Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 151
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
PLoS Pathog ; 19(5): e1011203, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37253065

RESUMO

The oncolytic autonomous parvovirus Minute Virus of Mice (MVM) establishes infection in the nuclear environment by usurping host DNA damage signaling proteins in the vicinity of cellular DNA break sites. MVM replication induces a global cellular DNA Damage Response (DDR) that is dependent on signaling by the ATM kinase and inactivates the cellular ATR-kinase pathway. However, the mechanism of how MVM generates cellular DNA breaks remains unknown. Using single molecule DNA Fiber Analysis, we have discovered that MVM infection leads to a shortening of host replication forks as infection progresses, as well as induction of replication stress prior to the initiation of virus replication. Ectopically expressed viral non-structural proteins NS1 and NS2 are sufficient to cause host-cell replication stress, as is the presence of UV-inactivated non-replicative MVM genomes. The host single-stranded DNA binding protein Replication Protein A (RPA) associates with the UV-inactivated MVM genomes, suggesting MVM genomes might serve as a sink for cellular stores of RPA. Overexpressing RPA in host cells prior to UV-MVM infection rescues DNA fiber lengths and increases MVM replication, confirming that MVM genomes deplete RPA stores to cause replication stress. Together, these results indicate that parvovirus genomes induce replication stress through RPA exhaustion, rendering the host genome vulnerable to additional DNA breaks.


Assuntos
Vírus Miúdo do Camundongo , Infecções por Parvoviridae , Parvovirus , Animais , Camundongos , Vírus Miúdo do Camundongo/genética , Proteína de Replicação A/genética , Parvovirus/genética , Replicação Viral/genética , Infecções por Parvoviridae/genética , Replicação do DNA/genética
2.
J Virol ; 97(5): e0046123, 2023 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-37098896

RESUMO

Parvoviruses are single-stranded DNA viruses that utilize host proteins to vigorously replicate in the nuclei of host cells, leading to cell cycle arrest. The autonomous parvovirus, minute virus of mice (MVM), forms viral replication centers in the nucleus which are adjacent to cellular DNA damage response (DDR) sites, many of which are fragile genomic regions prone to undergoing DDR during the S phase. Since the cellular DDR machinery has evolved to transcriptionally suppress the host epigenome to maintain genomic fidelity, the successful expression and replication of MVM genomes at these cellular sites suggest that MVM interacts with DDR machinery distinctly. Here, we show that efficient replication of MVM requires binding of the host DNA repair protein MRE11 in a manner that is independent of the MRE11-RAD50-NBS1 (MRN) complex. MRE11 binds to the replicating MVM genome at the P4 promoter, remaining distinct from RAD50 and NBS1, which associate with cellular DNA break sites to generate DDR signals in the host genome. Ectopic expression of wild-type MRE11 in CRISPR knockout cells rescues virus replication, revealing a dependence on MRE11 for efficient MVM replication. Our findings suggest a new model utilized by autonomous parvoviruses to usurp local DDR proteins that are crucial for viral pathogenesis and distinct from those of dependoparvoviruses, like adeno-associated virus (AAV), which require a coinfected helper virus to inactivate the local host DDR. IMPORTANCE The cellular DNA damage response (DDR) machinery protects the host genome from the deleterious consequences of DNA breaks and recognizes invading viral pathogens. DNA viruses that replicate in the nucleus have evolved distinct strategies to evade or usurp these DDR proteins. We have discovered that the autonomous parvovirus, MVM, which is used to target cancer cells as an oncolytic agent, depends on the initial DDR sensor protein MRE11 to express and replicate efficiently in host cells. Our studies reveal that the host DDR interacts with replicating MVM molecules in ways that are distinct from viral genomes being recognized as simple broken DNA molecules. These findings suggest that autonomous parvoviruses have evolved distinct mechanisms to usurp DDR proteins, which can be used to design potent DDR-dependent oncolytic agents.


Assuntos
Proteína Homóloga a MRE11 , Vírus Miúdo do Camundongo , Infecções por Parvoviridae , Animais , Camundongos , Proteínas de Ciclo Celular/metabolismo , Receptores com Domínio Discoidina/genética , Receptores com Domínio Discoidina/metabolismo , Dano ao DNA , Replicação do DNA , Vírus Miúdo do Camundongo/genética , Infecções por Parvoviridae/genética , Replicação Viral/fisiologia , Proteína Homóloga a MRE11/metabolismo
3.
PLoS Pathog ; 16(10): e1009002, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33064772

RESUMO

The autonomous parvovirus Minute Virus of Mice (MVM) localizes to cellular DNA damage sites to establish and sustain viral replication centers, which can be visualized by focal deposition of the essential MVM non-structural phosphoprotein NS1. How such foci are established remains unknown. Here, we show that NS1 localized to cellular sites of DNA damage independently of its ability to covalently bind the 5' end of the viral genome, or its consensus DNA binding sequence. Many of these sites were identical to those occupied by virus during infection. However, localization of the MVM genome to DNA damage sites occurred only when wild-type NS1, but not its DNA-binding mutant was expressed. Additionally, wild-type NS1, but not its DNA binding mutant, could localize a heterologous DNA molecule containing the NS1 binding sequence to DNA damage sites. These findings suggest that NS1 may function as a bridging molecule, helping the MVM genome localize to cellular DNA damage sites to facilitate ongoing virus replication.


Assuntos
Dano ao DNA , Vírus Miúdo do Camundongo/genética , Vírus Miúdo do Camundongo/metabolismo , Proteínas não Estruturais Virais/metabolismo , Animais , Linhagem Celular , Replicação do DNA , DNA Viral/genética , Proteínas de Ligação a DNA/genética , Feminino , Genoma Viral , Humanos , Masculino , Camundongos , Infecções por Parvoviridae/genética , Infecções por Parvoviridae/virologia , Parvovirus/genética , Replicação Viral
4.
BMC Vet Res ; 18(1): 195, 2022 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-35606875

RESUMO

BACKGROUND: Feline parvovirus (FPV) is a member of the family Parvoviridae, which is a major enteric pathogen of cats worldwide. This study aimed to investigate the prevalence of feline parvovirus in Beijing of China and analyze the genetic features of detected viruses. RESULTS: In this study, a total of 60 (8.5%) parvovirus-positive samples were detected from 702 cat fecal samples using parvovirus-specific PCR. The complete VP2 genes were amplified from all these samples. Among them, 55 (91.7%) sequences were characterized as FPV, and the other five (8.3%) were typed as canine parvovirus type 2 (CPV-2) variants, comprised of four CPV-2c and a new CPV-2b strain. In order to investigate the origin of CPV-2 variants in cats, we amplified full-length VP2 genes from seven fecal samples of dogs infected with CPV-2, which were further classified as CPV-2c. The sequences of new CPV-2b/MT270586 and CPV-2c/MT270587 detected from feline samples shared 100% identity with previous canine isolates KT156833 and MF467242 respectively, suggesting the CPV-2 variants circulating in cats might be derived from dogs. Sequence analysis indicated new mutations, Ala91Ser and Ser192Phe, in the FPV sequences, while obtained CPV-2c carried mutations reported in Asian CPV variants, showing they share a common evolutionary pattern with the Asian 2c strains. Interestingly, the FPV sequence (MT270571), displaying four CPV-specific residues, was found to be a putative recombinant sequence between CPV-2c and FPV. Phylogenetic analysis of the VP2 gene showed that amino acid and nucleotide mutations promoted the evolution of FPV and CPV lineages. CONCLUSIONS: Our findings will be helpful to further understand the circulation and evolution of feline and canine parvovirus in Beijing.


Assuntos
Doenças do Gato , Vírus da Panleucopenia Felina , Infecções por Parvoviridae , Animais , Pequim , Doenças do Gato/epidemiologia , Doenças do Gato/genética , Doenças do Gato/virologia , Gatos/virologia , Doenças do Cão/epidemiologia , Doenças do Cão/genética , Doenças do Cão/virologia , Cães , Fezes/virologia , Vírus da Panleucopenia Felina/genética , Vírus da Panleucopenia Felina/isolamento & purificação , Infecções por Parvoviridae/epidemiologia , Infecções por Parvoviridae/genética , Infecções por Parvoviridae/veterinária , Infecções por Parvoviridae/virologia , Parvovirus Canino/genética , Parvovirus Canino/isolamento & purificação , Filogenia
5.
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
6.
Vet Res ; 52(1): 73, 2021 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-34034820

RESUMO

Porcine Parvovirus (PPV), a pathogen causing porcine reproductive disorders, encodes two capsid proteins (VP1 and VP2) and three nonstructural proteins (NS1, NS2 and SAT) in infected cells. The PPV NS2 mRNA is from NS1 mRNA after alternative splicing, yet the corresponding mechanism is unclear. In this study, we identified a PPV NS1 mRNA binding protein SYNCRIP, which belongs to the hnRNP family and has been identified to be involved in host pre-mRNA splicing by RNA-pulldown and mass spectrometry approaches. SYNCRIP was found to be significantly up-regulated by PPV infection in vivo and in vitro. We confirmed that it directly interacts with PPV NS1 mRNA and is co-localized at the cytoplasm in PPV-infected cells. Overexpression of SYNCRIP significantly reduced the NS1 mRNA and protein levels, whereas deletion of SYNCRIP significantly reduced NS2 mRNA and protein levels and the ratio of NS2 to NS1, and further impaired replication of the PPV. Furthermore, we found that SYNCRIP was able to bind the 3'-terminal site of NS1 mRNA to promote the cleavage of NS1 mRNA into NS2 mRNA. Taken together, the results presented here demonstrate that SYNCRIP is a critical molecule in the alternative splicing process of PPV mRNA, while revealing a novel function for this protein and providing a potential target of antiviral intervention for the control of porcine parvovirus disease.


Assuntos
DNA Viral/fisiologia , Ribonucleoproteínas Nucleares Heterogêneas/genética , Infecções por Parvoviridae/veterinária , Parvovirus Suíno/fisiologia , RNA Mensageiro/genética , Doenças dos Suínos/genética , Proteínas não Estruturais Virais/genética , Processamento Alternativo , Animais , Replicação do DNA , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Infecções por Parvoviridae/genética , Infecções por Parvoviridae/metabolismo , Parvovirus Suíno/genética , RNA Mensageiro/metabolismo , Sus scrofa , Suínos , Doenças dos Suínos/metabolismo , Proteínas não Estruturais Virais/metabolismo
7.
Virus Genes ; 57(5): 426-433, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34255270

RESUMO

Enteritis caused by CPV-2 antigenic variants (CPV-2a, 2b, and 2c) is frequently reported in dogs worldwide leading to significant morbidity and mortality. Here, we describe about a simple, single-step, ARMS-PCR strategy targeting the mutant 426 amino acid of VP2 to differentiate CPV-2 antigenic types. A total of 150 fecal samples were subjected to ARMS-PCR of which 18 were typed as CPV-2a, 79 were typed as CPV-2b, and 6 were typed as CPV-2c. The ARMS-PCR results were validated by randomly sequencing partial VP2 gene of 14 samples. Phylogenetic analysis of partial VP2 gene sequencing of each of the CPV-2 variants revealed that CPV-2a and CPV-2b isolates formed a separate clade of Indian lineage, while CPV-2c shared common evolutionary origin with Asian lineage. The developed technique is first of its kind, one-step, rapid, sequencing independent method for typing of CPV-2 antigenic variants.


Assuntos
Proteínas do Capsídeo/isolamento & purificação , Infecções por Parvoviridae/diagnóstico , Parvovirus Canino/isolamento & purificação , Reação em Cadeia da Polimerase/métodos , Animais , Proteínas do Capsídeo/genética , Doenças do Cão/diagnóstico , Doenças do Cão/genética , Doenças do Cão/virologia , Cães , Fezes/virologia , Variação Genética/genética , Mutação/genética , Infecções por Parvoviridae/genética , Infecções por Parvoviridae/veterinária , Infecções por Parvoviridae/virologia , Parvovirus Canino/genética , Reação em Cadeia da Polimerase/veterinária
8.
J Virol ; 93(6)2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30626673

RESUMO

Parvovirus-derived endogenous viral elements (EVEs) have been found in the genomes of many different animal species, resulting from integration events that may have occurred from more than 50 million years ago to much more recently. Here, we further investigate the properties of autonomous parvovirus EVEs and describe their relationships to contemporary viruses. While we did not find any intact capsid protein open reading frames in the integrated viral sequences, we examined three EVEs that were repaired to form full-length sequences with relatively few changes. These sequences were found in the genomes of Rattus norvegicus (brown rat), Mus spretus (Algerian mouse), and Apodemus sylvaticus (wood mouse). The R. norvegicus sequence was not present in the genomes of the closely related species R. rattus, R. tanezumi, R. exulans, and R. everetti, indicating that it was less than 2 million years old, and the M. spretus and A. sylvaticus sequences were not found in the published genomes of other mouse species, also indicating relatively recent insertions. The M. spretus VP2 sequence assembled into capsids, which had high thermal stability, bound the sialic acid N-acetylneuraminic acid, and entered murine L cells. The 3.89-Å structure of the M. spretus virus-like particles (VLPs), determined using cryo-electron microscopy, showed similarities to rodent and porcine parvovirus capsids. The repaired VP2 sequences from R. norvegicus and A. sylvaticus did not assemble as first prepared, but chimeras combining capsid surface loops from R. norvegicus with canine parvovirus assembled, allowing some of that capsid's structures and functions to be examined.IMPORTANCE Parvovirus endogenous viral elements (EVEs) that have been incorporated into the genomes of different animals represent remnants of the DNA sequences of ancient viruses that infected the ancestors of those animals millions of years ago, but we know little about their properties or how they differ from currently circulating parvoviruses. By expressing the capsid proteins of different parvovirus EVEs that were found integrated into the genomes of three different rodents, we can examine their structures and functions. A VP2 (major capsid protein) EVE sequence from a mouse genome assembled into capsids that had a similar structure and biophysical properties to extant parvoviruses and also bound sialic acids and entered rodent cells. Chimeras formed from combinations of canine parvovirus and portions of the parvovirus sequences from the brown rat genome allowed us to examine the structures and functions of the surface loops of that EVE capsid.


Assuntos
Proteínas do Capsídeo/genética , Genoma/genética , Infecções por Parvoviridae/genética , Parvovirus/genética , Roedores/genética , Roedores/virologia , Animais , Capsídeo , Gatos , Linhagem Celular , Cães , Células HEK293 , Humanos , Camundongos , Infecções por Parvoviridae/virologia , Ratos , Células Sf9 , Suínos
9.
Rheumatology (Oxford) ; 59(11): 3526-3532, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32556240

RESUMO

OBJECTIVE: Fibrosis is the most characteristic pathological hallmark of SSc, a connective tissue disease characterized by vascular and immunological abnormalities, inflammation and enhanced extracellular matrix production, leading to progressive fibrosis of skin and internal organs. We previously demonstrated that parvovirus B19 (B19V) can infect normal human dermal fibroblasts (NHDFs) and that B19V persists in SSc fibroblasts. In this study, we investigated whether parvovirus B19V is able to activate in vitro NHDFs and to induce in these cells some phenotypic features similar to that observed in the SSc fibroblasts. METHODS: We preliminarily analysed the time course of B19V infection in cultured NHDFs, then we investigated the ability of B19V to induce cell migration, invasive phenotype and mRNA expression of some profibrotic and/or proinflammatory genes. RESULTS: We confirmed our previous findings that B19V infects NHDFs, but the infection is not productive. After incubation with B19V, NHDFs showed a significant increase of both migration and invasiveness, along with mRNA expression of different profibrotic genes (α-SMA, EDN-1, IL-6, TGF-ß1 receptors 1 and 2, Col1α2), some genes associated with inflammasome platform (AIM2, IFI16, IL-1ß, CASP-1) and genes for metalloprotease (MMP 2, 9 and 12). CONCLUSION: These data suggest that B19V can activate dermal fibroblasts and may have a role in the pathogenesis of fibrosis. B19V-induced fibroblast migration and invasiveness could be due to the B19V-associated MMP9 overexpression and activation. Moreover, the up-regulation of MMP12, typical of SSc, could link the B19V infection of fibroblasts to the anti-angiogenic process.


Assuntos
Movimento Celular , Fibroblastos/metabolismo , Fibrose/genética , Inflamação/genética , Infecções por Parvoviridae/genética , RNA Mensageiro/metabolismo , Actinas/genética , Caspase 1/genética , Células Cultivadas , Colágeno Tipo I/genética , Proteínas de Ligação a DNA/genética , Endotelina-1/genética , Fibroblastos/patologia , Fibroblastos/virologia , Fibrose/patologia , Humanos , Técnicas In Vitro , Interleucina-1beta/genética , Interleucina-6/genética , Metaloproteinase 12 da Matriz/genética , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 9 da Matriz/genética , Proteínas Nucleares/genética , Infecções por Parvoviridae/patologia , Parvovirus B19 Humano , Fosfoproteínas/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Escleroderma Sistêmico/genética , Escleroderma Sistêmico/metabolismo , Escleroderma Sistêmico/patologia , Escleroderma Sistêmico/virologia , Pele/citologia , Pele/patologia , Transcriptoma
10.
Virus Genes ; 56(4): 522-526, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32306155

RESUMO

A novel parvovirus was identified as a cell culture contaminant by metagenomic analysis. Droplet digital PCR (ddPCR) was used to determine viral loads in the cell culture supernatant and further analysis, by ddPCR and DNA sequencing, demonstrated that fetal bovine serum (FBS) used during cell culture was the source of the parvovirus contamination. The FBS contained ~ 50,000 copies of the novel parvovirus DNA per ml of serum. The viral DNA was resistant to DNAse digestion. Near-full length sequence of the novel parvovirus was determined. Phylogenetic analysis demonstrated that virus belongs to the Copiparvovirus genus, being most closely related to bovine parvovirus 2 (BPV2) with 41% identity with the non-structural protein NS1 and 47% identity with the virus capsid protein of BPV2. A screen of individual and pooled bovine sera identified a closely related variant of the novel virus in a second serum pool. For classification purposes, the novel virus has been designated bovine copiparvovirus species 3 isolate JB9 (bocopivirus 3-JB9).


Assuntos
Bocavirus/isolamento & purificação , Metagenômica , Infecções por Parvoviridae/genética , Parvovirinae/isolamento & purificação , Animais , Bovinos , Feto/virologia , Genoma Viral/genética , Infecções por Parvoviridae/virologia , Parvovirinae/genética , Soroalbumina Bovina/genética
11.
Biochem Genet ; 58(1): 63-73, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31250332

RESUMO

Chronic inflammation plays a prominent role in cancer initiation and development. On the other hand, the Inflammation can be established by a number of factors such as viral infections. Parvovirus B19 (B19V) is a pathogen with widespread infection, which infects bone marrow erythroid progenitor cells. It has been shown that B19V can also enter human bone marrow mesenchymal stem cells (BM-MSCs). In this study, we hypothesized that BM-MSCs as the main cellular component of bone marrow niche may be induced to secret pro-inflammatory cytokines after B19V infection. BM-MSCs were cultured up to passage 3. The cells were then subjected to nucleofection to transfer a plasmid containing B19V genome. After 36 h, total RNA was extracted and the expression levels of IL-1ß, IL-6, TNF-α and NF-κB genes were examined using qRT-PCR. Data analysis showed the significant increase in expression levels of all studied genes in the B19V-transfected cells (P < 0.05). Although further researches are required, our findings for the first time suggest the importance of B19V infection to establish an inflammatory microenvironment in the bone marrow and its involvement in inflammation-related diseases. Finally, based on our results, molecular assay to diagnose B19V infection of BM-MSCs prior to stem cell therapy is strongly recommended.


Assuntos
Citocinas/genética , Expressão Gênica , Células-Tronco Mesenquimais/virologia , Infecções por Parvoviridae/genética , Infecções por Parvoviridae/patologia , Parvovirus B19 Humano , Linhagem Celular , Humanos , Inflamação/genética , Inflamação/virologia , Infecções por Parvoviridae/diagnóstico , Regulação para Cima
12.
J Virol ; 91(1)2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27733644

RESUMO

Human bocavirus 1 (HBoV1), an emerging human-pathogenic respiratory virus, is a member of the genus Bocaparvovirus of the Parvoviridae family. In human airway epithelium air-liquid interface (HAE-ALI) cultures, HBoV1 infection initiates a DNA damage response (DDR), activating all three phosphatidylinositol 3-kinase-related kinases (PI3KKs): ATM, ATR, and DNA-PKcs. In this context, activation of PI3KKs is a requirement for amplification of the HBoV1 genome (X. Deng, Z. Yan, F. Cheng, J. F. Engelhardt, and J. Qiu, PLoS Pathog, 12:e1005399, 2016, https://doi.org/10.1371/journal.ppat.1005399), and HBoV1 replicates only in terminally differentiated, nondividing cells. This report builds on the previous discovery that the replication of HBoV1 DNA can also occur in dividing HEK293 cells, demonstrating that such replication is likewise dependent on a DDR. Transfection of HEK293 cells with the duplex DNA genome of HBoV1 induces hallmarks of DDR, including phosphorylation of H2AX and RPA32, as well as activation of all three PI3KKs. The large viral nonstructural protein NS1 is sufficient to induce the DDR and the activation of the three PI3KKs. Pharmacological inhibition or knockdown of any one of the PI3KKs significantly decreases both the replication of HBoV1 DNA and the downstream production of progeny virions. The DDR induced by the HBoV1 NS1 protein does not cause obvious damage to cellular DNA or arrest of the cell cycle. Notably, key DNA replication factors and major DNA repair DNA polymerases (polymerase η [Pol η] and polymerase κ [Pol κ]) are recruited to the viral DNA replication centers and facilitate HBoV1 DNA replication. Our study provides the first evidence of the DDR-dependent parvovirus DNA replication that occurs in dividing cells and is independent of cell cycle arrest. IMPORTANCE: The parvovirus human bocavirus 1 (HBoV1) is an emerging respiratory virus that causes lower respiratory tract infections in young children worldwide. HEK293 cells are the only dividing cells tested that fully support the replication of the duplex genome of this virus and allow the production of progeny virions. In this study, we demonstrate that HBoV1 induces a DDR that plays significant roles in the replication of the viral DNA and the production of progeny virions in HEK293 cells. We also show that both cellular DNA replication factors and DNA repair DNA polymerases colocalize within centers of viral DNA replication and that Pol η and Pol κ play an important role in HBoV1 DNA replication. Whereas the DDR that leads to the replication of the DNA of other parvoviruses is facilitated by the cell cycle, the DDR triggered by HBoV1 DNA replication or NS1 is not. HBoV1 is the first parvovirus whose NS1 has been shown to be able to activate all three PI3KKs (ATM, ATR, and DNA-PKcs).


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/genética , Replicação do DNA , DNA Viral/genética , Proteína Quinase Ativada por DNA/genética , Bocavirus Humano/genética , Proteínas Nucleares/genética , Infecções por Parvoviridae/genética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Divisão Celular , Dano ao DNA , DNA Viral/metabolismo , Proteína Quinase Ativada por DNA/metabolismo , DNA Polimerase Dirigida por DNA/genética , DNA Polimerase Dirigida por DNA/metabolismo , Regulação da Expressão Gênica , Células HEK293 , Repetição Terminal Longa de HIV , Histonas/genética , Histonas/metabolismo , Interações Hospedeiro-Patógeno , Bocavirus Humano/crescimento & desenvolvimento , Bocavirus Humano/metabolismo , Humanos , Proteínas Nucleares/metabolismo , Infecções por Parvoviridae/metabolismo , Infecções por Parvoviridae/virologia , Fosforilação , Regiões Promotoras Genéticas , Proteína de Replicação A/genética , Proteína de Replicação A/metabolismo , Transdução de Sinais , Transcrição Gênica , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Replicação Viral
13.
J Pediatr Hematol Oncol ; 40(8): e550-e552, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29432306

RESUMO

We report a case of a 12-year-old male with glucose-6-phosphate dehydrogenase deficiency presenting with clinical signs of sepsis and pancytopenia. Investigations revealed parvovirus B19 (PVB19)-associated hemophagocytic lymphohistiocytosis (HLH). The patient recovered fully and quickly with symptomatic treatment. Current evidence suggests that PVB19-associated HLH has a favorable prognosis. Mild undiagnosed cases of HLH may be the cause of pancytopenia in PVB19 infections.


Assuntos
Deficiência de Glucosefosfato Desidrogenase , Linfo-Histiocitose Hemofagocítica , Infecções por Parvoviridae , Parvovirus B19 Humano , Criança , Deficiência de Glucosefosfato Desidrogenase/genética , Deficiência de Glucosefosfato Desidrogenase/patologia , Deficiência de Glucosefosfato Desidrogenase/terapia , Humanos , Linfo-Histiocitose Hemofagocítica/diagnóstico , Linfo-Histiocitose Hemofagocítica/genética , Linfo-Histiocitose Hemofagocítica/patologia , Linfo-Histiocitose Hemofagocítica/terapia , Masculino , Infecções por Parvoviridae/diagnóstico , Infecções por Parvoviridae/genética , Infecções por Parvoviridae/patologia , Infecções por Parvoviridae/terapia , Sepse/diagnóstico , Sepse/genética , Sepse/patologia , Sepse/terapia
14.
Indian J Med Res ; 147(3): 239-247, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29923512

RESUMO

Erythrovirus B19 (B19V) is one of the erythroviruses known to be pathogenic in humans. B19V is classified into three distinct genotypes; 1, 2 and 3, differing from each other by 2-13 per cent. Genotype 1 consists of the prototype B19V isolates, genotype 2 comprises the A6, LaLi and their related isolates while genotype 3 includes the V9- and V9-related isolates. The classification of genotype 1 into two subtypes (1A and 1B) and genotype 3 into two subtypes (3a and 3b) with an estimated nucleotide difference of about 5 per cent has been done. Predominance of genotype 1 across all the continents is seen followed by genotypes 2 and 3. There are no disease-specific genotypes. All the three genotypes have been found in symptomatic as well as asymptomatic individuals and have been reported from several countries across the world. The prevalence of genotype 2 in older populations and its absence from current circulation in Northern Europe has also been reported. The present review focuses on geographic distribution and association of genotypes of B19V with different clinical manifestations.


Assuntos
Infecções por Parvoviridae/epidemiologia , Parvovirus B19 Humano/genética , DNA Viral , Erythrovirus , Variação Genética , Genótipo , Humanos , Infecções por Parvoviridae/genética , Parvovirus B19 Humano/patogenicidade
15.
J Virol ; 90(16): 7196-7204, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27252527

RESUMO

UNLABELLED: We previously reported that an amino acid substitution, Y704A, near the 2-fold interface of adeno-associated virus (AAV) was defective for transcription of the packaged genome (M. Salganik, F. Aydemir, H. J. Nam, R. McKenna, M. Agbandje-McKenna, and N. Muzyczka, J Virol 88:1071-1079, 2013, doi: http://dx.doi.org/10.1128/JVI.02093-13). In this report, we have characterized the defect in 6 additional capsid mutants located in a region ∼30 Šin diameter on the surface of the AAV type 2 (AAV2) capsid near the 2-fold interface. These mutants, which are highly conserved among primate serotypes, displayed a severe defect (3 to 6 logs) in infectivity. All of the mutants accumulated significant levels of uncoated DNA in the nucleus, but none of the mutants were able to accumulate significant amounts of genomic mRNA postinfection. In addition, wild-type (wt) capsids that were bound to the conformational antibody A20, which is known to bind the capsid surface in the region of the mutants, were also defective for transcription. In all cases, the mutant virus particles, as well as the antibody-bound wild-type capsids, were able to enter the cell, travel to the nucleus, uncoat, and synthesize a second strand but were unable to transcribe their genomes. Taken together, the phenotype of these mutants provides compelling evidence that the AAV capsid plays a role in the transcription of its genome, and the mutants map this functional region on the surface of the capsid near the 2-fold interface. This appears to be the first example of a viral structural protein that is also involved in the transcription of the viral genome that it delivers to the nucleus. IMPORTANCE: Many viruses package enzymes within their capsids that assist in expressing their genomes postinfection, e.g., retroviruses. A number of nonenveloped viruses, including AAV, carry proteases that are needed for capsid maturation or for capsid modification during infection. We describe here what appears to be the first example of a nonenveloped viral capsid that appears to have a role in promoting transcription. A total of six mutants at the AAV capsid 2-fold interface were shown to have a severe defect in expressing their genomes, and the defect was at the level of mRNA accumulation. This suggests that AAV capsids have a novel role in promoting the transcription of the genomes that they have packaged. Since wt virions could not complement the mutant viruses, and the mutant viruses did not effectively inhibit wt gene expression, our results suggest that the capsid exerts its effect on transcription in cis.


Assuntos
Capsídeo/fisiologia , Dependovirus/genética , Genoma Viral/genética , Mutação/genética , Infecções por Parvoviridae/virologia , Transcrição Gênica/genética , Proteínas Estruturais Virais/genética , Substituição de Aminoácidos , Células HEK293 , Células HeLa , Humanos , Modelos Moleculares , Infecções por Parvoviridae/genética , Infecções por Parvoviridae/metabolismo , Fenótipo , RNA Viral/genética , Proteínas Estruturais Virais/química , Proteínas Estruturais Virais/metabolismo , Vírion
16.
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
17.
PLoS Pathog ; 10(4): e1004055, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24699724

RESUMO

Infection by the autonomous parvovirus minute virus of mice (MVM) induces a vigorous DNA damage response in host cells which it utilizes for its efficient replication. Although p53 remains activated, p21 protein levels remain low throughout the course of infection. We show here that efficient MVM replication required the targeting for degradation of p21 during this time by the CRL4Cdt2 E3-ubiquitin ligase which became re-localized to MVM replication centers. PCNA provides a molecular platform for substrate recognition by the CRL4Cdt2 E3-ubiquitin ligase and p21 targeting during MVM infection required its interaction both with Cdt2 and PCNA. PCNA is also an important co-factor for MVM replication which can be antagonized by p21 in vitro. Expression of a stable p21 mutant that retained interaction with PCNA inhibited MVM replication, while a stable p21 mutant which lacked this interaction did not. Thus, while interaction with PCNA was important for targeting p21 to the CRL4Cdt2 ligase re-localized to MVM replication centers, efficient viral replication required subsequent depletion of p21 to abrogate its inhibition of PCNA.


Assuntos
Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Vírus Miúdo do Camundongo/fisiologia , Proteínas Nucleares/metabolismo , Infecções por Parvoviridae/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Replicação Viral/fisiologia , Animais , Linhagem Celular , Inibidor de Quinase Dependente de Ciclina p21/genética , Humanos , Camundongos , Mutação , Proteínas Nucleares/genética , Infecções por Parvoviridae/genética , Antígeno Nuclear de Célula em Proliferação/genética , Ubiquitina-Proteína Ligases/genética
18.
PLoS Pathog ; 10(1): e1003891, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24415942

RESUMO

Parvoviruses halt cell cycle progression following initiation of their replication during S-phase and continue to replicate their genomes for extended periods of time in arrested cells. The parvovirus minute virus of mice (MVM) induces a DNA damage response that is required for viral replication and induction of the S/G2 cell cycle block. However, p21 and Chk1, major effectors typically associated with S-phase and G2-phase cell cycle arrest in response to diverse DNA damage stimuli, are either down-regulated, or inactivated, respectively, during MVM infection. This suggested that parvoviruses can modulate cell cycle progression by another mechanism. In this work we show that the MVM-induced, p21- and Chk1-independent, cell cycle block proceeds via a two-step process unlike that seen in response to other DNA-damaging agents or virus infections. MVM infection induced Chk2 activation early in infection which led to a transient S-phase block associated with proteasome-mediated CDC25A degradation. This step was necessary for efficient viral replication; however, Chk2 activation and CDC25A loss were not sufficient to keep infected cells in the sustained G2-arrested state which characterizes this infection. Rather, although the phosphorylation of CDK1 that normally inhibits entry into mitosis was lost, the MVM induced DDR resulted first in a targeted mis-localization and then significant depletion of cyclin B1, thus directly inhibiting cyclin B1-CDK1 complex function and preventing mitotic entry. MVM infection thus uses a novel strategy to ensure a pseudo S-phase, pre-mitotic, nuclear environment for sustained viral replication.


Assuntos
Ciclina B1/metabolismo , Vírus Miúdo do Camundongo/metabolismo , Mitose , Infecções por Parvoviridae/metabolismo , Animais , Proteína Quinase CDC2 , Linhagem Celular , Quinase do Ponto de Checagem 2/genética , Quinase do Ponto de Checagem 2/metabolismo , Ciclina B1/genética , Quinases Ciclina-Dependentes/genética , Quinases Ciclina-Dependentes/metabolismo , Ativação Enzimática/genética , Pontos de Checagem da Fase G2 do Ciclo Celular/genética , Humanos , Camundongos , Vírus Miúdo do Camundongo/genética , Infecções por Parvoviridae/genética , Infecções por Parvoviridae/patologia , Pontos de Checagem da Fase S do Ciclo Celular/genética , Fosfatases cdc25/genética , Fosfatases cdc25/metabolismo
19.
Clin Genet ; 90(1): 69-78, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26830532

RESUMO

The aim of this study was to describe the mutational characteristics in Korean hereditary spherocytosis (HS) patients. Relevant literatures including genetically confirmed cases with well-documented clinical summaries and relevant information were also reviewed to investigate the mutational gene- or domain-specific laboratory and clinical association. Twenty-five HS patients carried one heterozygous mutation of ANK1 (n = 13) or SPTB (n = 12) but not in SPTA1, SLC4A1, or EPB42. Deleterious mutations including frameshift, nonsense, and splice site mutations were identified in 91% (21/23), and non-hotspot mutations were dispersed across multiple exons. Genotype-phenotype correlation was clarified after combined analysis of the cases and the literature review; anemia was most severe in HS patients with mutations on the ANK1 spectrin-binding domain (p < 0.05), and SPTB mutations in HS patients spared the tetramerization domain in which mutations of hereditary elliptocytosis and pyropoikilocytosis are located. Splenectomy (17/75) was more frequent in ANK1 mutant HS (32%) than in HS with SPTB mutation (10%) (p = 0.028). Aplastic crisis occurred in 32.0% of the patients (8/25; 3 ANK1 and 5 SPTB), and parvovirus B19 was detected in 88%. The study clarifies ANK1 or SPTB mutational characteristics in HS Korean patients. The genetic association of laboratory and clinical aspects suggests comprehensive considerations for genetic-based management of HS.


Assuntos
Anemia Aplástica/genética , Anquirinas/genética , Mutação , Infecções por Parvoviridae/genética , Espectrina/genética , Esferocitose Hereditária/genética , Adolescente , Adulto , Anemia Aplástica/complicações , Anemia Aplástica/diagnóstico , Anemia Aplástica/cirurgia , Povo Asiático , Criança , Pré-Escolar , Éxons , Expressão Gênica , Estudos de Associação Genética , Estudo de Associação Genômica Ampla , Heterozigoto , Humanos , Lactente , Infecções por Parvoviridae/complicações , Infecções por Parvoviridae/diagnóstico , Infecções por Parvoviridae/cirurgia , Parvovirus B19 Humano/isolamento & purificação , Índice de Gravidade de Doença , Esferocitose Hereditária/complicações , Esferocitose Hereditária/diagnóstico , Esferocitose Hereditária/cirurgia , Esplenectomia
20.
Mol Biol Rep ; 43(3): 175-81, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26878856

RESUMO

Human Parvovirus B19 (PVB19) is one of the most important pathogens that targets erythroid lineage. Many factors were mentioned for restriction to erythroid progenitor cells (EPCs). Previous studies showed that in non-permissive cells VP1 and VP2 (structural proteins) mRNAs were detected but could not translate to proteins. A bioinformatics study showed that this inhibition might be due to specific microRNAs (miRNAs) present in non-permissive cells but not in permissive EPCs. To confirm the hypothesis, we evaluated the effect of miRNAs on VP expression. CD34(+) HSCs were separated from cord blood. Then, CD34(+) cells were treated with differentiation medium to obtain CD36(+) EPCs. To evaluate the effect of miRNAs on VP expression in MCF7 and HEK-293 cell lines (non-permissive cells) and CD36(+) EPCs, dual luciferase assay was performed in presence of shRNAs against Dicer and Drosha to disrupt miRNA biogenesis. QRT-PCR was performed to check down-regulation of Dicer and Drosha after transfection. All measurements were done in triplicate. Data means were compared using one-way ANOVAs. MicroRNA prediction was done by the online microRNA prediction tools. No significant difference was shown in luciferase activity of CD36(+) EPCs after co-transfection with shRNAs, while it was significant in non-permissive cells. Our study revealed that miRNAs may be involved in inhibition of VP expression in non-permissive cells, although further studies are required to demonstrate which miRNAs exactly are involved in regulation of PVB19 replication.


Assuntos
Proteínas do Capsídeo/genética , Células Precursoras Eritroides/metabolismo , MicroRNAs , Infecções por Parvoviridae/metabolismo , Parvovirus B19 Humano/metabolismo , Tropismo , Biologia Computacional , Células Precursoras Eritroides/virologia , Regulação da Expressão Gênica , Células HEK293 , Humanos , Células MCF-7 , Infecções por Parvoviridae/genética , Parvovirus B19 Humano/fisiologia , RNA Mensageiro
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA