Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Microbiol Spectr ; 12(6): e0391423, 2024 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-38742903

RESUMO

Porcine parvovirus (PPV) is one of the most important pathogens that cause reproductive failure in pigs. However, the pathogenesis of PPV infection remains unclear. Proteomics is a powerful tool to understand the interaction between virus and host cells. In the present study, we analyzed the proteomics of PPV-infected PK-15 cells. A total of 32 and 345 proteins were differentially expressed at the early and replication stages, respectively. Subsequent gene ontology annotation and Kyoto Encyclopedia of Genes and Genomes enrichment analysis showed these differentially expressed proteins were significantly enriched in pathways including toll-like receptor signaling pathway, tumor necrosis factor signaling pathway, and viral carcinogenesis. The expression of poly (rC) binding protein 1 (PCBP1) was observed to decrease after PPV infection. Overexpressed or silenced PCBP1 expression inhibited or promoted PPV infection. Our studies established a foundation for further exploration of the multiplication mechanism of PPV. IMPORTANCE: Porcine parvovirus (PPV) is a cause of reproductive failure in the swine industry. Our knowledge of PPV remains limited, and there is no effective treatment for PPV infection. Proteomics of PPV-infected PK-15 cells was conducted to identify differentially expressed proteins at 6 hours post-infection (hpi) and 36 hpi. Gene ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analysis showed that various pathways participate in PPV infection. Poly (rC) binding protein 1 was confirmed to inhibit PPV replication, which provided potential targets for anti-PPV infection. Our findings improve the understanding of PPV infection and pave the way for future research in this area.


Assuntos
Infecções por Parvoviridae , Parvovirus Suíno , Proteômica , Proteínas de Ligação a RNA , Doenças dos Suínos , Replicação Viral , Parvovirus Suíno/genética , Parvovirus Suíno/fisiologia , Animais , Suínos , Linhagem Celular , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Infecções por Parvoviridae/virologia , Infecções por Parvoviridae/metabolismo , Infecções por Parvoviridae/veterinária , Doenças dos Suínos/virologia , Doenças dos Suínos/metabolismo , Doenças dos Suínos/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo
2.
J Virol ; 97(7): e0016123, 2023 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-37367301

RESUMO

Parvoviruses are among the smallest and superficially simplest animal viruses, infecting a broad range of hosts, including humans, and causing some deadly infections. In 1990, the first atomic structure of the canine parvovirus (CPV) capsid revealed a 26-nm-diameter T=1 particle made up of two or three versions of a single protein, and packaging about 5,100 nucleotides of single-stranded DNA. Our structural and functional understanding of parvovirus capsids and their ligands has increased as imaging and molecular techniques have advanced, and capsid structures for most groups within the Parvoviridae family have now been determined. Despite those advances, significant questions remain unanswered about the functioning of those viral capsids and their roles in release, transmission, or cellular infection. In addition, the interactions of capsids with host receptors, antibodies, or other biological components are also still incompletely understood. The parvovirus capsid's apparent simplicity likely conceals important functions carried out by small, transient, or asymmetric structures. Here, we highlight some remaining open questions that may need to be answered to provide a more thorough understanding of how these viruses carry out their various functions. The many different members of the family Parvoviridae share a capsid architecture, and while many functions are likely similar, others may differ in detail. Many of those parvoviruses have not been experimentally examined in detail (or at all in some cases), so we, therefore, focus this minireview on the widely studied protoparvoviruses, as well as the most thoroughly investigated examples of adeno-associated viruses.


Assuntos
Parvoviridae , Animais , Humanos , Capsídeo/ultraestrutura , Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , DNA Viral/metabolismo , Parvoviridae/genética , Parvoviridae/ultraestrutura , Infecções por Parvoviridae/metabolismo , Infecções por Parvoviridae/virologia , Dependovirus/genética , Dependovirus/metabolismo , Dependovirus/ultraestrutura
3.
J Virol ; 96(17): e0111822, 2022 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-35950857

RESUMO

Being nonpathogenic to humans, rodent parvoviruses (PVs) are naturally oncolytic viruses with great potential as anti-cancer agents. As these viruses replicate in the host cell nucleus, they must gain access to the nucleus during infection. The PV minute virus of mice (MVM) and several other PVs transiently disrupt the nuclear envelope (NE) and enter the nucleus through the resulting breaks. However, the molecular basis of this unique nuclear entry pathway remains uncharacterized. In this study, we used MVM as a model to investigate the molecular mechanism by which PVs induce NE disruption during viral nuclear entry. By combining bioinformatics analyses, metabolic labeling assays, mutagenesis, and pharmacological inhibition, we identified a functional myristoylation site at the sequence 78GGKVGH83 of the unique portion of the capsid protein VP1 (VP1u) of MVM. Performing proteolytic cleavage studies with a peptide containing this myristoylation site or with purified virions, we found tryptophan at position 77 of MVM VP1u is susceptible to chymotrypsin cleavage, implying this cleavage exposes G (glycine) 78 at the N-terminus of VP1u for myristoylation. Subsequent experiments using inhibitors of myristoylation and cellular proteases with MVM-infected cells, or an imaging-based quantitative NE permeabilization assay, further indicate protein myristoylation and a chymotrypsin-like activity are essential for MVM to locally disrupt the NE during viral nuclear entry. We thus propose a model for the nuclear entry of MVM in which NE disruption is mediated by VP1u myristoylation after the intact capsid undergoes proteolytic processing to expose the required N-terminal G for myristoylation. IMPORTANCE Rodent parvoviruses (PVs), including minute virus of mice (MVM), have the ability to infect and kill cancer cells and thereby possess great potential in anti-cancer therapy. In fact, some of these viruses are currently being investigated in both preclinical studies and clinical trials to treat a wide variety of cancers. However, the detailed mechanism of how PVs enter the cell nucleus remains unknown. In this study, we for the first time demonstrated a chemical modification called "myristoylation" of a MVM protein plays an essential role in the nuclear entry of the virus. We also showed, in addition to protein myristoylation, a chymotrypsin-like activity, which may come from cellular proteasomes, is required for MVM to get myristoylated and enter the nucleus. These findings deepen our understanding on how MVM and other related PVs infect host cells and provide new insights for the development of PV-based anti-cancer therapies.


Assuntos
Proteínas do Capsídeo , Núcleo Celular , Vírus Miúdo do Camundongo , Infecções por Parvoviridae , Animais , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Linhagem Celular , Núcleo Celular/virologia , Quimotripsina/metabolismo , Camundongos , Vírus Miúdo do Camundongo/fisiologia , Infecções por Parvoviridae/metabolismo , Processamento de Proteína Pós-Traducional
4.
J Virol ; 96(3): e0110321, 2022 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-34851146

RESUMO

Adeno-associated virus (AAV) is extensively used as a viral vector to deliver therapeutic genes during human gene therapy. A high-affinity cellular receptor (AAVR) for most serotypes was recently identified; however, its biological function as a gene product remains unclear. In this study, we used AAVR knockdown cell models to show that AAVR depletion significantly attenuated cells to activate unfolded protein response (UPR) pathways when exposed to the endoplasmic reticulum (ER) stress inducer, tunicamycin. By analyzing three major UPR pathways, we found that ATF6 signaling was most affected in an AAVR-dependent fashion, distinct from CHOP and XBP1 branches. AAVR capacity in UPR regulation required the full native AAVR protein, and AAV2 capsid binding to the receptor altered ATF6 dynamics. Conversely, the transduction efficiency of AAV2 was associated with changes in ATF6 signaling in host cells following treatment with different small molecules. Thus, AAVR served as an inhibitory molecule to repress UPR responses via a specificity for ATF6 signaling, and the AAV2 infection route involved the release from AAVR-mediated ATF6 repression, thereby facilitating viral intracellular trafficking and transduction. IMPORTANCE The native function of the AAVR as an ER-Golgi localized protein is largely unknown. We showed that AAVR acted as a functional molecule to regulate UPR signaling under induced ER stress. AAVR inhibited the activation of the transcription factor, ATF6, whereas receptor binding to AAV2 released the suppression effects. This finding has expanded our understanding of AAV infection biology in terms of the physiological properties of AAVR in host cells. Importantly, our research provides a possible strategy which may improve the efficiency of AAV-mediated gene delivery during gene therapy.


Assuntos
Fator 6 Ativador da Transcrição/metabolismo , Dependovirus/fisiologia , Estresse do Retículo Endoplasmático , Infecções por Parvoviridae/metabolismo , Infecções por Parvoviridae/virologia , Receptores de Superfície Celular/metabolismo , Resposta a Proteínas não Dobradas , Linhagem Celular , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Células HeLa , Hepatócitos , Interações Hospedeiro-Patógeno , Humanos , Especificidade de Órgãos , Receptores de Superfície Celular/genética , Transdução de Sinais , Transdução Genética , Tunicamicina/metabolismo , Replicação Viral
5.
J Exp Med ; 217(12)2020 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-32750141

RESUMO

Severe acute respiratory syndrome-coronavirus 2 (SARS-Cov-2) has caused over 13,000,000 cases of coronavirus disease (COVID-19) with a significant fatality rate. Laboratory mice have been the stalwart of therapeutic and vaccine development; however, they do not support infection by SARS-CoV-2 due to the virus's inability to use the mouse orthologue of its human entry receptor angiotensin-converting enzyme 2 (hACE2). While hACE2 transgenic mice support infection and pathogenesis, these mice are currently limited in availability and are restricted to a single genetic background. Here we report the development of a mouse model of SARS-CoV-2 based on adeno-associated virus (AAV)-mediated expression of hACE2. These mice support viral replication and exhibit pathological findings found in COVID-19 patients. Moreover, we show that type I interferons do not control SARS-CoV-2 replication in vivo but are significant drivers of pathological responses. Thus, the AAV-hACE2 mouse model enables rapid deployment for in-depth analysis following robust SARS-CoV-2 infection with authentic patient-derived virus in mice of diverse genetic backgrounds.


Assuntos
Betacoronavirus/metabolismo , Infecções por Coronavirus/metabolismo , Modelos Animais de Doenças , Interferon Tipo I/metabolismo , Camundongos/genética , Peptidil Dipeptidase A/metabolismo , Pneumonia Viral/metabolismo , Enzima de Conversão de Angiotensina 2 , Animais , COVID-19 , Linhagem Celular Tumoral , Infecções por Coronavirus/patologia , Infecções por Coronavirus/virologia , Dependovirus/genética , Feminino , Humanos , Inflamação/metabolismo , Pulmão/patologia , Pulmão/virologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pandemias , Infecções por Parvoviridae/metabolismo , Infecções por Parvoviridae/virologia , Peptidil Dipeptidase A/genética , Pneumonia Viral/patologia , Pneumonia Viral/virologia , SARS-CoV-2 , Transdução de Sinais/genética , Replicação Viral/genética
6.
Curr Protoc Microbiol ; 58(1): e107, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32639683

RESUMO

Human bocavirus 1 (HBoV1) is a small DNA virus that belongs to the Bocaparvovirus genus of the Parvoviridae family. HBoV1 is a common respiratory pathogen that causes mild to life-threatening acute respiratory tract infections in children and immunocompromised individuals, infecting both the upper and lower respiratory tracts. HBoV1 infection causes death of airway epithelial cells, resulting in airway injury and inflammation. In vitro, HBoV1 only infects well-differentiated (polarized) human airway epithelium cultured at an air-liquid interface (HAE-ALI), but not any dividing human cells. A full-length HBoV1 genome of 5543 nucleotides has been cloned from DNA extracted from a human nasopharyngeal swab into a plasmid called HBoV1 infectious clone pIHBoV1. Transfection of pIHBoV1 replicates efficiently in human embryonic kidney 293 (HEK293) cells and produces virions that are highly infectious. This article describes protocols for production of HBoV1 in HEK293 cells, generation of HAE-ALI cultures, and infection with HBoV1 in HAE-ALI. © 2020 Wiley Periodicals LLC. Basic Protocol 1: Human bocavirus 1 production in HEK293 cells Support Protocol 1: HEK293 cell culture and transfection Support Protocol 2: Quantification of human bocavirus 1 using real-time quantitative PCR Basic Protocol 2: Differentiation of human airway cells at an air-liquid interface Support Protocol 3: Expansion of human airway epithelial cell line CuFi-8 Support Protocol 4: Expansion of human airway basal cells Support Protocol 5: Coating of plastic dishes and permeable membranes of inserts Support Protocol 6: Transepithelial electrical resistance measurement Basic Protocol 3: Human bocavirus 1 infection in human airway epithelium cultured at an air-liquid interface Support Protocol 7: Isolation of infected human airway epithelium cells from inserts Basic Protocol 4: Transduction of airway basal cells with lentiviral vector.


Assuntos
Células Epiteliais/virologia , Bocavirus Humano/metabolismo , Transfecção/métodos , Vírion/metabolismo , Cultura de Vírus/métodos , Diferenciação Celular , Linhagem Celular , Genoma Viral , Células HEK293 , Humanos , Infecções por Parvoviridae/metabolismo , Plasmídeos , Sistema Respiratório/virologia , Replicação Viral
7.
Protein Expr Purif ; 174: 105682, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32502709

RESUMO

Canine parvovirus (CPV) non-structural protein-1 (NS1) plays crucial roles in CPV replication and transcription, as well as pathogenic effects to the host. However, the mechanism was not fully understood. Lack of NS1 antibody is one of the restricting factors for NS1 function investigation. To prepare NS1 monoclonal antibody (mAb), the NS1 epitope (AA461 ~ AA650) gene was amplified by PCR, and inserted into pGEX-4T-1vector to construct the prokaryotic expression vector of GST-tag-fused NS1 epitope gene. The NS1 fusion protein was expressed in E. coli, and purified with GSH-magnetic beads, and then used to immunize BALB/c mice. The mouse splenic lymphocytes were isolated and fused with myeloma cells (SP 2/0) to generate hybridoma cells. After several rounds of screening by ELISA, a hybridoma cell clone (1B8) stably expressing NS1 mAb was developed. A large amount of NS1 mAb was prepared from mouse ascites fluid. The isotype of NS1 mAb was identified as IgG1, which can specifically bind NS1 protein in either CPV-infected cells or NS1 vector-transfected cells, indicating the NS1 mAb is effective in detecting NS1 protein. Meanwhile, we used the NS1 mAb to investigate NS1 dynamic changes by qRT-PCR and location by confocal imaging in CPV-infected host cells and showed that NS1 began to appear in the cells at 12 h after CPV infection and reached the highest level at 42 h, NS1 protein was mainly located in nucleus of the cells. This study provided a necessary condition for further investigation on molecular mechanism of NS1 function and pathogenicity.


Assuntos
Anticorpos Monoclonais Murinos , Anticorpos Antivirais , Epitopos , Infecções por Parvoviridae , Parvovirus Canino , Proteínas não Estruturais Virais , Animais , Anticorpos Monoclonais Murinos/química , Anticorpos Monoclonais Murinos/imunologia , Anticorpos Antivirais/química , Anticorpos Antivirais/imunologia , Linhagem Celular , Epitopos/química , Epitopos/genética , Epitopos/imunologia , Epitopos/metabolismo , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Parvoviridae/imunologia , Infecções por Parvoviridae/metabolismo , Parvovirus Canino/química , Parvovirus Canino/genética , Parvovirus Canino/imunologia , Parvovirus Canino/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/imunologia , Proteínas não Estruturais Virais/metabolismo
8.
J Vet Sci ; 21(3): e50, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32476323

RESUMO

BACKGROUND: Porcine parvovirus (PPV) is a single-stranded DNA virus that causes porcine reproductive failure. It is of critical importance to study PPV pathogenesis for the prevention and control of the disease. NS1, a PPV non-structural protein, is participated in viral DNA replication, transcriptional regulation, and cytotoxicity. Our previous research showed that PPV can activate nuclear factor kappa B (NF-κB) signaling pathway and then up-regulate the expression of interleukin (IL)-6. OBJECTIVES: Herein, the purpose of this study is to determine whether the non-structural protein NS1 of PPV also has the same function. METHODS: Real-time quantitative reverse transcription polymerase chain reaction (RT-qPCR), enzyme-linked immunosorbent assay, western blot, immunofluorescence assay and small interfering RNA (siRNA) were used. RESULTS: Our findings demonstrated that PPV NS1 protein can up-regulate the expression levels of IL-6 and tumor necrosis factor-alpha in a dose-dependent manner. Moreover, PPV NS1 protein was found to induce the phosphorylation of IκBα, then leading to the phosphorylation and nuclear translocation of NF-κB. In addition, the NS1 protein activated the upstream pathways of NF-κB. Meanwhile, TLR2-siRNA assay showed TLR2 plays an important role in the activation of NF-κB signaling pathway induced by PPV-NS1. CONCLUSIONS: These findings indicated that PPV NS1 protein induced the up-regulated of IL-6 expression through activating the TLR2 and NF-κB signaling pathways. In conclusion, these findings provide a new avenue to study the innate immune mechanism of PPV infection.


Assuntos
NF-kappa B/metabolismo , Infecções por Parvoviridae/veterinária , Parvovirus Suíno/fisiologia , Transdução de Sinais , Doenças dos Suínos/genética , Receptor 2 Toll-Like/metabolismo , Proteínas não Estruturais Virais/genética , Animais , Regulação da Expressão Gênica/imunologia , NF-kappa B/genética , Infecções por Parvoviridae/genética , Infecções por Parvoviridae/metabolismo , Parvovirus Suíno/genética , Suínos , Doenças dos Suínos/metabolismo , Receptor 2 Toll-Like/genética , Proteínas não Estruturais Virais/metabolismo
9.
Adv Virus Res ; 106: 39-84, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32327148

RESUMO

Adeno-associated virus (AAV) is a nonenveloped, ssDNA virus in the parvovirus family, which has become one of the leading candidate vectors for human gene therapy. AAV has been studied extensively to identify host cellular factors involved in infection, as well as to identify capsid variants that confer clinically favorable transduction profiles ex vivo and in vivo. Recent advances in technology have allowed for direct genetic approaches to be used to more comprehensively characterize host factors required for AAV infection and allowed for identification of a critical multi-serotype receptor, adeno-associated virus receptor (AAVR). In this chapter, we will discuss the interactions of AAV with its glycan and proteinaceous receptors and describe the host and viral components involved in AAV entry, which requires cellular attachment, endocytosis, trafficking to the trans-Golgi network and nuclear import. AAV serves as a paradigm for entry of nonenveloped viruses. Furthermore, we will discuss the potential of utilizing our increased understanding of virus-host interactions during AAV entry to develop better AAV-based therapeutics, with a focus on host factors and capsid interactions involved in in vivo tropism.


Assuntos
Dependovirus/fisiologia , Ligação Viral , Internalização do Vírus , Animais , Dependovirus/química , Dependovirus/genética , Vetores Genéticos/química , Vetores Genéticos/genética , Vetores Genéticos/fisiologia , Humanos , Infecções por Parvoviridae/genética , Infecções por Parvoviridae/metabolismo , Infecções por Parvoviridae/virologia , Receptores Virais/genética , Receptores Virais/metabolismo
10.
J Virol ; 94(11)2020 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-32213611

RESUMO

Human bocavirus 1 (HBoV1) is a parvovirus that gathers increasing attention due to its pleiotropic role as a pathogen and emerging vector for human gene therapy. Curiously, albeit a large variety of HBoV1 capsid variants has been isolated from human samples, only one has been studied as a gene transfer vector to date. Here, we analyzed a cohort of HBoV1-positive samples and managed to PCR amplify and sequence 29 distinct HBoV1 capsid variants. These differed from the originally reported HBoV1 reference strain in 32 nucleotides or four amino acids, including a frequent change of threonine to serine at position 590. Interestingly, this T590S mutation was associated with lower viral loads in infected patients. Analysis of the time course of infection in two patients for up to 15 weeks revealed a gradual accumulation of T590S, concurrent with drops in viral loads. Surprisingly, in a recombinant vector context, T590S was beneficial and significantly increased titers compared to that of T590 variants but had no major impact on their transduction ability or immunoreactivity. Additional targeted mutations in the HBoV1 capsid identified several residues that are critical for transduction, capsid assembly, or DNA packaging. Our new findings on the phylogeny, infectivity, and immunoreactivity of HBoV1 capsid variants improve our understanding of bocaviral biology and suggest strategies to enhance HBoV1 gene transfer vectors.IMPORTANCE The family of Parvoviridae comprises a wide variety of members that exhibit a unique biology and that are concurrently highly interesting as a scaffold for the development of human gene therapy vectors. A most notable example is human bocavirus 1 (HBoV1), which we and others have recently harnessed to cross-package and deliver recombinant genomes derived from another parvovirus, the adeno-associated virus (AAV). Here, we expanded the repertoire of known HBoV1 variants by cloning 29 distinct HBoV1 capsid sequences from primary human samples and by analyzing their properties as AAV/HBoV1 gene transfer vectors. This led to our discovery of a mutational hot spot at HBoV1 capsid position 590 that accumulated in two patients during natural infection and that lowers viral loads but increases vector yields. Thereby, our study expands our current understanding of HBoV1 biology in infected human subjects and concomitantly provides avenues to improve AAV/HBoV1 gene transfer vectors.


Assuntos
Proteínas do Capsídeo , Capsídeo/metabolismo , DNA Viral , Bocavirus Humano/fisiologia , Infecções por Parvoviridae , Montagem de Vírus , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , DNA Viral/genética , DNA Viral/metabolismo , Terapia Genética , Vetores Genéticos , Células HEK293 , Humanos , Infecções por Parvoviridae/genética , Infecções por Parvoviridae/metabolismo , Infecções por Parvoviridae/patologia , Transdução Genética
11.
J Virol ; 94(4)2020 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-31748386

RESUMO

Parvoviruses are an important platform for gene and cancer therapy. Their cell entry and the following steps, including nuclear import, are inefficient, limiting their use in therapeutic applications. Two models exist on parvoviral nuclear entry: the classical import of the viral capsid using nuclear transport receptors of the importin (karyopherin) family or the direct attachment of the capsid to the nuclear pore complex leading to the local disintegration of the nuclear envelope. Here, by laser scanning confocal microscopy and in situ proximity ligation analyses combined with coimmunoprecipitation, we show that infection requires importin ß-mediated access to the nuclear pore complex and nucleoporin 153-mediated interactions on the nuclear side. The importin ß-capsid interaction continued within the nucleoplasm, which suggests a mixed model of nuclear entry in which the classical nuclear import across the nuclear pore complex is accompanied by transient ruptures of the nuclear envelope, also allowing the passive entry of importin ß-capsid complexes into the nucleus.IMPORTANCE Parvoviruses are small DNA viruses that deliver their DNA into the postmitotic nuclei, which is an important step for parvoviral gene and cancer therapies. Limitations in virus-receptor interactions or endocytic entry do not fully explain the low transduction/infection efficiency, indicating a bottleneck after virus entry into the cytoplasm. We thus investigated the transfer of parvovirus capsids from the cytoplasm to the nucleus, showing that the nuclear import of the parvovirus capsid follows a unique strategy, which differs from classical nuclear import and those of other viruses.


Assuntos
Infecções por Parvoviridae/metabolismo , Parvovirus/metabolismo , beta Carioferinas/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Capsídeo/metabolismo , Proteínas do Capsídeo/genética , Linhagem Celular , Núcleo Celular/virologia , Citoplasma/metabolismo , Citosol/metabolismo , Carioferinas/metabolismo , Membrana Nuclear/metabolismo , Poro Nuclear/metabolismo , Parvovirus/imunologia , Internalização do Vírus , Replicação Viral , alfa Carioferinas/metabolismo
12.
J Virol ; 93(21)2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31391273

RESUMO

Adeno-associated viruses (AAV) are helper-dependent parvoviruses that have been developed into promising gene therapy vectors. Many studies, including a recent unbiased genomic screen, have identified host factors essential for AAV cell entry, but no genome-wide screens that address inhibitory host factors have been reported. Here, we utilize a novel CRISPR screen to identify AAV restriction factors in a human hepatocyte cell line. The major hit from our gain-of-function screen is the apical polarity determinant Crumbs 3 (Crb3). Knockout (KO) of Crb3 enhances AAV transduction, while overexpression exerts the opposite effect. Further, Crb3 appears to restrict AAV transduction in a serotype- and cell type-specific manner. Particularly, for AAV serotype 9 and a rationally engineered AAV variant, we demonstrate that increased availability of galactosylated glycans on the surfaces of Crb3 KO cells, but not the universal AAV receptor, leads to increased capsid attachment and enhanced transduction. We postulate that Crb3 could serve as a key molecular determinant that restricts the availability of AAV glycan attachment factors on the cell surface by maintaining apical-basal polarity and tight junction integrity.IMPORTANCE Adeno-associated viruses (AAVs) have recently emerged at the forefront as gene therapy vectors; however, our understanding of host factors that influence AAV transduction in different cell types is still evolving. In the present study, we perform a genome-scale CRISPR knockout screen to identify cellular host factors that restrict AAV infection in hepatocyte cultures. We discover that Crumbs 3, which determines cellular polarity, also influences the distribution of certain carbohydrate attachment factors on the cell surface. This in turn affects the ability of virions to bind and enter the cells. This study underscores the importance of cell polarity in AAV transduction and provides a potential molecular basis for the differential infectious mechanism(s) in cell culture versus organ systems.


Assuntos
Dependovirus/fisiologia , Hepatócitos/metabolismo , Glicoproteínas de Membrana/metabolismo , Infecções por Parvoviridae/virologia , Sistemas CRISPR-Cas , Capsídeo/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Polaridade Celular , Claudinas/genética , Claudinas/metabolismo , Dependovirus/genética , Expressão Gênica , Técnicas de Inativação de Genes , Hepatócitos/fisiologia , Hepatócitos/virologia , Humanos , Glicoproteínas de Membrana/genética , Infecções por Parvoviridae/metabolismo , Polissacarídeos/metabolismo , Sorogrupo , Junções Íntimas , Transdução Genética , Ligação Viral
13.
J Virol ; 93(17)2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31217242

RESUMO

Sequences derived from parvoviruses (family Parvoviridae) are relatively common in animal genomes, but the functional significance of these endogenous parvoviral element (EPV) sequences remains unclear. In this study, we used a combination of in silico and molecular biological approaches to investigate a fusion gene carried by guinea pigs (genus Cavia) that is partially derived from an EPV. This gene, named enRep-M9l, encodes a predicted polypeptide gene product comprising a partial myosin9-like (M9l) gene fused to a 3' truncated, EPV-encoded replicase. We examined the genomic and phylogenetic characteristics of the EPV locus (enRep) that encodes the viral portions of enRep-M9l, revealing that it derives from an ancient dependoparvovirus (genus Dependoparvovirus) that was incorporated into the guinea pig germ line between approximately 22 and 35 million years ago (MYA). Despite these ancient origins, the regions of the enRep locus that are expressed in the enRep-M9l gene are conserved across multiple species in the family Caviidae (guinea pigs and cavies), consistent with a potential function at the amino acid level. Using molecular biological approaches, we further demonstrated that (i) enRep-M9l mRNA is broadly transcribed in guinea pig cells, (ii) the cloned enRep-M9l transcript can express a protein of the expected size in guinea pig cells in vitro, and (iii) the expressed protein localizes to the cytosol. Our findings demonstrate that, consistent with a functional role, the enRep-M9l fusion gene is evolutionarily conserved, broadly transcribed, and capable of expressing protein.IMPORTANCE DNA from viruses has been "horizontally transferred" to mammalian genomes during evolution, but the impact of this process on mammalian biology remains poorly understood. The findings of our study indicate that a novel gene has evolved in guinea pigs through fusion of host and virus genes.


Assuntos
DNA Polimerase Dirigida por DNA/genética , Miosinas/genética , Infecções por Parvoviridae/virologia , Parvovirus/enzimologia , Proteínas Recombinantes de Fusão/metabolismo , Sequência de Aminoácidos , Animais , DNA Polimerase Dirigida por DNA/metabolismo , Evolução Molecular , Células Germinativas/virologia , Cobaias , Miosinas/metabolismo , Infecções por Parvoviridae/metabolismo , Parvovirus/genética , Filogenia , Homologia de Sequência de Aminoácidos , Proteínas Virais/genética , Proteínas Virais/metabolismo
14.
Cell Rep ; 27(13): 3780-3789.e4, 2019 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-31242412

RESUMO

Genetically engineered mouse models harboring large sequence insertions or modifications are critical for a wide range of applications including endogenous gene tagging, conditional knockout, site-specific transgene insertion, and gene replacement; however, existing methods to generate such animals remain laborious and costly. To address this, we developed an approach called CRISPR-READI (CRISPR RNP electroporation and AAV donor infection), combining adeno-associated virus (AAV)-mediated HDR donor delivery with Cas9/sgRNA RNP electroporation to engineer large site-specific modifications in the mouse genome with high efficiency and throughput. We successfully targeted a 774 bp fluorescent reporter, a 2.1 kb CreERT2 driver, and a 3.3 kb expression cassette into endogenous loci in both embryos and live mice. CRISPR-READI is applicable to most widely used knockin schemes requiring donor lengths within the 4.9 kb AAV packaging capacity. Altogether, CRISPR-READI is an efficient, high-throughput, microinjection-free approach for sophisticated mouse genome engineering with potential applications in other mammalian species.


Assuntos
Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Dependovirus , Eletroporação , Técnicas de Introdução de Genes , Infecções por Parvoviridae , Ribonucleoproteínas , Animais , Dependovirus/genética , Dependovirus/metabolismo , Feminino , Camundongos , Camundongos Transgênicos , Infecções por Parvoviridae/genética , Infecções por Parvoviridae/metabolismo , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo
15.
Infect Genet Evol ; 73: 66-70, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31026603

RESUMO

Human papillomavirus (HPV) is one of the most common oncogenic viruses which cause malignancy in different epithelial surfaces of the human body and its infection is the main cause of cervical cancer. However, research suggests that this virus might not be the sole cause of infection in target cells. It is believed that, other infectious agents could co-infect the same cell with HPV including; bacteria, viruses, and parasites, which may have different effects on the carcinogenesis of HPV infections. One of the most important viruses is adeno-associated virus (AAV), which comes from the parvoviridae family. The function of this virus is associated with several stages of HPV carcinogenicity, which leads to the suppression of HPV oncogenesis. The inhibition effects of AAV are exerted not only in viral parts but also in cellular parts. This suppression illuminates a new therapeutic approach in the way of HPV-associated cervical cancer. In the present review we consider the exact roles of AAV infection in this suppression.


Assuntos
Coinfecção , Dependovirus/fisiologia , Interações Microbianas , Papillomaviridae/fisiologia , Infecções por Papillomavirus/virologia , Infecções por Parvoviridae/virologia , Apoptose , Biomarcadores , Suscetibilidade a Doenças , Interações Hospedeiro-Patógeno , Humanos , Infecções por Papillomavirus/metabolismo , Infecções por Parvoviridae/metabolismo
16.
Proc Natl Acad Sci U S A ; 115(15): E3529-E3538, 2018 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-29581310

RESUMO

Adeno-associated virus (AAV) is a small human Dependovirus whose low immunogenicity and capacity for long-term persistence have led to its widespread use as vector for gene therapy. Despite great recent successes in AAV-based gene therapy, further improvements in vector technology may be hindered by an inadequate understanding of various aspects of basic AAV biology. AAV is unique in that its replication is largely dependent on a helper virus and cellular factors. In the absence of helper virus coinfection, wild-type AAV establishes latency through mechanisms that are not yet fully understood. Challenging the currently held model for AAV latency, we show here that the corepressor Krüppel-associated box domain-associated protein 1 (KAP1) binds the latent AAV2 genome at the rep ORF, leading to trimethylation of AAV2-associated histone 3 lysine 9 and that the inactivation of KAP1 repression is necessary for AAV2 reactivation and replication. We identify a viral mechanism for the counteraction of KAP1 in which interference with the KAP1 phosphatase protein phosphatase 1 (PP1) by the AAV2 Rep proteins mediates enhanced phosphorylation of KAP1-S824 and thus relief from KAP1 repression. Furthermore, we show that this phenomenon involves recruitment of the NIPP1 (nuclear inhibitor of PP1)-PP1α holoenzyme to KAP1 in a manner dependent upon the NIPP1 FHA domain, identifying NIPP1 as an interaction partner for KAP1 and shedding light on the mechanism through which PP1 regulates cellular KAP1 activity.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Dependovirus/metabolismo , Receptores de Neuropeptídeo Y/antagonistas & inibidores , Proteína 28 com Motivo Tripartido/metabolismo , Proteínas Virais/metabolismo , Linhagem Celular , Replicação do DNA/fisiologia , DNA Viral/genética , Proteínas de Ligação a DNA/genética , Dependovirus/genética , Epigênese Genética , Genoma Viral , Células HEK293 , Células HeLa , Humanos , Infecções por Parvoviridae/metabolismo , Infecções por Parvoviridae/virologia , Receptores de Neuropeptídeo Y/metabolismo , Proteínas Virais/genética , Vírion/metabolismo , Latência Viral , Replicação Viral/fisiologia
17.
Biol Reprod ; 98(4): 558-569, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29360968

RESUMO

Porcine parvovirus (PPV) is a major virus that leads to fetal death in swine. However, the effects of PPV infection on sows are poorly understood. The aim of this study was to investigate the effects of PPV on porcine steroidogenic luteal cells (SLCs) survival and functions and underlying mechanisms. In vivo experiment results showed that artificial infection of PPV significantly reduced the concentration of serum progesterone and induced histopathological lesions and SLCs apoptosis in porcine corpora luteum. In in vitro cultured primary porcine SLCs, PPV could infect and replicate in SLCs and induced SLCs apoptosis through mitochondria, but not the death receptor, mediated apoptosis pathway. Meanwhile, PPV infection also decreased progesterone production in SLCs. Moreover, PPV infection could increase active p53 transcriptional activity and protein expression as well as promoting p53 translocation to nucleus. Using the p53-specific pharmacological inhibitor (pifithrin-α) and siRNA could partially attenuate PPV-induced Bax upregulation, caspase-3 activation, apoptosis, and the reduction of progesterone production in primary porcine SLCs. Furthermore, the phosphorylation of p38 mitogen-activated protein kinase (MAPK) was also increased in PPV-infected SLCs. Pretreatment with p38 MAPK inhibitor (SB203580) suppressed PPV-induced p53 accumulation and translocation, SLCs apoptosis, and progesterone production reduction. In summary, these findings indicate that PPV could induce SLCs apoptosis and a decrease of progesterone production in vivo and in vitro via p38 MAPK signaling and p53-dependent mitochondrial pathway, which provides the potential clinical therapy methods for PPV infection.


Assuntos
Apoptose/fisiologia , Células Lúteas/metabolismo , Mitocôndrias/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Infecções por Parvoviridae/veterinária , Progesterona/biossíntese , Proteína Supressora de Tumor p53/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Animais , Feminino , Células Lúteas/virologia , Mitocôndrias/virologia , Infecções por Parvoviridae/metabolismo , Parvovirus Suíno , Fosforilação , Transdução de Sinais/fisiologia , Suínos
18.
Biochemistry ; 55(47): 6577-6593, 2016 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-27809499

RESUMO

Infection with human parvovirus B19 (B19V) has been associated with a myriad of illnesses, including erythema infectiosum (Fifth disease), hydrops fetalis, arthropathy, hepatitis, and cardiomyopathy, and also possibly the triggering of any number of different autoimmune diseases. B19V NS1 is a multidomain protein that plays a critical role in viral replication, with predicted nuclease, helicase, and gene transactivation activities. Herein, we investigate the biochemical activities of the nuclease domain (residues 2-176) of B19V NS1 (NS1-nuc) in sequence-specific DNA binding of the viral origin of replication sequences, as well as those of promoter sequences, including the viral p6 and the human p21, TNFα, and IL-6 promoters previously identified in NS1-dependent transcriptional transactivation. NS1-nuc was found to bind with high cooperativity and with multiple (five to seven) copies to the NS1 binding elements (NSBE) found in the viral origin of replication and the overlapping viral p6 promoter DNA sequence. NS1-nuc was also found to bind cooperatively with at least three copies to the GC-rich Sp1 binding sites of the human p21 gene promoter. Only weak or nonspecific binding of NS1-nuc to the segments of the TNFα and IL-6 promoters was found. Cleavage of DNA by NS1-nuc occurred at the expected viral sequence (the terminal resolution site), but only in single-stranded DNA, and NS1-nuc was found to covalently attach to the 5' end of the DNA at the cleavage site. Off-target cleavage by NS1-nuc was also identified.


Assuntos
DNA Viral/metabolismo , Proteínas de Ligação a DNA/metabolismo , DNA/metabolismo , Parvovirus B19 Humano/metabolismo , Proteínas não Estruturais Virais/metabolismo , Sequência de Bases , Sítios de Ligação/genética , Inibidor de Quinase Dependente de Ciclina p21/genética , DNA/genética , Replicação do DNA/genética , DNA de Cadeia Simples/genética , DNA de Cadeia Simples/metabolismo , DNA Viral/genética , Proteínas de Ligação a DNA/genética , Interações Hospedeiro-Patógeno/genética , Humanos , Interleucina-6/genética , Modelos Genéticos , Infecções por Parvoviridae/genética , Infecções por Parvoviridae/metabolismo , Infecções por Parvoviridae/virologia , Parvovirus B19 Humano/genética , Parvovirus B19 Humano/fisiologia , Regiões Promotoras Genéticas/genética , Fator de Necrose Tumoral alfa/genética , Proteínas não Estruturais Virais/genética , Replicação Viral/genética
19.
Virology ; 498: 149-163, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27584592

RESUMO

Old World monkey (OWM) and hominid APOBEC3Aproteins exhibit differential restriction activities against lentiviruses and DNA viruses. Human APOBEC3A(hA3A)has weak restriction activity against HIV-1Δvifbut is efficiently restricted by an artificially generated chimeric from mandrills (mndA3A/G). We show that a chimeric hA3Acontaining the "WVS" insertion (hA3A[(27)WVS(29)]) conferred potent HIV-1restriction activity. Analysis of each amino acid of the "WVS" motif show that the length and not necessarily the charge or hydrophobicity of the amino acids accounted for restriction activity. Our results suggest that hA3A[(27)WVS(29)]restricts HIV-1at the level of reverse transcription in target cells. Finally, our results suggest that insertion of "WVS" into hA3Amodestly reduces restriction of adeno-associated virus 2(AAV-2)while insertion of the AC Loop1region of the mndA3A/G into hA3A abolished AAV-2 restriction, strengthening the role of this molecular interface in the functional evolution of primate A3A.


Assuntos
Aminoácidos/genética , Citidina Desaminase/genética , Citidina Desaminase/metabolismo , Resistência à Doença , Infecções por HIV/metabolismo , Infecções por HIV/virologia , Mutagênese Insercional , Infecções por Parvoviridae/metabolismo , Infecções por Parvoviridae/virologia , Proteínas/genética , Proteínas/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Citidina Desaminase/química , Dependovirus , HIV-1 , Interações Hospedeiro-Patógeno , Humanos , Transporte Proteico , Proteínas/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Deleção de Sequência , Proteínas Virais/metabolismo , Replicação Viral
20.
Sci Rep ; 6: 29560, 2016 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-27406444

RESUMO

Canine parvovirus (CPV) reproduces by co-opting the resources of host cells, inevitably causing cytotoxic effects to the host cells. Feline kidney F81 cells are sensitive to CPV infection and show disparate growing statuses at different time points post-infection. This study analysed the response of F81 cells to CPV infection at successive infection time points by iTRAQ-based quantitative proteomics. Differentially expressed proteins (DEPs) during 60 h of infection and at selected time points post-infection were identified by an analysis of variance test and a two-tailed unpaired t test, respectively. DEPs with similar quantitative changes were clustered by hierarchical clustering and analysed by gene ontology enrichment, revealing that 12 h and 60 h post-infection were the optimal times to analyse the autonomous parvovirus replication and apoptosis processes, respectively. Using the Metacore(TM) database, 29 DEPs were enriched in a network involved in p53 regulation. Besides, a significantly enriched pathway suggests that the CPV-induced cytopathic effect was probably due to the deficiency of functional CFTR caused by CPV infection. This study uncovered the systemic changes in key cellular factors involved in CPV infection and help to understand the molecular mechanisms of the anti-cancer activity of CPV and the cytopathic effects induced by CPV infection.


Assuntos
Panleucopenia Felina/metabolismo , Redes Reguladoras de Genes , Infecções por Parvoviridae/veterinária , Parvovirus Canino/fisiologia , Proteômica/métodos , Animais , Gatos , Linhagem Celular , Regulador de Condutância Transmembrana em Fibrose Cística , Panleucopenia Felina/virologia , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , Infecções por Parvoviridae/metabolismo , Infecções por Parvoviridae/virologia , Mapas de Interação de Proteínas , Tempo , Proteína Supressora de Tumor p53/metabolismo , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA