Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 482
Filtrar
1.
Int J Mol Sci ; 25(8)2024 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-38674125

RESUMO

Polyomavirus (PyV) Large T-antigen (LT) is the major viral regulatory protein that targets numerous cellular pathways for cellular transformation and viral replication. LT directly recruits the cellular replication factors involved in initiation of viral DNA replication through mutual interactions between LT, DNA polymerase alpha-primase (Polprim), and single-stranded DNA binding complex, (RPA). Activities and interactions of these complexes are known to be modulated by post-translational modifications; however, high-sensitivity proteomic analyses of the PTMs and proteins associated have been lacking. High-resolution liquid chromatography tandem mass spectrometry (LC-MS/MS) of the immunoprecipitated factors (IPMS) identified 479 novel phosphorylated amino acid residues (PAARs) on the three factors; the function of one has been validated. IPMS revealed 374, 453, and 183 novel proteins associated with the three, respectively. A significant transcription-related process network identified by Gene Ontology (GO) enrichment analysis was unique to LT. Although unidentified by IPMS, the ETS protooncogene 1, transcription factor (ETS1) was significantly overconnected to our dataset indicating its involvement in PyV processes. This result was validated by demonstrating that ETS1 coimmunoprecipitates with LT. Identification of a novel PAAR that regulates PyV replication and LT's association with the protooncogenic Ets1 transcription factor demonstrates the value of these results for studies in PyV biology.


Assuntos
Replicação do DNA , Polyomavirus , Proteômica , Replicação Viral , Fosforilação , Humanos , Proteômica/métodos , Polyomavirus/metabolismo , Polyomavirus/genética , Espectrometria de Massas em Tandem , Proteína Proto-Oncogênica c-ets-1/metabolismo , Proteína Proto-Oncogênica c-ets-1/genética , Cromatografia Líquida , Antígenos Virais de Tumores/metabolismo , Antígenos Virais de Tumores/genética , Processamento de Proteína Pós-Traducional , DNA Viral/metabolismo , DNA Viral/genética
2.
Viruses ; 15(11)2023 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-38005912

RESUMO

Protein phosphorylation and dephosphorylation are the most common post-translational modifications mediated by protein kinases and protein phosphatases, respectively. These reversible processes can modulate the function of the target protein, such as its activity, subcellular localization, stability, and interaction with other proteins. Phosphorylation of viral proteins plays an important role in the life cycle of a virus. In this review, we highlight biological implications of the phosphorylation of the monkey polyomavirus SV40 large T and small t antigens, summarize our current knowledge of the phosphorylation of these proteins of human polyomaviruses, and conclude with gaps in the knowledge and a proposal for future research directions.


Assuntos
Infecções por Polyomavirus , Polyomavirus , Humanos , Polyomavirus/metabolismo , Antígenos Virais de Tumores/metabolismo , Fosforilação , Proteínas Quinases/metabolismo
3.
Cell Rep ; 42(2): 112114, 2023 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-36790933

RESUMO

BK polyomavirus (BKPyV) is an opportunistic pathogen that uses the b-series gangliosides GD1b and GT1b as entry receptors. Here, we characterize the impact of naturally occurring VP1 mutations on ganglioside binding, VP1 protein structure, and virus tropism. Infectious entry of single mutants E73Q and E73A and the triple mutant A72V-E73Q-E82Q (VQQ) remains sialic acid dependent, and all three variants acquire binding to a-series gangliosides, including GD1a. However, the E73A and VQQ variants lose the ability to infect ganglioside-complemented cells, and this correlates with a clear shift of the BC2 loop in the crystal structures of E73A and VQQ. On the other hand, the K69N mutation in the K69N-E82Q variant leads to a steric clash that precludes sialic acid binding. Nevertheless, this mutant retains significant infectivity in 293TT cells, which is not dependent on heparan sulfate proteoglycans, implying that an unknown sialic acid-independent entry receptor for BKPyV exists.


Assuntos
Vírus BK , Polyomavirus , Vírus BK/genética , Vírus BK/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Polyomavirus/genética , Polyomavirus/metabolismo , Capsídeo/metabolismo , Proteínas do Capsídeo/metabolismo , Gangliosídeos/metabolismo
4.
Biochim Biophys Acta Mol Basis Dis ; 1868(12): 166537, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36089125

RESUMO

Human polyomaviruses are relatively common in the general population. Polyomaviruses maintain a persistent infection after initial infection in childhood, acting as an opportunistic pathogen in immunocompromised populations and their association has been linked to carcinogenesis. A comprehensive understanding of the underlying molecular mechanisms of carcinogenesis in consequence of polyomavirus infection remains elusive. However, the critical role of viral miRNAs and their potential targets in modifying the transcriptome profile of the host remains largely unknown. Polyomavirus-derived miRNAs have the potential to play a substantial role in carcinogenesis. Employing computational approaches, putative viral miRNAs along with their target genes have been predicted and possible roles of the targeted genes in many significant biological processes have been obtained. Polyomaviruses have been observed to target intracellular signal transduction pathways through miRNA-mediated epigenetic regulation, which may contribute to cancer development. In addition, BKPyV-infected human renal cell microarray data was coupled with predicted target genes and analysis of the downregulated genes indicated that viruses target multiple signaling pathways (e.g. MAPK signaling pathway, PI3K-Akt signaling pathway, PPAR signaling pathway) in the host as well as turning off several tumor suppression genes (e.g. FGGY, EPHX2, CACNA2D3, CDH16) through miRNA-induced mechanisms, assuring cell transformation. This study provides a conceptual framework for the underlying molecular mechanisms involved in the course of carcinogenesis upon polyomavirus infection.


Assuntos
MicroRNAs , Infecções por Polyomavirus , Polyomavirus , Carcinogênese/genética , Epigênese Genética , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/genética , Fosfatidilinositol 3-Quinases/metabolismo , Polyomavirus/genética , Polyomavirus/metabolismo , Infecções por Polyomavirus/genética , Infecções por Polyomavirus/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo
5.
J Virol ; 96(14): e0206121, 2022 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-35770990

RESUMO

Several studies reported the presence of a recently discovered polyomavirus (PyV), Lyon IARC PyV (LIPyV), in human and domestic animal specimens. LIPyV has some structural similarities to well-established animal and human oncogenic PyVs, such as raccoon PyV and Merkel cell PyV (MCPyV), respectively. In this study, we demonstrate that LIPyV early proteins immortalize human foreskin keratinocytes. LIPyV LT binds pRb, accordingly cell cycle checkpoints are altered in primary human fibroblasts and keratinocytes expressing LIPyV early genes. Mutation of the pRb binding site in LT strongly affected the ability of LIPyV ER to induced HFK immortalization. LIPyV LT also binds p53 and alters p53 functions activated by cellular stresses. Finally, LIPyV early proteins activate telomerase reverse transcriptase (hTERT) gene expression, via accumulation of the Sp1 transcription factor. Sp1 recruitment to the hTERT promoter is controlled by its phosphorylation, which is mediated by ERK1 and CDK2. Together, these data highlight the transforming properties of LIPyV in in vitro experimental models, supporting its possible oncogenic nature. IMPORTANCE Lyon IARC PyV is a recently discovered polyomavirus that shows some structural similarities to well-established animal and human oncogenic PyVs, such as raccoon PyV and Merkel cell PyV, respectively. Here, we show the capability of LIPyV to efficiently promote cellular transformation of primary human cells, suggesting a possible oncogenic role of this virus in domestic animals and/or humans. Our study identified a novel virus-mediated mechanism of activation of telomerase reverse transcriptase gene expression, via accumulation of the Sp1 transcription factor. In addition, because the persistence of infection is a key event in virus-mediated carcinogenesis, it will be important to determine whether LIPyV can deregulate immune-related pathways, similarly to the well-established oncogenic viruses.


Assuntos
Infecções por Polyomavirus , Polyomavirus , Animais , Carcinogênese , Fibroblastos/virologia , Humanos , Queratinócitos/virologia , Poliomavírus das Células de Merkel/genética , Polyomavirus/genética , Polyomavirus/metabolismo , Infecções por Polyomavirus/virologia , Fator de Transcrição Sp1/metabolismo , Telomerase/genética , Proteína Supressora de Tumor p53/metabolismo
6.
Genes (Basel) ; 13(5)2022 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-35627238

RESUMO

Although genetic transformation has opened up a new era for cotton molecular breeding, it still suffers from the limitation problem of long transformation periods, which slows down the generation of new cotton germplasms. In this study, LT gene (SV40 large T antigen), which promotes the transformation efficiency of animal cells, was codon-optimized. Its overexpression vector was transformed into cotton. It was observed that EC (embryogenic callus) formation period was 33% shorter and transformation efficiency was slightly higher in the LT T0 generation than that of control. RNA-seq data of NEC (non-embryonic callus) and EC from LT and control revealed that more DEGs (differential expression genes) in NEC were identified than that of EC, indicating LT mainly functioned in NEC. Further KEGG, GO, and transcription factor analyses showed that DEGs were significantly enriched in brassinosteroid biosynthesis pathways and that bHLH, MYB, and AP2/ERF were the top three gene families, which are involved in EC formation. In addition, the key genes related to the auxin pathway were differentially expressed only in LT overexpression NEC, which caused early response, biosynthesis, and transportation of the hormone, resulting in EC earlier formation. In summary, the results demonstrated that LT can promote somatic embryogenesis in cotton, which provides a new strategy for improving cotton transformation and shortening EC formation time.


Assuntos
Regulação da Expressão Gênica de Plantas , Polyomavirus , Fibra de Algodão , Desenvolvimento Embrionário , Regulação da Expressão Gênica de Plantas/genética , Ácidos Indolacéticos/metabolismo , Proteínas Oncogênicas/genética , Polyomavirus/metabolismo
7.
Viruses ; 14(3)2022 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-35336880

RESUMO

Merkel cell polyomavirus (MCV) causes one of the most aggressive human skin cancers, but laboratory studies on MCV replication have proven technically difficult. We report the first recombinase-mediated MCV minicircle (MCVmc) system that generates high levels of circularized virus, allowing facile MCV genetic manipulation and characterization of viral gene expression kinetics during replication. Mutations to Fbw7, Skp2, ß-TrCP and hVam6p interaction sites, or to the stem loop sequence for the MCV-encoded miRNA precursor, markedly increase viral replication, whereas point mutation to an origin-binding site eliminates active virus replication. To further increase the utility of this system, an mScarlet fusion protein was inserted into the VP1 c-terminus to generate a non-infectious reporter virus for studies on virus kinetics. When this reporter virus genome is heterologously expressed together with MCV VP1 and VP2, virus-like particles are generated. The reporter virus genome is encapsidated and can be used at lower biosafety levels for one-round infection studies. Our findings reveal that MCV has multiple, self-encoded viral restriction mechanisms to promote viral latency over lytic replication, and these mechanisms are now amenable to examination using a recombinase technology.


Assuntos
Poliomavírus das Células de Merkel , Infecções por Polyomavirus , Polyomavirus , Infecções Tumorais por Vírus , Antígenos Virais de Tumores/genética , Humanos , Cinética , Poliomavírus das Células de Merkel/genética , Poliomavírus das Células de Merkel/metabolismo , Polyomavirus/genética , Polyomavirus/metabolismo , Recombinases/metabolismo , Replicação Viral/genética
8.
ACS Synth Biol ; 10(12): 3251-3263, 2021 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-34591448

RESUMO

Metabolic pathways are commonly organized by sequestration into discrete cellular compartments. Compartments prevent unfavorable interactions with other pathways and provide local environments conducive to the activity of encapsulated enzymes. Such compartments are also useful synthetic biology tools for examining enzyme/pathway behavior and for metabolic engineering. Here, we expand the intracellular compartmentalization toolbox for budding yeast (Saccharomyces cerevisiae) with Murine polyomavirus virus-like particles (MPyV VLPs). The MPyV system has two components: VP1 which self-assembles into the compartment shell and a short anchor, VP2C, which mediates cargo protein encapsulation via binding to the inner surface of the VP1 shell. Destabilized green fluorescent protein (GFP) fused to VP2C was specifically sorted into VLPs and thereby protected from host-mediated degradation. An engineered VP1 variant displayed improved cargo capture properties and differential subcellular localization compared to wild-type VP1. To demonstrate their ability to function as a metabolic compartment, MPyV VLPs were used to encapsulate myo-inositol oxygenase (MIOX), an unstable and rate-limiting enzyme in d-glucaric acid biosynthesis. Strains with encapsulated MIOX produced ∼20% more d-glucaric acid compared to controls expressing "free" MIOX─despite accumulating dramatically less expressed protein─and also grew to higher cell densities. This is the first demonstration in yeast of an artificial biocatalytic compartment that can participate in a metabolic pathway and establishes the MPyV platform as a promising synthetic biology tool for yeast engineering.


Assuntos
Polyomavirus , Saccharomyces cerevisiae , Animais , Proteínas do Capsídeo/metabolismo , Ácido Glucárico/metabolismo , Inositol Oxigenase/metabolismo , Redes e Vias Metabólicas , Camundongos , Polyomavirus/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
9.
Viruses ; 12(10)2020 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-32993049

RESUMO

Extracellular vesicles have recently emerged as a novel mode of viral transmission exploited by naked viruses to exit host cells through a nonlytic pathway. Extracellular vesicles can allow multiple viral particles to collectively traffic in and out of cells, thus enhancing the viral fitness and diversifying the transmission routes while evading the immune system. This has been shown for several RNA viruses that belong to the Picornaviridae, Hepeviridae, Reoviridae, and Caliciviridae families; however, recent studies also demonstrated that the BK and JC viruses, two DNA viruses that belong to the Polyomaviridae family, use a similar strategy. In this review, we provide an update on recent advances in understanding the mechanisms used by naked viruses to hijack extracellular vesicles, and we discuss the implications for the biology of polyomaviruses.


Assuntos
Vesículas Extracelulares/virologia , Evasão da Resposta Imune/fisiologia , Infecções por Polyomavirus/transmissão , Polyomavirus/metabolismo , Infecções Tumorais por Vírus/transmissão , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Endocitose/fisiologia , Humanos , Polyomavirus/imunologia , Infecções Tumorais por Vírus/virologia , Internalização do Vírus , Replicação Viral/fisiologia
10.
Viruses ; 12(10)2020 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-32987952

RESUMO

Polyomavirus infection is widespread in the human population. This family of viruses normally maintains latent infection within the host cell but can cause a range of human pathologies, especially in immunocompromised individuals. Among several known pathogenic human polyomaviruses, JC polyomavirus (JCPyV) has the potential to cause the demyelinating disease progressive multifocal leukoencephalopathy (PML); BK polyomavirus (BKPyV) can cause nephropathy in kidney transplant recipients, and Merkel cell polyomavirus (MCPyV) is associated with a highly aggressive form of skin cancer, Merkel cell carcinoma (MCC). While the mechanisms by which these viruses give rise to the relevant diseases are not well understood, it is clear that the control of gene expression in each polyomavirus plays an important role in determining the infectious tropism of the virus as well as their potential to promote disease progression. In this review, we discuss the mechanisms governing the transcriptional regulation of these pathogenic human polyomaviruses in addition to the best-studied simian vacuolating virus 40 (SV40). We highlight the roles of viral cis-acting DNA elements, encoded proteins and miRNAs that control the viral gene expression. We will also underline the cellular transcription factors and epigenetic modifications that regulate the gene expression of these viruses.


Assuntos
Regulação Viral da Expressão Gênica/genética , Infecções por Polyomavirus/patologia , Polyomavirus/genética , Polyomavirus/metabolismo , Infecções Tumorais por Vírus/patologia , Vírus BK/genética , Vírus BK/metabolismo , Carcinoma de Célula de Merkel/patologia , Carcinoma de Célula de Merkel/virologia , Humanos , Vírus JC/genética , Vírus JC/metabolismo , Infecção Latente/virologia , Leucoencefalopatia Multifocal Progressiva/patologia , Leucoencefalopatia Multifocal Progressiva/virologia , Poliomavírus das Células de Merkel/genética , Poliomavírus das Células de Merkel/metabolismo , Infecções por Polyomavirus/virologia , Vírus 40 dos Símios/genética , Vírus 40 dos Símios/metabolismo , Infecções Tumorais por Vírus/virologia
11.
J Virol ; 94(14)2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32404521

RESUMO

UNC5B is a dependence receptor that promotes survival in the presence of its ligand, netrin-1, while inducing cell death in its absence. The receptor has an important role in the development of the nervous and vascular systems. It is also involved in the normal turnover of intestinal epithelium. Netrin-1 and UNC5B are deregulated in multiple cancers, including colorectal, neuroblastoma, and breast tumors. However, the detailed mechanism of UNC5B function is not fully understood. We have utilized the murine polyomavirus small T antigen (PyST) as a tool to study UNC5B-mediated apoptosis. PyST is known to induce mitotic arrest followed by extensive cell death in mammalian cells. Our results show that the expression of PyST increases mRNA levels of UNC5B by approximately 3-fold in osteosarcoma cells (U2OS) and also stabilizes UNC5B at the posttranslational level. Furthermore, UNC5B is upregulated predominantly in those cells that undergo mitotic arrest upon PyST expression. Interestingly, although its expression was previously reported to be regulated by p53, our data show that the increase in UNC5B levels by PyST is p53 independent. The posttranslational stabilization of UNC5B by PyST is regulated by the interaction of PyST with PP2A. We also show that netrin-1 expression, which is known to inhibit UNC5B apoptotic activity, promotes survival of PyST-expressing cells. Our results thus suggest an important role of UNC5B in small-T antigen-induced mitotic catastrophe that also requires PP2A.IMPORTANCE UNC5B, PP2A, and netrin-1 are deregulated in a variety of cancers. UNC5B and PP2A are regarded as tumor suppressors, as they promote apoptosis and are deleted or mutated in many cancers. In contrast, netrin-1 promotes survival by inhibiting dependence receptors, including UNC5B, and is upregulated in many cancers. Here, we show that UNC5B-mediated apoptosis can occur independently of p53 but in a PP2A-dependent manner. A substantial percentage of cancers arise due to p53 mutations and are insensitive to chemotherapeutic treatments that activate p53. Unexpectedly, treatment of cancers having functional p53 with many conventional drugs leads to the upregulation of netrin-1 through activated p53, which is counterintuitive. Therefore, understanding the p53-independent mechanisms of the netrin-UNC5B axis, such as those involving PP2A, assumes greater clinical significance. Anticancer strategies utilizing anti-netrin-1 antibody treatment are already in clinical trials.


Assuntos
Antígenos Virais de Tumores/metabolismo , Apoptose , Receptores de Netrina/metabolismo , Polyomavirus/metabolismo , Proteína Fosfatase 2/metabolismo , Células A549 , Animais , Antígenos Virais de Tumores/genética , Células HeLa , Humanos , Camundongos , Receptores de Netrina/genética , Polyomavirus/genética , Proteína Fosfatase 2/genética
12.
Viruses ; 12(2)2020 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-32085463

RESUMO

Viruses have evolved mechanisms to manipulate microtubules (MTs) for the efficient realization of their replication programs. Studying the mechanisms of replication of mouse polyomavirus (MPyV), we observed previously that in the late phase of infection, a considerable amount of the main structural protein, VP1, remains in the cytoplasm associated with hyperacetylated microtubules. VP1-microtubule interactions resulted in blocking the cell cycle in the G2/M phase. We are interested in the mechanism leading to microtubule hyperacetylation and stabilization and the roles of tubulin acetyltransferase 1 (αTAT1) and deacetylase histone deacetylase 6 (HDAC6) and VP1 in this mechanism. Therefore, HDAC6 inhibition assays, αTAT1 knock out cell infections, in situ cell fractionation, and confocal and TIRF microscopy were used. The experiments revealed that the direct interaction of isolated microtubules and VP1 results in MT stabilization and a restriction of their dynamics. VP1 leads to an increase in polymerized tubulin in cells, thus favoring αTAT1 activity. The acetylation status of MTs did not affect MPyV infection. However, the stabilization of MTs by VP1 in the late phase of infection may compensate for the previously described cytoskeleton destabilization by MPyV early gene products and is important for the observed inhibition of the G2→M transition of infected cells to prolong the S phase.


Assuntos
Acetiltransferases/genética , Proteínas do Capsídeo/genética , Interações entre Hospedeiro e Microrganismos , Microtúbulos/metabolismo , Polyomavirus/metabolismo , Acetilação , Acetiltransferases/metabolismo , Animais , Proteínas do Capsídeo/metabolismo , Ciclo Celular , Linhagem Celular , Citoplasma/metabolismo , Fibroblastos/virologia , Desacetilase 6 de Histona/genética , Desacetilase 6 de Histona/metabolismo , Camundongos , Microtúbulos/virologia , Polyomavirus/genética , Processamento de Proteína Pós-Traducional , Tubulina (Proteína)/metabolismo
13.
Int J Pharm ; 576: 119008, 2020 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-31901358

RESUMO

Viral nanoparticles represent potential natural versatile platforms for targeted gene and drug delivery. Improving the efficiency of gene transfer mediated by viral vectors could not only enhance their therapeutic potential, but also contribute to understanding the limitations in interactions of nanoparticles with cells and the development of new therapeutic approaches. In this study, four cell-penetrating peptides (CPPs), cationic octaarginine (R8), histidine-rich peptides (LAH4 and KH27K) and fusogenic peptide (FUSO), are investigated for their effect on infection by mouse polyomavirus (MPyV) or on transduction of reporter genes delivered by MPyV or related viral vectors. Peptides noncovalently associated with viral particles enhance gene transfer (with the exception of FUSO). Removal of cellular heparan sulfates by the heparinase does not significantly change the enhancing potential of CPPs. Instead, CPPs influences the physical state of viral particles: R8 slightly destabilizes the intact virus, KH27K induces its aggregation and LAH4 promotes disassembly and aggregation of the particles that massively and rapidly associate with cells. The findings indicate that peptides acting as transduction-enhancing agents of polyomavirus-based nanoparticles modulate their physical state, which can be an important prerequisite for sensitization of cells and determination of the further fate of viral particles inside cells.


Assuntos
Peptídeos Penetradores de Células/metabolismo , Vetores Genéticos , Polyomavirus/metabolismo , Transdução Genética , Vírion/metabolismo , Animais , Capsídeo/metabolismo , Capsídeo/ultraestrutura , Peptídeos Penetradores de Células/química , Células HEK293 , Humanos , Camundongos , Oligopeptídeos/química , Oligopeptídeos/metabolismo , Polyomavirus/genética , Polyomavirus/ultraestrutura , Vírion/genética , Vírion/ultraestrutura
15.
Acta Crystallogr F Struct Biol Commun ; 74(Pt 8): 451-462, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-30084394

RESUMO

Biomolecular NMR spectroscopy has limitations in the determination of protein structures: an inherent size limit and the requirement for expensive and potentially difficult isotope labelling pose considerable hurdles. Therefore, structural analysis of larger proteins is almost exclusively performed by crystallography. However, the diversity of biological NMR applications outperforms that of any other structural biology technique. For the characterization of transient complexes formed by proteins and small ligands, notably oligosaccharides, one NMR technique has recently proven to be particularly powerful: saturation-transfer difference NMR (STD-NMR) spectroscopy. STD-NMR experiments are fast and simple to set up, with no general protein size limit and no requirement for isotope labelling. The method performs best in the moderate-to-low affinity range that is of interest in most of glycobiology. With small amounts of unlabelled protein, STD-NMR experiments can identify hits from mixtures of potential ligands, characterize mutant proteins and pinpoint binding epitopes on the ligand side. STD-NMR can thus be employed to complement and improve protein-ligand complex models obtained by other structural biology techniques or by purely computational means. With a set of protein-glycan interactions from our own work, this review provides an introduction to the technique for structural biologists. It exemplifies how crystallography and STD-NMR can be combined to elucidate protein-glycan (and other protein-ligand) interactions in atomic detail, and how the technique can extend structural biology from simplified systems amenable to crystallization to more complex biological entities such as membranes, live viruses or entire cells.


Assuntos
Lectinas/química , Ressonância Magnética Nuclear Biomolecular/métodos , Polyomavirus/química , Polissacarídeos/química , Animais , Cristalografia por Raios X/métodos , Humanos , Lectinas/metabolismo , Polyomavirus/metabolismo , Polissacarídeos/metabolismo , Ligação Proteica/fisiologia , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
16.
ACS Nano ; 12(5): 4615-4623, 2018 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-29697964

RESUMO

Bioinspired self-sorting and self-assembling systems using engineered versions of natural protein cages are being developed for biocatalysis and therapeutic delivery. The packaging and intracellular delivery of guest proteins is of particular interest for both in vitro and in vivo cell engineering. However, there is a lack of bionanotechnology platforms that combine programmable guest protein encapsidation with efficient intracellular uptake. We report a minimal peptide anchor for in vivo self-sorting of cargo-linked capsomeres of murine polyomavirus (MPyV) that enables controlled encapsidation of guest proteins by in vitro self-assembly. Using Förster resonance energy transfer, we demonstrate the flexibility in this system to support coencapsidation of multiple proteins. Complementing these ensemble measurements with single-particle analysis by super-resolution microscopy shows that the stochastic nature of coencapsidation is an overriding principle. This has implications for the design and deployment of both native and engineered self-sorting encapsulation systems and for the assembly of infectious virions. Taking advantage of the encoded affinity for sialic acids ubiquitously displayed on the surface of mammalian cells, we demonstrate the ability of self-assembled MPyV virus-like particles to mediate efficient delivery of guest proteins to the cytosol of primary human cells. This platform for programmable coencapsidation and efficient cytosolic delivery of complementary biomolecules therefore has enormous potential in cell engineering.


Assuntos
Proteínas do Capsídeo/química , Capsídeo/química , Proteínas de Fluorescência Verde/química , Nanopartículas/química , Polyomavirus/metabolismo , Proteínas Recombinantes de Fusão/química , Animais , Capsídeo/metabolismo , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Engenharia Celular/métodos , Citosol/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Transferência Ressonante de Energia de Fluorescência , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Polyomavirus/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Montagem de Vírus
17.
Virology ; 506: 45-54, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28342387

RESUMO

The small double-stranded DNA polyomaviruses (PyVs) form a family of 73 species, whose natural hosts are primarily mammals and birds. So far, 13 PyVs have been isolated in humans, and some of them have clearly been associated with several diseases, including cancer. In this study, we describe the isolation of a novel PyV in human skin using a sensitive degenerate PCR protocol combined with next-generation sequencing. The new virus, named Lyon IARC PyV (LIPyV), has a circular genome of 5269 nucleotides. Phylogenetic analyses showed that LIPyV is related to the raccoon PyV identified in neuroglial tumours in free-ranging raccoons. Analysis of human specimens from cancer-free individuals showed that 9 skin swabs (9/445; 2.0%), 3 oral gargles (3/140; 2.1%), and one eyebrow hair sample (1/439; 0.2%) tested positive for LIPyV. Future biological and epidemiological studies are needed to confirm the human tropism and provide insights into its biological properties.


Assuntos
Infecções por Polyomavirus/virologia , Polyomavirus/isolamento & purificação , Sequência de Aminoácidos , Animais , Genoma Viral , Glioma/virologia , Humanos , Dados de Sequência Molecular , Filogenia , Polyomavirus/classificação , Polyomavirus/genética , Polyomavirus/metabolismo , Guaxinins/virologia , Proteínas Virais/genética , Proteínas Virais/metabolismo
18.
FEBS J ; 284(6): 883-902, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28164464

RESUMO

The minor structural protein VP2 and its shorter variant, VP3, of mouse polyomavirus (MPyV) are essential for virus exit from the endoplasmic reticulum (ER) during viral trafficking to the nucleus. Here, we followed the role of putative hydrophobic domains (HD) of the minor proteins in membrane affinity and viral infectivity. We prepared variants of VP2, each mutated to decrease hydrophobicity of one of three predicted hydrophobic domains: VP2-mHD1, VP2-mHD2 or VP2-mHD3 mutated in HD1 (amino acids (aa) 60-101), HD2 (aa 125-165) or HD3 (aa 287-307), respectively. Transient production of the mutated proteins revealed that only VP2-mHD2 lost the affinity for intracellular membranes. Cytotoxicity connected with the ability of VP2/VP3 to perforate membranes decreased markedly for VP2-mHD2, but only slightly for VP2-mHD1. The mutant VP2-mHD3 exhibited properties similar to the wild-type protein. MPyV genomes, each carrying one of the mutations, were prepared for virus production. MPyV-mHD1 and MPyV-mHD2 viruses could be isolated, while the HD3 mutation in VP2/VP3 prevented virus assembly. We found that both MPyV-mHD1 and MPyV-mHD2 viruses arrived at the ER without delay and were processed by ER residential enzymes. However, the ability to associate with ER membranes was decreased in the case of MPyV-mHD1 and practically abolished in the case of MPyV-mHD2. Interestingly, while MPyV-mHD2 was not infectious, infection of MPyV-mHD1 virus was delayed. These findings reveal that HD2, common to both VP2 and VP3, is responsible for the membrane binding properties of the minor proteins, while HD1 of VP2 is likely required to stabilize VP2-membrane association and to enhance viral exit from the ER.


Assuntos
Proteínas do Capsídeo/metabolismo , Retículo Endoplasmático/metabolismo , Membranas Intracelulares/metabolismo , Polyomavirus/metabolismo , Sequência de Aminoácidos , Animais , Proteínas do Capsídeo/genética , Núcleo Celular/metabolismo , Retículo Endoplasmático/genética , Humanos , Interações Hidrofóbicas e Hidrofílicas , Camundongos , Polyomavirus/genética , Polyomavirus/patogenicidade , Ligação Proteica , Domínios Proteicos
19.
FEBS J ; 284(2): 301-323, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27885808

RESUMO

VP1, the major structural protein of the mouse polyomavirus (MPyV), is the major architectural component of the viral capsid. Its pentamers are able to self-assemble into capsid-like particles and to non-specifically bind DNA. Surface loops of the protein interact with sialic acid of ganglioside receptors. Although the replication cycle of the virus, including virion morphogenesis, proceeds in the cell nucleus, a substantial fraction of the protein is detected in the cytoplasm of late-phase MPyV-infected cells. In this work, we detected VP1 mainly in the cytoplasm of mammalian cells transfected with plasmid expressing VP1. In the cytoplasm, VP1-bound microtubules, including the mitotic spindle, and the interaction of VP1 with microtubules resulted in cell cycle block at the G2/M phase. Furthermore, in the late phase of MPyV infection and in cells expressing VP1, microtubules were found to be hyperacetylated. We then sought to understand how VP1 interacts with microtubules. Dynein is not responsible for the VP1-microtubule association, as neither overexpression of p53/dynamitin nor treatment with ciliobrevin-D (an inhibitor of dynein activity) prevented binding of VP1 to microtubules. A pull-down assay for VP1-interacting proteins identified the heat shock protein 90 (Hsp90) chaperone, and Hsp90 was also detected in the VP1-microtubule complexes. Although Hsp90 is known to be associated with acetylated microtubules, it does not mediate the interaction between VP1 and microtubules. Our study provides insight into the role of the major structural protein in MPyV replication, indicating that VP1 is a multifunctional protein that participates in the regulation of cell cycle progression in MPyV-infected cells.


Assuntos
Proteínas do Capsídeo/metabolismo , Células Epiteliais/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Microtúbulos/metabolismo , Polyomavirus/metabolismo , Vírion/metabolismo , Acetilação , Animais , Proteínas do Capsídeo/genética , Núcleo Celular/metabolismo , Núcleo Celular/virologia , Citoplasma/metabolismo , Citoplasma/virologia , Células Epiteliais/virologia , Feminino , Pontos de Checagem da Fase G2 do Ciclo Celular , Expressão Gênica , Células HEK293 , Proteínas de Choque Térmico HSP90/genética , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/virologia , Camundongos , Microtúbulos/virologia , Células NIH 3T3 , Plasmídeos/química , Plasmídeos/metabolismo , Polyomavirus/genética , Ligação Proteica , Transfecção , Vírion/genética
20.
J Gen Virol ; 97(11): 2939-2948, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27600312

RESUMO

Polyomavirus infection often results in persistence of the viral genome with little or no virion production. However, infection of certain cell types can result in high viral gene transcription and either cytolysis or neoplastic transformation. While infection by polyomavirus is common in humans and many animals, major questions regarding viral persistence of most polyomaviruses remain unanswered. Specifically, identification of target cells for viral infection and the mechanisms polyomaviruses employ to maintain viral genomes within cells are important not only in ascribing causality to polyomaviruses in disease, but in understanding specific mechanisms by which they cause disease. Here, we characterize the cell of origin in raccoon polyomavirus (RacPyV)-associated neuroglial brain tumours as a neural stem cell. Moreover, we identify an association between the viral genome and the host cell bromodomain protein, BRD4, which is involved in numerous cellular functions, including cell cycle progression, differentiation of stem cells, tethering of persistent DNA viruses, and regulation of viral and host-cell gene transcription. We demonstrate that inhibition of BRD4 by the small molecule inhibitors (+)-JQ1 and IBET-151 (GSK1210151A) results in reduced RacPyV genome within cells in vitro, as well as significant reduction of viral gene transcripts LT and VP1, highlighting its importance in both maintenance of the viral genome and in driving oncogenic transformation by RacPyV. This work implicates BRD4 as a central protein involved in RacPyV neuroglial tumour cell proliferation and in the maintenance of a stem cell state.


Assuntos
Neuroglia/virologia , Infecções por Polyomavirus/veterinária , Polyomavirus/genética , Guaxinins/virologia , Células-Tronco/virologia , Fatores de Transcrição/metabolismo , Infecções Tumorais por Vírus/veterinária , Proteínas Virais/genética , Animais , Proliferação de Células , Transformação Celular Neoplásica , Genoma Viral , Neuroglia/metabolismo , Polyomavirus/metabolismo , Infecções por Polyomavirus/metabolismo , Infecções por Polyomavirus/fisiopatologia , Infecções por Polyomavirus/virologia , Células-Tronco/metabolismo , Fatores de Transcrição/genética , Transcrição Gênica , Infecções Tumorais por Vírus/metabolismo , Infecções Tumorais por Vírus/fisiopatologia , Infecções Tumorais por Vírus/virologia , Proteínas Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA