Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Virol J ; 20(1): 82, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-37127643

RESUMO

Promyelocytic leukemia nuclear bodies (PM NBs), often referred to as membraneless organelles, are dynamic macromolecular protein complexes composed of a PML protein core and other transient or permanent components. PML NBs have been shown to play a role in a wide variety of cellular processes. This review describes in detail the diverse and complex interactions between small and medium size DNA viruses and PML NBs that have been described to date. The PML NB components that interact with small and medium size DNA viruses include PML protein isoforms, ATRX/Daxx, Sp100, Sp110, HP1, and p53, among others. Interaction between viruses and components of these NBs can result in different outcomes, such as influencing viral genome expression and/or replication or impacting IFN-mediated or apoptotic cell responses to viral infection. We discuss how PML NB components abrogate the ability of adenoviruses or Hepatitis B virus to transcribe and/or replicate their genomes and how papillomaviruses use PML NBs and their components to promote their propagation. Interactions between polyomaviruses and PML NBs that are poorly understood but nevertheless suggest that the NBs can serve as scaffolds for viral replication or assembly are also presented. Furthermore, complex interactions between the HBx protein of hepadnaviruses and several PML NBs-associated proteins are also described. Finally, current but scarce information regarding the interactions of VP3/apoptin of the avian anellovirus with PML NBs is provided. Despite the considerable number of studies that have investigated the functions of the PML NBs in the context of viral infection, gaps in our understanding of the fine interactions between viruses and the very dynamic PML NBs remain. The complexity of the bodies is undoubtedly a great challenge that needs to be further addressed.


Assuntos
Vírus de DNA , Proteínas Nucleares , Adenoviridae , Proteínas Nucleares/metabolismo , Corpos Nucleares da Leucemia Promielocítica , Proteína da Leucemia Promielocítica/metabolismo , Fatores de Transcrição/metabolismo , Vírus , Vírus de DNA/genética
2.
Viruses ; 14(4)2022 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-35458396

RESUMO

DNA virus infections are often lifelong and can cause serious diseases in their hosts. Their recognition by the sensors of the innate immune system represents the front line of host defence. Understanding the molecular mechanisms of innate immunity responses is an important prerequisite for the design of effective antivirotics. This review focuses on the present state of knowledge surrounding the mechanisms of viral DNA genome sensing and the main induced pathways of innate immunity responses. The studies that have been performed to date indicate that herpesviruses, adenoviruses, and polyomaviruses are sensed by various DNA sensors. In non-immune cells, STING pathways have been shown to be activated by cGAS, IFI16, DDX41, or DNA-PK. The activation of TLR9 has mainly been described in pDCs and in other immune cells. Importantly, studies on herpesviruses have unveiled novel participants (BRCA1, H2B, or DNA-PK) in the IFI16 sensing pathway. Polyomavirus studies have revealed that, in addition to viral DNA, micronuclei are released into the cytosol due to genotoxic stress. Papillomaviruses, HBV, and HIV have been shown to evade DNA sensing by sophisticated intracellular trafficking, unique cell tropism, and viral or cellular protein actions that prevent or block DNA sensing. Further research is required to fully understand the interplay between viruses and DNA sensors.


Assuntos
Infecções por Vírus de DNA , Herpesviridae , Polyomavirus , DNA Viral/metabolismo , Herpesviridae/genética , Herpesviridae/metabolismo , Humanos , Imunidade Inata , Polyomavirus/genética
3.
Int J Mol Sci ; 23(1)2022 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-35009004

RESUMO

The nuclear lamina is the main component of the nuclear cytoskeleton that maintains the integrity of the nucleus. However, it represents a natural barrier for viruses replicating in the cell nucleus. The lamina blocks viruses from being trafficked to the nucleus for replication, but it also impedes the nuclear egress of the progeny of viral particles. Thus, viruses have evolved mechanisms to overcome this obstacle. Large viruses induce the assembly of multiprotein complexes that are anchored to the inner nuclear membrane. Important components of these complexes are the viral and cellular kinases phosphorylating the lamina and promoting its disaggregation, therefore allowing virus egress. Small viruses also use cellular kinases to induce lamina phosphorylation and the subsequent disruption in order to facilitate the import of viral particles during the early stages of infection or during their nuclear egress. Another component of the nuclear cytoskeleton, nuclear actin, is exploited by viruses for the intranuclear movement of their particles from the replication sites to the nuclear periphery. This study focuses on exploitation of the nuclear cytoskeleton by viruses, although this is just the beginning for many viruses, and promises to reveal the mechanisms and dynamic of physiological and pathological processes in the nucleus.


Assuntos
Núcleo Celular/metabolismo , Citoesqueleto/metabolismo , Suscetibilidade a Doenças , Interações Hospedeiro-Patógeno , Viroses/etiologia , Viroses/metabolismo , Actinas/metabolismo , Animais , Citoesqueleto/genética , Regulação Viral da Expressão Gênica , Humanos , Laminas/metabolismo , Membrana Nuclear/metabolismo , Lâmina Nuclear/metabolismo , Especificidade da Espécie , Replicação Viral
4.
Int J Pharm ; 611: 121308, 2022 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-34800617

RESUMO

The utilization of nanoparticles for the intracellular delivery of theranostic agents faces one substantial limitation. Sequestration in intracellular vesicles prevents them from reaching the desired location in the cytoplasm or nucleus to deliver their cargo. We investigated whether three different cell-penetrating peptides (CPPs), namely, octa-arginine R8, polyhistidine KH27K and histidine-rich LAH4, could promote cytosolic and/or nuclear transfer of unique model nanoparticles-pseudovirions derived from murine polyomavirus. Two types of CPP-modified pseudovirions that carry the luciferase reporter gene were created: VirPorters-IN with CPPs genetically attached to the capsid interior and VirPorters-EX with CPPs noncovalently associated with the capsid exterior. We tested their transduction ability by luciferase assay and monitored their presence in subcellular fractions. Our results confirmed the overall effect of CPPs on the intracellular destination of the particles and suggested that KH27K has the potential to improve the cytosolic release of pseudovirions. None of the VirPorters caused endomembrane damage detectable by the Galectin-3 assay. Remarkably, a noncovalent modification was required to promote high transduction of the reporter gene and cytosolic delivery of pseudovirions mediated by LAH4. Together, CPPs in different arrangements have demonstrated their potential to improve pseudovirion invasion into cells, and these findings could be useful for the development of other nanoparticle-based delivery systems.


Assuntos
Peptídeos Penetradores de Células , Animais , Bioensaio , Cátions , Citosol , Histidina , Camundongos
5.
FEBS J ; 288(20): 5964-5985, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33969628

RESUMO

The mechanism by which DNA viruses interact with different DNA sensors and their connection with the activation of interferon (IFN) type I pathway are poorly understood. We investigated the roles of protein 204 (p204) and cyclic guanosine-adenosine synthetase (cGAS) sensors during infection with mouse polyomavirus (MPyV). The phosphorylation of IFN regulatory factor 3 (IRF3) and the stimulator of IFN genes (STING) proteins and the upregulation of IFN beta (IFN-ß) and MX Dynamin Like GTPase 1 (MX-1) genes were detected at the time of replication of MPyV genomes in the nucleus. STING knockout abolished the IFN response. Infection with a mutant virus that exhibits defective nuclear entry via nucleopores and that accumulates in the cytoplasm confirmed that replication of viral genomes in the nucleus is required for IFN induction. The importance of both DNA sensors, p204 and cGAS, in MPyV-induced IFN response was demonstrated by downregulation of the IFN pathway observed in p204-knockdown and cGAS-knockout cells. Confocal microscopy revealed the colocalization of p204 with MPyV genomes in the nucleus. cGAS was found in the cytoplasm, colocalizing with viral DNA leaked from the nucleus and with DNA within micronucleus-like bodies, but also with the MPyV genomes in the nucleus. However, 2'3'-Cyclic guanosine monophosphate-adenosine monophosphate synthesized by cGAS was detected exclusively in the cytoplasm. Biochemical assays revealed no evidence of functional interaction between cGAS and p204 in the nucleus. Our results provide evidence for the complex interactions of MPyV and DNA sensors including the sensing of viral genomes in the nucleus by p204 and of leaked viral DNA and micronucleus-like bodies in the cytoplasm by cGAS.


Assuntos
DNA Viral/imunologia , Imunidade Inata/imunologia , Proteínas de Membrana/metabolismo , Proteínas Nucleares/metabolismo , Nucleotidiltransferases/metabolismo , Fosfoproteínas/metabolismo , Infecções por Polyomavirus/imunologia , Polyomavirus/imunologia , Infecções Tumorais por Vírus/imunologia , Animais , DNA Viral/genética , Interações Hospedeiro-Patógeno , Interferon beta/metabolismo , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Camundongos , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/genética , Nucleotidiltransferases/antagonistas & inibidores , Nucleotidiltransferases/genética , Fosfoproteínas/antagonistas & inibidores , Fosfoproteínas/genética , Fosforilação , Polyomavirus/genética , Infecções por Polyomavirus/virologia , Infecções Tumorais por Vírus/virologia
6.
Cancers (Basel) ; 13(9)2021 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-33923020

RESUMO

The tumorigenic potential of mouse polyomavirus (MPyV) has been studied for decades in cell culture models and has been mainly attributed to nonstructural middle T antigen (MT), which acts as a scaffold signal adaptor, activates Src tyrosine kinases, and possesses transforming ability. We hypothesized that MPyV could also transform mouse cells independent of MT via a Toll-like receptor 4 (TLR4)-mediated inflammatory mechanism. To this end, we investigated the interaction of MPyV with TLR4 in mouse embryonic fibroblasts (MEFs) and 3T6 cells, resulting in secretion of interleukin 6 (IL-6), independent of active viral replication. TLR4 colocalized with MPyV capsid protein VP1 in MEFs. Neither TLR4 activation nor recombinant IL-6 inhibited MPyV replication in MEFs and 3T6 cells. MPyV induced STAT3 phosphorylation through both direct and MT-dependent and indirect and TLR4/IL-6-dependent mechanisms. We demonstrate that uninfected mouse fibroblasts exposed to the cytokine environment from MPyV-infected fibroblasts upregulated the expressions of MCP-1, CCL-5, and α-SMA. Moreover, the cytokine microenvironment increased the invasiveness of MEFs and CT26 carcinoma cells. Collectively, TLR4 recognition of MPyV induces a cytokine environment that promotes the cancer-associated fibroblast (CAF)-like phenotype in noninfected fibroblasts and increases cell invasiveness.

7.
Bioconjug Chem ; 31(5): 1575-1585, 2020 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-32329599

RESUMO

Protein corona formation has been regarded as an obstacle to developing diagnostic and therapeutic nanoparticles for in vivo applications. Serum proteins that assemble around nanoparticles can hinder their targeting efficiency. Virus-based nanoparticles should be naturally predisposed to evade such barriers in host organisms. Here, we demonstrate that virus-like particles derived from mouse polyomavirus do not form a rich protein corona. These particles can be efficiently targeted to cells that overproduce transferrin receptors, e.g., cancer cells, by conjugating transferrin to the particle surface. In this study, we provide evidence that the interaction of virus-like particles with their newly assigned target receptor is not obstructed by serum proteins. The particles enter target cells via a clathrin-dependent endocytic pathway that is not naturally used by the virus. Our results support the notion that the natural properties of virus-like particles make them well-suited for development of nanosized theranostic tools resistant to detargeting by protein coronas.


Assuntos
Nanopartículas/química , Polyomavirus/química , Coroa de Proteína/química , Coroa de Proteína/metabolismo , Receptores da Transferrina/metabolismo , Animais , Proteínas Sanguíneas/química , Proteínas Sanguíneas/metabolismo , Humanos , Camundongos
8.
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
9.
Viruses ; 12(1)2020 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-31963741

RESUMO

Microtubules, part of the cytoskeleton, are indispensable for intracellular movement, cell division, and maintaining cell shape and polarity. In addition, microtubules play an important role in viral infection. In this review, we summarize the role of the microtubules' network during polyomavirus infection. Polyomaviruses usurp microtubules and their motors to travel via early and late acidic endosomes to the endoplasmic reticulum. As shown for SV40, kinesin-1 and microtubules are engaged in the release of partially disassembled virus from the endoplasmic reticulum to the cytosol, and dynein apparently assists in the further disassembly of virions prior to their translocation to the cell nucleus-the place of their replication. Polyomavirus gene products affect the regulation of microtubule dynamics. Early T antigens destabilize microtubules and cause aberrant mitosis. The role of these activities in tumorigenesis has been documented. However, its importance for productive infection remains elusive. On the other hand, in the late phase of infection, the major capsid protein, VP1, of the mouse polyomavirus, counteracts T-antigen-induced destabilization. It physically binds microtubules and stabilizes them. The interaction results in the G2/M block of the cell cycle and prolonged S phase, which is apparently required for successful completion of the viral replication cycle.


Assuntos
Proteínas do Capsídeo/metabolismo , Núcleo Celular/virologia , Interações Hospedeiro-Patógeno , Microtúbulos/fisiologia , Microtúbulos/virologia , Polyomavirus/patogenicidade , Animais , Proteínas do Capsídeo/genética , Citosol/virologia , Retículo Endoplasmático/virologia , Endossomos/virologia , Humanos , Camundongos , Polyomavirus/genética , Ligação Proteica , Replicação Viral
10.
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
11.
J Med Virol ; 91(5): 856-864, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30609063

RESUMO

Active infection with BK polyomavirus (BKPyV) may cause serious complications in transplantation settings. Recently, the level of BKPyV IgG seroreactivity in graft donors has been shown to predict viremia and BKPyV-associated nephropathy in kidney transplant (KTx) recipients. Pretransplantation testing of the donor and recipient BKPyV serostatus could, therefore, identify patients at high risk. For the development of serological immunoassays, antibody response to the predominant BKPyV subtypes (BKPyV-I and BKPyV-IV) was studied using virus-like particle (VLP)-based enzyme-linked immunosorbent assay (ELISA). VLPs made from the capsid protein, VP1, derived from BKPyV-I and BKPyV-IV subtypes were produced using a baculovirus expression system and used as antigens. The tests were used for IgG antibody determination in 50 KTx recipients and 111 healthy blood donors. While 87% of samples reacted with mixed BKPyV-I and BKPyV-IV antigens, only 49% of samples were reactive in both ELISA tests when using BKPyV-I or BKPyV-IV antigens separately. Twenty-seven percent of healthy blood donors and 26% of KTx recipients were reactive only with BKPyV-I, while 9% and 20% were reactive only with BKPyV-IV, respectively. To determine the specificities of the antigens, selected seropositive samples were retested after preadsorption with soluble BKPyV-I, BKPyV-IV, or JC polyomavirus antigens. The experiments confirmed that recombinant VP1 VLP-based ELISAs predominantly detected BKPyV type-specific antibodies. The results imply that anti-BKPyV antibody ELISA tests should contain a mixture of subtype-specific VLP-based antigens instead of antigen derived from the most prevalent BKPyV-I subtype. The tests can be used for serological surveys of BKPyV infection and improved KTx patient management.


Assuntos
Anticorpos Antivirais/sangue , Vírus BK/imunologia , Transplante de Rim , Infecções por Polyomavirus/epidemiologia , Transplantados , República Tcheca/epidemiologia , Ensaio de Imunoadsorção Enzimática/métodos , Humanos , Imunoglobulina G/sangue , Estudos Soroepidemiológicos
12.
Viruses ; 10(4)2018 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-29614718

RESUMO

The mechanism used by mouse polyomavirus (MPyV) overcomes the crowded cytosol to reach the nucleus has not been fully elucidated. Here, we investigated the involvement of importin α/ß1 mediated transport in the delivery of MPyV genomes into the nucleus. Interactions of the virus with importin ß1 were studied by co-immunoprecipitation and proximity ligation assay. For infectivity and nucleus delivery assays, the virus and its capsid proteins mutated in the nuclear localization signals (NLSs) were prepared and produced. We found that at early times post infection, virions bound importin ß1 in a time dependent manner with a peak of interactions at 6 h post infection. Mutation analysis revealed that only when the NLSs of both VP1 and VP2/3 were disrupted, virus did not bind efficiently to importin ß1 and its infectivity remarkably decreased (by 80%). Nuclear targeting of capsid proteins was improved when VP1 and VP2 were co-expressed. VP1 and VP2 were effectively delivered into the nucleus, even when one of the NLS, either VP1 or VP2, was disrupted. Altogether, our results showed that MPyV virions can use VP1 and/or VP2/VP3 NLSs in concert or individually to bind importins to deliver their genomes into the cell nucleus.


Assuntos
Proteínas do Capsídeo/metabolismo , DNA Viral/metabolismo , Carioferinas/metabolismo , Infecções por Polyomavirus/metabolismo , Infecções por Polyomavirus/virologia , Polyomavirus/fisiologia , Substituição de Aminoácidos , Animais , Transporte Biológico , Proteínas do Capsídeo/genética , Linhagem Celular , Núcleo Celular , Imunofluorescência , Camundongos , Mutação , Sinais de Localização Nuclear/genética , Polyomavirus/ultraestrutura , Ligação Proteica , Montagem de Vírus
13.
Mol Pharm ; 15(8): 2932-2945, 2018 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-29389139

RESUMO

Glutamate carboxypeptidase II (GCPII) is a membrane protease overexpressed by prostate cancer cells and detected in the neovasculature of most solid tumors. Targeting GCPII with inhibitor-bearing nanoparticles can enable recognition, imaging, and delivery of treatments to cancer cells. Compared to methods based on antibodies and other large biomolecules, inhibitor-mediated targeting benefits from the low molecular weight of the inhibitor molecules, which are typically stable, easy-to-handle, and able to bind the enzyme with very high affinity. Although GCPII is established as a molecular target, comparing previously reported results is difficult due to the different methodological approaches used. In this work, we investigate the robustness and limitations of GCPII targeting with a diverse range of inhibitor-bearing nanoparticles (various structures, sizes, bionanointerfaces, conjugation chemistry, and surface densities of attached inhibitors). Polymer-coated nanodiamonds, virus-like particles based on bacteriophage Qß and mouse polyomavirus, and polymeric poly(HPMA) nanoparticles with inhibitors attached by different means were synthesized and characterized. We evaluated their ability to bind GCPII and interact with cancer cells using surface plasmon resonance, inhibition assay, flow cytometry, and confocal microscopy. Regardless of the diversity of the investigated nanosystems, they all strongly interact with GCPII (most with low picomolar Ki values) and effectively target GCPII-expressing cells. The robustness of this approach was limited only by the quality of the nanoparticle bionanointerface, which must be properly designed by adding a sufficient density of hydrophilic protective polymers. We conclude that the targeting of cancer cells overexpressing GCPII is a viable approach transferable to a broad diversity of nanosystems.


Assuntos
Antineoplásicos/administração & dosagem , Inibidores Enzimáticos/administração & dosagem , Glutamato Carboxipeptidase II/antagonistas & inibidores , Nanoconjugados/química , Neoplasias/tratamento farmacológico , Antígenos de Superfície/metabolismo , Linhagem Celular Tumoral , Química Farmacêutica , Química Click , Glutamato Carboxipeptidase II/metabolismo , Humanos , Interações Hidrofóbicas e Hidrofílicas , Ligantes , Neoplasias/patologia , Proteínas Recombinantes/metabolismo , Tiazolidinas/química
14.
PLoS One ; 12(9): e0184870, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28922413

RESUMO

The aim of this study was to develop a suitable vaccine antigen against porcine circovirus 2 (PCV2), the causative agent of post-weaning multi-systemic wasting syndrome, which causes significant economic losses in swine breeding. Chimeric antigens containing PCV2b Cap protein sequences based on the mouse polyomavirus (MPyV) nanostructures were developed. First, universal vectors for baculovirus-directed production of chimeric MPyV VLPs or pentamers of the major capsid protein, VP1, were designed for their exploitation as vaccines against other pathogens. Various strategies were employed based on: A) exposure of selected immunogenic epitopes on the surface of MPyV VLPs by insertion into a surface loop of the VP1 protein, B) insertion of foreign protein molecules inside the VLPs, or C) fusion of a foreign protein or its part with the C-terminus of VP1 protein, to form giant pentamers of a chimeric protein. We evaluated these strategies by developing a recombinant vaccine against porcine circovirus 2. All candidate vaccines induced the production of antibodies against the capsid protein of porcine circovirus after immunization of mice. The candidate vaccine, Var C, based on fusion of mouse polyomavirus and porcine circovirus capsid proteins, could induce the production of antibodies with the highest PCV2 neutralizing capacity. Its ability to induce the production of neutralization antibodies was verified after immunization of pigs. The advantage of this vaccine, apart from its efficient production in insect cells and easy purification, is that it represents a DIVA (differentiating infected from vaccinated animals) vaccine, which also induces an immune response against the mouse polyoma VP1 protein and is thus able to distinguish between vaccinated and naturally infected animals.


Assuntos
Proteínas do Capsídeo , Circovirus , Nanoestruturas , Polyomavirus , Proteínas Recombinantes de Fusão , Vacinas Virais , Animais , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/imunologia , Circovirus/genética , Circovirus/imunologia , Camundongos , Polyomavirus/genética , Polyomavirus/imunologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Células Sf9 , Spodoptera , Suínos , Vacinas Virais/genética , Vacinas Virais/imunologia , Vacinas Virais/farmacologia
15.
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
16.
Bioconjug Chem ; 28(2): 307-313, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-28035816

RESUMO

Virus-like particles based on polyomaviruses (PVLPs) are promising delivery devices for various cargoes, including nucleic acids, imaging probes, and therapeutic agents. In biological environments, the major coat protein VP1 interacts with ubiquitously distributed sialic acid residues, and therefore PVLPs show a broad tropism. For selective targeting, appropriate engineering of the PVLP surface is needed. Here, we describe a chemical approach to retarget PVLPs to cancer cells displaying abnormally high levels of transferrin receptor. We created an array of transferrin molecules on the surface of PVLPs by combining a high-yielding bioconjugation approach with specific point modification of transferrin. This artificial surface protein architecture enables (i) suppression of natural VP1-specific interactions by blocking the surface conformational epitope on the VP1 protein, (ii) unusually high cellular uptake efficiency, and (iii) selective retargeting of PVLPs to osteosarcoma (U2OS) and lymphoblastoid leukemia (CCRF-CEM) cells.


Assuntos
Capsídeo/química , Portadores de Fármacos/química , Polyomavirus/química , Transporte Biológico , Capsídeo/metabolismo , Linhagem Celular Tumoral , Portadores de Fármacos/metabolismo , Humanos , Modelos Moleculares , Conformação Molecular , Propriedades de Superfície
17.
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
18.
ACS Appl Mater Interfaces ; 8(38): 25127-36, 2016 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-27589368

RESUMO

A simple nanoprecipitation method was used for preparation of stable photoactive polystyrene nanoparticles (NPs, diameter 30 ± 10 nm) from sulfonated electrospun polystyrene nanofiber membranes with encapsulated 5,10,15,20-tetraphenylporphyrin (TPP) or platinum octaethylporphyrin (Pt-OEP). The NPs prepared with TPP have strong antibacterial and antiviral properties and can be applied to the photooxidation of external substrates based on photogenerated singlet oxygen. In contrast to nanofiber membranes, which have limited photooxidation ability near the surface, NPs are able to travel toward target species/structures. NPs with Pt-OEP were used for oxygen sensing in aqueous media, and they presented strong linear responses to a broad range of oxygen concentrations. The nanofiber membranes can be applied not only as a source of NPs but also as an effective filter for their removal from solution.


Assuntos
Nanopartículas , Antibacterianos , Antivirais , Oxigênio , Oxigênio Singlete
19.
Curr Protoc Microbiol ; 38: 14F.1.1-26, 2015 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-26237106

RESUMO

Mouse polyomavirus (MPyV) is a member of the Polyomaviridae family, which comprises non-enveloped tumorigenic viruses infecting various vertebrates including humans and causing different pathogenic responses in the infected organisms. Despite the variations in host tropism and pathogenicity, the structure of the virions of these viruses is similar. The capsid, with icosahedral symmetry (ø, 45 nm, T = 7d), is composed of a shell of 72 capsomeres of structural proteins, arranged around the nucleocore containing approximately 5-kbp-long circular dsDNA in complex with cellular histones. MPyV has been one of the most studied polyomaviruses and serves as a model virus for studies of the mechanisms of cell transformation and virus trafficking, and for use in nanotechnology. It can be propagated in primary mouse cells (e.g., in whole mouse embryo cells) or in mouse epithelial or fibroblast cell lines. In this unit, propagation, purification, quantification, and storage of MPyV virions are presented.


Assuntos
Polyomavirus/crescimento & desenvolvimento , Polyomavirus/isolamento & purificação , Preservação Biológica/métodos , Carga Viral/métodos , Cultura de Vírus/métodos , Animais , Células Cultivadas , Camundongos
20.
Viruses ; 6(7): 2899-937, 2014 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-25055856

RESUMO

To get access to the replication site, small non-enveloped DNA viruses have to cross the cell membrane using a limited number of capsid proteins, which also protect the viral genome in the extracellular environment. Most of DNA viruses have to reach the nucleus to replicate. The capsid proteins involved in transmembrane penetration are exposed or released during endosomal trafficking of the virus. Subsequently, the conserved domains of capsid proteins interact with cellular membranes and ensure their efficient permeabilization. This review summarizes our current knowledge concerning the role of capsid proteins of small non-enveloped DNA viruses in intracellular membrane perturbation in the early stages of infection.


Assuntos
Proteínas do Capsídeo/metabolismo , Membrana Celular/virologia , Células Eucarióticas/virologia , Internalização do Vírus , Adenoviridae/fisiologia , Proteínas do Capsídeo/química , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Núcleo Celular/virologia , Endossomos/metabolismo , Endossomos/virologia , Células Eucarióticas/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Papillomaviridae/fisiologia , Parvoviridae/fisiologia , Polyomaviridae/fisiologia , Ligação Proteica , Transporte Proteico , Receptores Virais/metabolismo , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA