Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Virol ; 97(2): e0163122, 2023 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-36749078

RESUMO

B19 virus (B19V) is a pathogenic human parvovirus that infects erythroid progenitor cells. Because there are limited in vitro culture systems to propagate this virus, little is known about the molecular mechanisms by which it propagates in cells. In this study, we introduced a HiBiT peptide tag into various loops of VP2 located on the surface of B19V particles and evaluated their ability to form virus-like particles (VLPs). Three independent sites were identified as permissive sites for peptide tag insertion without affecting VLP formation. When the HiBiT tag was introduced into B19V clones (pB19-M20) and transfected into a semipermissive erythroleukemia cell line (UT7/Epo-S1), HiBiT-dependent luciferase activities (HiBiT activities) increased depending on helicase activity of viral NS1. Furthermore, we used a GFP11 tag-split system to visualize VLPs in the GFP1-10-expressing live cells. Time-lapse imaging of green fluorescent protein (GFP)-labeled VLPs revealed that nuclear VLPs were translocated into the cytoplasm only after cell division, suggesting that the breakdown of the nuclear envelope during mitosis contributes to VLP nuclear export. Moreover, HiBiT activities of culture supernatants were dependent on the presence of a detergent, and the released VLPs were associated with extracellular vesicles, as observed under electron microscopy. Treatment with an antimitotic agent (nocodazole) enhanced the release of VLPs. These results suggest that the virions accumulated in the cytoplasm are constitutively released from the cell as membrane-coated vesicles. These properties are likely responsible for viral escape from host immune responses and enhance membrane fusion-mediated transmission. IMPORTANCE Parvovirus particles are expected to be applied as nanoparticles in drug delivery systems. However, little is known about how nuclear-assembled B19 virus (B19V) virions are released from host cells. This study provides evidence of mitosis-dependent nuclear export of B19V and extracellular vesicle-mediated virion release. Moreover, this study provides methods for modifying particle surfaces with various exogenous factors and contributes to the development of fine nanoparticles with novel valuable functions. The pB19-M20 plasmid expressing HiBiT-tagged VP2 is a novel tool to easily quantify VP2 expression. Furthermore, this system can be applied in high-throughput screening of reagents that affect VP2 expression, which might be associated with viral propagation.


Assuntos
Infecções por Parvoviridae , Parvovirus B19 Humano , Humanos , Linhagem Celular , Proteínas de Fluorescência Verde/metabolismo , Parvovirus B19 Humano/metabolismo , Peptídeos/metabolismo , Partículas Artificiais Semelhantes a Vírus
2.
J Virol ; 96(9): e0216421, 2022 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-35435730

RESUMO

Two new structures of the N-terminal domain of the main replication protein, NS1, of human parvovirus B19 (B19V) are presented here. This domain (NS1-nuc) plays an important role in the "rolling hairpin" replication of the single-stranded B19V DNA genome, recognizing origin of replication sequences in double-stranded DNA, and cleaving (i.e., nicking) single-stranded DNA at a nearby site known as the terminal resolution site (trs). The three-dimensional structure of NS1-nuc is well conserved between the two forms, as well as with a previously solved structure of a sequence variant of the same domain; however, it is shown here at a significantly higher resolution (2.4 Å). Using structures of NS1-nuc homologues bound to single- and double-stranded DNA, models for DNA recognition and nicking by B19V NS1-nuc are presented that predict residues important for DNA cleavage and for sequence-specific recognition at the viral origin of replication. IMPORTANCE The high-resolution structure of the DNA binding and cleavage domain of the main replicative protein, NS1, from the human-pathogenic virus human parvovirus B19 is presented here. Included also are predictions of how the protein recognizes important sequences in the viral DNA which are required for viral replication. These predictions can be used to further investigate the function of this protein, as well as to predict the effects on viral viability due to mutations in the viral protein and viral DNA sequences. Finally, the high-resolution structure facilitates structure-guided drug design efforts to develop antiviral compounds against this important human pathogen.


Assuntos
Modelos Moleculares , Parvovirus B19 Humano , Proteínas não Estruturais Virais , DNA Viral/genética , Endonucleases/química , Endonucleases/genética , Humanos , Parvovirus B19 Humano/genética , Parvovirus B19 Humano/metabolismo , Domínios Proteicos , Estrutura Terciária de Proteína , Proteínas não Estruturais Virais/química , Replicação Viral/genética
3.
Br J Haematol ; 193(4): 827-840, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33899219

RESUMO

A total of 244 patients with hereditary haemolytic anaemias (HHA) were screened for acute symptomatic human parvovirus B19 infection (HPV-B19) in a prospective study. To assess the risks associated with HPV-B19 infection, patients were classified into Group I and Group II according to presence or absence (symptoms, signs and specific serology) of acute HPV-B19 infection respectively. In all, 131 (53·7%) patients had ß-thalassaemia, 75 (30·7%) hereditary spherocytosis (HS), 27 (11·1%) sickle cell anaemia (SCA) and 11 (4·5%) glucose-6-phosphate dehydrogenase (G6PD) deficiency. Of 33 (13·5%) patients who presented with symptomatic HPV-B19 infection, 19 (57·5%) had HS, nine (27·3%) had ß-thalassaemia and five (15·2%) had SCA. In Group I, there were significant differences in the mean white blood cell, red blood cell and platelet counts, haemoglobin concentration, total bilirubin (TB), alanine aminotransferase, aspartate aminotransferase and serum creatinine (all P < 0·001) compared to Group II. In all, 27 (81·8%) patients had arthropathy and bone marrow failure (BMF); 13 (39·4%) had acute kidney injury (AKI), more in SCA (80%); and 12 (36·4%) patients had hepatitis, more in HS (66·8%). Five (15·2%) patients with HS had BMF, AKI, nervous system involvement and extreme hyperbilirubinaemia (TB range 26·3-84·7 mg/dl). Five (15·2%) patients had haemophagocytic syndrome. Two patients with HS combined with Type-I autoimmune hepatitis presented with transient BMF. Complete recovery or stabilisation was noted at 12 months in every patient except for one patient with SCA who died during the infection. HPV-B19 must be suspected and screened in patients with HHA with typical and atypical presentations with careful follow-up.


Assuntos
Anemia Hemolítica Congênita , Transtornos da Insuficiência da Medula Óssea , Eritema Infeccioso , Hepatite , Hiperbilirrubinemia , Parvovirus B19 Humano/metabolismo , Doença Aguda , Adolescente , Adulto , Anemia Hemolítica Congênita/sangue , Anemia Hemolítica Congênita/mortalidade , Anemia Hemolítica Congênita/virologia , Transtornos da Insuficiência da Medula Óssea/sangue , Transtornos da Insuficiência da Medula Óssea/mortalidade , Transtornos da Insuficiência da Medula Óssea/virologia , Criança , Eritema Infeccioso/sangue , Eritema Infeccioso/mortalidade , Feminino , Seguimentos , Hepatite/sangue , Hepatite/mortalidade , Hepatite/virologia , Humanos , Hiperbilirrubinemia/sangue , Hiperbilirrubinemia/mortalidade , Hiperbilirrubinemia/virologia , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos
4.
J Virol ; 93(1)2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30282717

RESUMO

Lytic infection of human parvovirus B19 (B19V) takes place exclusively in human erythroid progenitor cells of bone marrow and fetal liver, which disrupts erythropoiesis. During infection, B19V expresses three nonstructural proteins (NS1, 11-kDa, and 7.5-kDa) and two structural proteins (VP1 and VP2). While NS1 is essential for B19V DNA replication, 11-kDa enhances viral DNA replication significantly. In this study, we confirmed the enhancement role of 11-kDa in viral DNA replication and elucidated the underlying mechanism. We found that 11-kDa specially interacts with cellular growth factor receptor-bound protein 2 (Grb2) during virus infection and in vitro We determined a high affinity interaction between 11-kDa and Grb2 that has an equilibrium dissociation constant (KD ) value of 18.13 nM. In vitro, one proline-rich motif was sufficient for 11-kDa to sustain a strong interaction with Grb2. In consistence, in vivo during infection, one proline-rich motif was enough for 11-kDa to significantly reduce phosphorylation of extracellular signal-regulated kinase (ERK). Mutations of all three proline-rich motifs of 11-kDa abolished its capability to reduce ERK activity and, accordingly, decreased viral DNA replication. Transduction of a lentiviral vector encoding a short hairpin RNA (shRNA) targeting Grb2 decreased the expression of Grb2 as well as the level of ERK phosphorylation, which resulted in an increase of B19V replication. These results, in concert, indicate that the B19V 11-kDa protein interacts with cellular Grb2 to downregulate ERK activity, which upregulates viral DNA replication.IMPORTANCE Human parvovirus B19 (B19V) infection causes hematological disorders and is the leading cause of nonimmunological fetal hydrops during pregnancy. During infection, B19V expresses two structural proteins, VP1 and VP2, and three nonstructural proteins, NS1, 11-kDa, and 7.5-kDa. While NS1 is essential, 11-kDa plays an enhancing role in viral DNA replication. Here, we elucidated a mechanism underlying 11-kDa protein-regulated B19V DNA replication. 11-kDa is tightly associated with cellular growth factor receptor-bound protein 2 (Grb2) during infection. In vitro, 11-kDa interacts with Grb2 with high affinity through three proline-rich motifs, of which at least one is indispensable for the regulation of viral DNA replication. 11-kDa and Grb2 interaction disrupts extracellular signal-regulated kinase (ERK) signaling, which mediates upregulation of B19V replication. Thus, our study reveals a novel mechanism of how a parvoviral small nonstructural protein regulates viral DNA replication by interacting with a host protein that is predominately expressed in the cytoplasm.


Assuntos
Proteína Adaptadora GRB2/metabolismo , Infecções por Parvoviridae/metabolismo , Parvovirus B19 Humano/fisiologia , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/metabolismo , Motivos de Aminoácidos , Sítios de Ligação , Replicação do DNA , Humanos , Peso Molecular , Mutação , Parvovirus B19 Humano/metabolismo , Fosforilação , Prolina/metabolismo , Ligação Proteica , Replicação Viral
5.
J Pediatr Hematol Oncol ; 42(4): e213-e218, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31972722

RESUMO

OBJECTIVE: Blood-derived products from patient with hemophilia treated by factor VIII concentrates are potential sources of transfusion-transmitted infections, including human immunodeficiency virus, hepatitis, human pegivirus-1 (HPgV-1), B19 virus, and also human hepegivirus-1 (HHpgV-1). In the current study, we investigated the impact of blood transfusion on the prevalence of HHpgV-1, HPgV-1, and B19 virus in plasma of Iranian patient with hemophilia after direct-acting antiviral treatment of hepatitis C virus (HCV) infections for the first time. MATERIALS AND METHODS: A total of 170 patients with hemophilia who received direct-acting antivirals were enrolled in this study. Among them, 92 patients had a history of blood transfusion. The presence of HHpgV-1, HPgV-1, and B19 virus was detected by nested polymerase chain reaction analysis using the conserved primers. The plasmids harboring 5'-UTR and NS3 were used as positive controls for HPgV-1 and HHpgV-1, respectively. RESULTS: Our data identified 3 individuals with HHpgV-1 viremia (1.76%), 11 individuals with HPgV-1 viremia (6.47%), and 33 individuals with B19 viremia (19.4%). All patients were negative for hepatitis B virus, human immunodeficiency virus, and HCV infections. These findings indicated lower transmissibility or higher rates of virus clearance for HHpgV-1, HPgV-1, and B19 virus as compared with other bloodborne human flaviviruses such as HCV. However, the prevalence of B19 virus was significantly higher than the other 2 viruses. CONCLUSION: In general, these findings showed that the history of blood transfusion could increase the risk of viral transmission of bloodborne viruses among patient with hemophilia.


Assuntos
Transfusão de Sangue , DNA Viral/sangue , Eritema Infeccioso/sangue , Hemofilia A/sangue , Hepacivirus/metabolismo , Hepatite C/sangue , Parvovirus B19 Humano/metabolismo , Adolescente , Adulto , Criança , Pré-Escolar , Estudos Transversais , Eritema Infeccioso/epidemiologia , Eritema Infeccioso/etiologia , Feminino , Hemofilia A/epidemiologia , Hemofilia A/terapia , Hemofilia A/virologia , Hepatite C/epidemiologia , Hepatite C/terapia , Hepatite C/virologia , Humanos , Irã (Geográfico)/epidemiologia , Masculino , Pessoa de Meia-Idade , Prevalência
6.
Transfus Med ; 30(3): 226-230, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32131139

RESUMO

OBJECTIVE: In the multicentre Haemoglobinopathy Blood Surveillance Project, to evaluate the seroprevalence of parvovirus B19 and DNA viral load in sickle cell disease (SCD). BACKGROUND: Although the epidemiology of parvovirus B19 seropositivity in SCD has been well documented, there are few studies that have assessed possible persistent parvovirus DNAemia and associated risk factors including blood transfusion. METHODS: A qualitative analysis of parvovirus B19 serology using ELISA and quantitative parvovirus B19 DNA by RT-PCR was performed in patients with SCD. RESULTS: Of 322 patients, 113 (35%) were parvovirus IgG positive and 119 (37%) were IgM positive at enrolment. The prevalence of IgG positivity increased with age. 71/322 (22%) were parvovirus DNA positive at enrolment with a mean viral load of 15 227 ± 55 227 SD. (range 72-329 238 IU/mL). Patients who were positive for parvovirus B19 DNA received a significantly higher red blood cell transfusion volume in the prior year compared to patients who were negative (mean RBC volume = 8310 mL vs 5435 mL, respectively; P = .0073). Seventy-seven patients had follow-up testing approximately 1 year after enrolment and 11/28 (39%) patients had persistently positive IgM. CONCLUSION: Further studies are needed to better understand the natural history of parvovirus B19 infection in SCD especially in relation to RBC transfusion as a risk factor, as well as disease outcome and severity.


Assuntos
Anemia Falciforme , Anticorpos Antivirais/sangue , DNA Viral/sangue , Eritema Infeccioso , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Parvovirus B19 Humano/metabolismo , Adolescente , Adulto , Anemia Falciforme/sangue , Anemia Falciforme/epidemiologia , Anemia Falciforme/terapia , Anemia Falciforme/virologia , Centers for Disease Control and Prevention, U.S. , Criança , Pré-Escolar , Eritema Infeccioso/sangue , Eritema Infeccioso/epidemiologia , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Fatores de Risco , Estados Unidos
7.
J Virol ; 92(5)2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29237843

RESUMO

Human parvovirus B19 (B19V) infection of human erythroid progenitor cells (EPCs) induces a DNA damage response and cell cycle arrest at late S phase, which facilitates viral DNA replication. However, it is not clear exactly which cellular factors are employed by this single-stranded DNA virus. Here, we used microarrays to systematically analyze the dynamic transcriptome of EPCs infected with B19V. We found that DNA metabolism, DNA replication, DNA repair, DNA damage response, cell cycle, and cell cycle arrest pathways were significantly regulated after B19V infection. Confocal microscopy analyses revealed that most cellular DNA replication proteins were recruited to the centers of viral DNA replication, but not the DNA repair DNA polymerases. Our results suggest that DNA replication polymerase δ and polymerase α are responsible for B19V DNA replication by knocking down its expression in EPCs. We further showed that although RPA32 is essential for B19V DNA replication and the phosphorylated forms of RPA32 colocalized with the replicating viral genomes, RPA32 phosphorylation was not necessary for B19V DNA replication. Thus, this report provides evidence that B19V uses the cellular DNA replication machinery for viral DNA replication.IMPORTANCE Human parvovirus B19 (B19V) infection can cause transient aplastic crisis, persistent viremia, and pure red cell aplasia. In fetuses, B19V infection can result in nonimmune hydrops fetalis and fetal death. These clinical manifestations of B19V infection are a direct outcome of the death of human erythroid progenitors that host B19V replication. B19V infection induces a DNA damage response that is important for cell cycle arrest at late S phase. Here, we analyzed dynamic changes in cellular gene expression and found that DNA metabolic processes are tightly regulated during B19V infection. Although genes involved in cellular DNA replication were downregulated overall, the cellular DNA replication machinery was tightly associated with the replicating single-stranded DNA viral genome and played a critical role in viral DNA replication. In contrast, the DNA damage response-induced phosphorylated forms of RPA32 were dispensable for viral DNA replication.


Assuntos
Divisão Celular , Replicação do DNA , Interações Hospedeiro-Patógeno , Infecções por Parvoviridae/virologia , Parvovirus B19 Humano/genética , Parvovirus B19 Humano/metabolismo , Replicação Viral , Bromodesoxiuridina/metabolismo , Antígenos CD36/análise , Antígenos CD36/metabolismo , Ciclo Celular , Pontos de Checagem do Ciclo Celular , Linhagem Celular , Dano ao DNA , DNA Polimerase III , DNA Polimerase beta , Reparo do DNA , DNA de Cadeia Simples/metabolismo , DNA Viral/genética , DNA Viral/metabolismo , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/virologia , Morte Fetal , Regulação Viral da Expressão Gênica/fisiologia , Genoma Viral , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Parvovirus B19 Humano/patogenicidade , Fosforilação , Mapas de Interação de Proteínas , Aplasia Pura de Série Vermelha/virologia , Proteína de Replicação A/genética , Fase S , Transcriptoma , Viremia/virologia
8.
Indian J Med Res ; 148(4): 373-384, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30666000

RESUMO

Human parvovirus B19 (B19V) causes myriads of clinical diseases; however, owing to lack of awareness and undetermined clinical impact, it has failed to become a virus pathogen of global concern. Cryptically, B19V causes significant morbidity and mortality. Half of the world population and 60 per cent of Indians are known to be serologically naive and are at risk of acquiring B19V infections. Cumulatively, our data showed 21.3 per cent B19V-infected patients with juvenile chronic arthropathy, recurrent abortions, multi-transfused thalassaemia and leukaemia. In addition, B19V-infected cases that ended fatally included patients with pure red cell aplasia, fulminant hepatitis and haemophagocytic syndrome. Novel clinical associations of B19V observed were amegakaryocytic thrombocytopaenia, myositis and non-occlusive ischaemic gangrene of bowel. B19V possesses multiple receptors which are distributed widely in human tissues. Vascular endothelial cell infection by B19V causes endothelialitis and vasculitic injuries besides antibody-dependent enhancement which empowered B19V to cause multiorgan diseases. Owing to lack of suitable animal model for B19V, true causal role remains to be determined, but numerous reports on B19V infections substantiate a causal role in multiorgan diseases. Hence, B19V infections need to be recognized, investigated and treated besides making efforts on vaccine developments.


Assuntos
Infecções por Parvoviridae/complicações , Infecções por Parvoviridae/epidemiologia , Parvovirus B19 Humano/patogenicidade , Doenças Cardiovasculares/epidemiologia , Doenças Cardiovasculares/virologia , Feminino , Doenças Hematológicas/epidemiologia , Doenças Hematológicas/virologia , Humanos , Índia/epidemiologia , Nefropatias/epidemiologia , Nefropatias/virologia , Hepatopatias/epidemiologia , Hepatopatias/virologia , Doenças do Sistema Nervoso/epidemiologia , Doenças do Sistema Nervoso/virologia , Infecções por Parvoviridae/transmissão , Parvovirus B19 Humano/metabolismo , Gravidez , Complicações na Gravidez/epidemiologia , Estudos Soroepidemiológicos
9.
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
10.
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
11.
Biochem Biophys Res Commun ; 450(4): 1396-401, 2014 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-25010641

RESUMO

Parvovirus B19 (B19V) can cause inflammatory cardiomyopathy and endothelial dysfunction. Pathophysiological mechanisms involved include lysophosphatidylcholine producing phospholipase A2 (PLA2) activity of the B19V capsid protein VP1. Most recently, VP1 and lysophosphatidylcholine have been shown to inhibit Na(+)/K(+) ATPase. The present study explored whether VP1 modifies the activity of Kv1.3 and Kv1.5 K(+) channels. cRNA encoding Kv1.3 or Kv1.5 was injected into Xenopus oocytes without or with cRNA encoding VP1 isolated from a patient suffering from fatal B19V-induced myocarditis. K(+) channel activity was determined by dual electrode voltage clamp. Injection of cRNA encoding Kv1.3 or Kv1.5 into Xenopus oocytes was followed by appearance of Kv K(+) channel activity, which was significantly decreased by additional injection of cRNA encoding VP1, but not by additional injection of cRNA encoding PLA2-negative VP1 mutant (H153A). The effect of VP1 on Kv current was not significantly modified by transcription inhibitor actinomycin (10 µM for 36 h) but was mimicked by lysophosphatidylcholine (1 µg/ml). The B19V capsid protein VP1 inhibits host cell Kv channels, an effect at least partially due to phospholipase A2 (PLA) dependent formation of lysophosphatidylcholine.


Assuntos
Proteínas do Capsídeo/fisiologia , Regulação para Baixo , Parvovirus B19 Humano/metabolismo , Canais de Potássio/fisiologia , Animais , Humanos , Xenopus
12.
Cells ; 13(15)2024 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-39120285

RESUMO

Human parvovirus B19 (B19V), like most parvoviruses, possesses phospholipase A2 (PLA2) activity, which is thought to mediate endosomal escape by membrane disruption. Here, we challenge this model and find evidence for a mechanism of B19V entry mediated by the glycosphingolipid globoside without endosome disruption and retrograde transport to the Golgi. We show that B19V PLA2 activity requires specific calcium levels and pH conditions that are not optimal in endosomes. Accordingly, endosomal membrane integrity was maintained during B19V entry. Furthermore, endosomes remained intact when loaded with MS2 bacteriophage particles pseudotyped with multiple B19V PLA2 subunits, providing superior enzymatic potential compared to native B19V. In globoside knockout cells, incoming viruses are arrested in the endosomal compartment and the infection is blocked. Infection can be rescued by promoting endosomal leakage with polyethyleneimine (PEI), demonstrating the essential role of globoside in facilitating endosomal escape. Incoming virus colocalizes with Golgi markers and interfering with Golgi function blocks infection, suggesting that globoside-mediated entry involves the Golgi compartment, which provides conditions favorable for the lipolytic PLA2. Our study challenges the current model of B19V entry and identifies globoside as an essential intracellular receptor required for endosomal escape.


Assuntos
Endossomos , Globosídeos , Complexo de Golgi , Parvovirus B19 Humano , Internalização do Vírus , Endossomos/metabolismo , Endossomos/virologia , Humanos , Complexo de Golgi/metabolismo , Complexo de Golgi/virologia , Parvovirus B19 Humano/metabolismo , Parvovirus B19 Humano/fisiologia , Parvovirus B19 Humano/genética , Globosídeos/metabolismo , Fosfolipases A2/metabolismo , Cálcio/metabolismo
13.
Cell Physiol Biochem ; 31(4-5): 638-48, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23689698

RESUMO

BACKGROUND/AIMS: Human parvovirus B19 (B19V) may cause inflammatory cardiomyopathy (iCMP) which is accompanied by endothelial dysfunction. The B19V capsid protein VP1 contains a lysophosphatidylcholine producing phospholipase A2 (PLA) sequence. Lysophosphatidylcholine has in turn been shown to inhibit Na(+)/K(+) ATPase. The present study explored whether VP1 modifies Na(+)/K(+) ATPase activity. METHODS: Xenopus oocytes were injected with cRNA encoding VP1 isolated from a patient suffering from fatal B19V-iCMP or cRNA encoding PLA2-negative VP1 mutant (H153A) and K(+) induced pump current (I(pump)) as well as ouabain-inhibited current (I(ouabain)) both reflecting Na(+)/K(+)-ATPase activity were determined by dual electrode voltage clamp. RESULTS: Injection of cRNA encoding VP1, but not of VP1(H153A) or water, was followed by a significant decrease of both, I(pump) and I(ouabain) in Xenopus oocytes. The effect was not modified by inhibition of transcription with actinomycin (10 µM for 36 hours) but was abrogated in the presence of PLA2 specific blocker 4-bromophenacylbromide (50 µM) and was mimicked by lysophosphatidylcholine (0.5 - 1 µg/ml). According to whole cell patch clamp, lysophosphatidylcholine (1 µg /ml) similarly decreased I(pump) in human microvascular endothelial cells (HMEC). CONCLUSION: The B19V capsid protein VP1 is a powerful inhibitor of host cell Na(+)/K(+) ATPase, an effect at least partially due to phospholipase A2 (PLA2) dependent formation of lysophosphatidylcholine.


Assuntos
Proteínas do Capsídeo/metabolismo , Parvovirus B19 Humano/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Acetofenonas/farmacologia , Animais , Proteínas do Capsídeo/genética , Células Cultivadas , Regulação para Baixo , Células Endoteliais/citologia , Células Endoteliais/fisiologia , Humanos , Lisofosfatidilcolinas/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Oócitos/efeitos dos fármacos , Oócitos/fisiologia , Técnicas de Patch-Clamp , Fosfolipases A2/química , Fosfolipases A2/metabolismo , Plasmídeos/genética , Plasmídeos/metabolismo , ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores , Xenopus laevis/crescimento & desenvolvimento
14.
J Virol ; 86(19): 10748-58, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22837195

RESUMO

Human parvovirus B19 (B19V) infection is highly restricted to human erythroid progenitor cells, in which it induces a DNA damage response (DDR). The DDR signaling is mainly mediated by the ATR (ataxia telangiectasia-mutated and Rad3-related) pathway, which promotes replication of the viral genome; however, the exact mechanisms employed by B19V to take advantage of the DDR for virus replication remain unclear. In this study, we focused on the initiators of the DDR and the role of the DDR in cell cycle arrest during B19V infection. We examined the role of individual viral proteins, which were delivered by lentiviruses, in triggering a DDR in ex vivo-expanded primary human erythroid progenitor cells and the role of DNA replication of the B19V double-stranded DNA (dsDNA) genome in a human megakaryoblastoid cell line, UT7/Epo-S1 (S1). All the cells were cultured under hypoxic conditions. The results showed that none of the viral proteins induced phosphorylation of H2AX or replication protein A32 (RPA32), both hallmarks of a DDR. However, replication of the B19V dsDNA genome was capable of inducing the DDR. Moreover, the DDR per se did not arrest the cell cycle at the G(2)/M phase in cells with replicating B19V dsDNA genomes. Instead, the B19V nonstructural 1 (NS1) protein was the key factor in disrupting the cell cycle via a putative transactivation domain operating through a p53-independent pathway. Taken together, the results suggest that the replication of the B19V genome is largely responsible for triggering a DDR, which does not perturb cell cycle progression at G(2)/M significantly, during B19V infection.


Assuntos
Dano ao DNA , Replicação do DNA , Parvovirus B19 Humano/genética , Parvovirus B19 Humano/metabolismo , Replicação Viral , Antígenos CD34/biossíntese , Pontos de Checagem do Ciclo Celular , Divisão Celular , Fase G2 , Genoma Viral , Histonas/metabolismo , Humanos , Hipóxia , Lentivirus/genética , Mutação , Fosforilação , Regiões Promotoras Genéticas
15.
J Virol ; 86(9): 5099-109, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22357277

RESUMO

Despite its very narrow tropism for erythroid progenitor cells, human parvovirus B19 (B19V) has recently been shown to replicate and form infectious progeny virus in 293 cells in the presence of early adenoviral functions provided either by infection with adenovirus type 5 or by addition of the pHelper plasmid encoding the E2a, E4orf6, and VA RNA functions. In the present study we dissected the individual influence of these functions on B19V genome replication and expression of structural proteins VP1 and VP2. We show that, in the presence of the constitutively expressed E1A and E1B, E4orf6 alone is able to promote B19V DNA replication, resulting in a concomitant increase in VP expression levels. The stimulatory effects of E4orf6 require the integrity of the BC box motifs, which target cellular proteins such as p53 and the Mre11 DNA repair complex for proteosomal degradation through formation of an E3 ubiquitin ligase complex with E1B. VA RNA also strongly induces VP expression but, in contrast to E4orf6, in a replication-independent manner. This stimulation could be attributed exclusively to the VA I RNA transcript and does not involve major activating effects at the level of the B19V p6 promoter, but the nucleotide residues required for the well-defined pathway of VA I RNA mediated stimulation of translation through functional inactivation of protein kinase R. These data show that the cellular pathways regulating B19V replication may be very similar to those governing the productive cycle of the helper-dependent parvoviruses, the adeno-associated viruses.


Assuntos
Adenoviridae/metabolismo , Proteínas E4 de Adenovirus/metabolismo , Proteínas do Capsídeo/genética , Replicação do DNA , Regulação Viral da Expressão Gênica , Parvovirus B19 Humano/genética , RNA Viral/metabolismo , Adenoviridae/genética , Linhagem Celular , Proteínas Culina/metabolismo , DNA Viral/biossíntese , Humanos , Complexos Multiproteicos/metabolismo , Parvovirus B19 Humano/metabolismo , Parvovirus B19 Humano/fisiologia , Ligação Proteica , Ubiquitina-Proteína Ligases/metabolismo , eIF-2 Quinase/metabolismo
16.
Sci Adv ; 9(2): eade0869, 2023 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-36630517

RESUMO

Parvovirus B19 (B19V) infects human erythroid progenitor cells (EPCs) and causes several hematological disorders and fetal hydrops. Amino acid (aa) 5-68 of minor capsid protein VP1 (VP1u5-68aa) is the minimal receptor binding domain for B19V to enter EPCs. Here, we carried out a genome-wide CRISPR-Cas9 guide RNA screen and identified tyrosine protein kinase receptor UFO (AXL) as a proteinaceous receptor for B19V infection of EPCs. AXL gene silencing in ex vivo expanded EPCs remarkably decreased B19V internalization and replication. Additions of the recombinant AXL extracellular domain or a polyclonal antibody against it upon infection efficiently inhibited B19V infection of ex vivo expanded EPCs. Moreover, B19V VP1u interacted with the recombinant AXL extracellular domain in vitro at a relatively high affinity (KD = 103 nM). Collectively, we provide evidence that AXL is a co-receptor for B19V infection of EPCs.


Assuntos
Receptor Tirosina Quinase Axl , Eritema Infeccioso , Parvovirus B19 Humano , Humanos , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Eritema Infeccioso/metabolismo , Parvovirus B19 Humano/genética , Parvovirus B19 Humano/metabolismo , Ligação Proteica , Receptor Tirosina Quinase Axl/metabolismo
17.
J Biol Chem ; 286(28): 24793-805, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21622561

RESUMO

Alternative processing of parvovirus B19 (B19V) pre-mRNA is critical to generating appropriate levels of B19V mRNA transcripts encoding capsid proteins and small nonstructural proteins. Polyadenylation of the B19V pre-mRNA at the proximal polyadenylation site ((pA)p), which prevents generation of full-length capsid proteins encoding mRNA transcripts, has been suggested as a step that blocks B19V permissiveness. We report here that efficient splicing of the B19V pre-mRNA within the first intron (upstream of the (pA)p site) stimulated the polyadenylation; in contrast, splicing of the B19V pre-mRNA within the second intron (in which the (pA)p site resides) interfered with the polyadenylation, leading to the generation of a sufficient number of B19V mRNA transcripts polyadenylated at the distal polyadenylation site ((pA)d). We also found that splicing within the second intron and polyadenylation at the (pA)p site compete during processing of the B19V pre-mRNA. Furthermore, we discovered that the U1 RNA that binds to the 5' splice donor site of the second intron is fully responsible for inhibiting polyadenylation at the (pA)p site, whereas actual splicing, and perhaps assembly of the functional spliceosome, is not required. Finally, we demonstrated that inhibition of B19V pre-mRNA splicing within the second intron by targeting an intronic splicing enhancer using a Morpholino antisense oligonucleotide prevented B19V mRNA transcripts polyadenylated at the (pA)d site during B19V infection of human erythroid progenitors. Thus, our study reveals the mechanism by which alternative splicing coordinates alternative polyadenylation to generate full-length B19V mRNA transcripts at levels sufficient to support productive B19V infection.


Assuntos
Processamento Alternativo/fisiologia , Parvovirus B19 Humano/metabolismo , Sinais de Poliadenilação na Ponta 3' do RNA/fisiologia , Precursores de RNA/metabolismo , Processamento Pós-Transcricional do RNA/fisiologia , Sítios de Splice de RNA/fisiologia , RNA Viral/metabolismo , Animais , Células COS , Chlorocebus aethiops , Humanos , Íntrons/fisiologia , Morfolinos/metabolismo , Morfolinos/farmacologia , Parvovirus B19 Humano/genética , Precursores de RNA/genética , Processamento Pós-Transcricional do RNA/efeitos dos fármacos , RNA Viral/genética
18.
J Virol ; 85(16): 8046-55, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21680529

RESUMO

Human parvovirus B19 (B19V) infection is restricted to erythroid progenitor cells of the human bone marrow. Although the mechanism by which the B19V genome replicates in these cells has not been studied in great detail, accumulating evidence has implicated involvement of the cellular DNA damage machinery in this process. Here, we report that, in ex vivo-expanded human erythroid progenitor cells, B19V infection induces a broad range of DNA damage responses by triggering phosphorylation of all the upstream kinases of each of three repair pathways: ATM (ataxia-telangiectasi mutated), ATR (ATM and Rad3 related), and DNA-PKcs (DNA-dependent protein kinase catalytic subunit). We found that phosphorylated ATM, ATR, and DNA-PKcs, and also their downstream substrates and components (Chk2, Chk1, and Ku70/Ku80 complex, respectively), localized within the B19V replication center. Notably, inhibition of kinase phosphorylation (through treatment with either kinase-specific inhibitors or kinase-specific shRNAs) revealed requirements for signaling of ATR and DNA-PKcs, but not ATM, in virus replication. Inhibition of the ATR substrate Chk1 led to similar levels of decreased virus replication, indicating that signaling via the ATR-Chk1 pathway is critical to B19V replication. Notably, the cell cycle arrest characteristic of B19V infection was not rescued by interference with the activity of any of the three repair pathway kinases.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Células Precursoras Eritroides/virologia , Parvovirus B19 Humano/fisiologia , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Replicação Viral , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ligação ao Cálcio/metabolismo , Ciclo Celular , Linhagem Celular , Quinase 1 do Ponto de Checagem , Dano ao DNA , Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Citometria de Fluxo , Imunofluorescência , Histonas/metabolismo , Humanos , Hibridização in Situ Fluorescente , Parvovirus B19 Humano/genética , Parvovirus B19 Humano/metabolismo , Fosforilação , Proteína de Replicação A/metabolismo , Transdução de Sinais , Proteínas Supressoras de Tumor/metabolismo
19.
Blood ; 115(5): 1070-80, 2010 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-19861680

RESUMO

Human parvovirus B19 (B19V) infection shows a strong erythroid tropism and drastically destroys erythroid progenitor cells, thus leading to most of the disease outcomes associated with B19V infection. In this study, we systematically examined the 3 B19V nonstructural proteins, 7.5 kDa, 11 kDa, and NS1, for their function in inducing apoptosis in transfection of primary ex vivo-expanded erythroid progenitor cells, in comparison with apoptosis induced during B19V infection. Our results show that 11 kDa is a more significant inducer of apoptosis than NS1, whereas 7.5 kDa does not induce apoptosis. Furthermore, we determined that caspase-10, an initiator caspase in death receptor signaling, is the most active caspase in apoptotic erythroid progenitors induced by 11 kDa and NS1 as well as during B19V infection. More importantly, cytoplasm-localized 11 kDa is expressed at least 100 times more than nucleus-localized NS1 at the protein level in primary erythroid progenitor cells infected with B19V; and inhibition of 11 kDa expression using antisense oligos targeting specifically to the 11 kDa-encoding mRNAs reduces apoptosis significantly during B19V infection of erythroid progenitor cells. Taken together, these results demonstrate that the 11 kDa protein contributes to erythroid progenitor cell death during B19V infection.


Assuntos
Apoptose , Células Precursoras Eritroides/metabolismo , Parvovirus B19 Humano/genética , Proteínas não Estruturais Virais/genética , Clorometilcetonas de Aminoácidos/farmacologia , Caspase 10/metabolismo , Inibidores de Caspase , Linhagem Celular , Células Cultivadas , Inibidores de Cisteína Proteinase/farmacologia , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/virologia , Citometria de Fluxo , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Immunoblotting , Marcação In Situ das Extremidades Cortadas , Células K562 , Peso Molecular , Parvovirus B19 Humano/metabolismo , Parvovirus B19 Humano/fisiologia , Quinolinas/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/fisiologia
20.
Viruses ; 14(9)2022 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-36146728

RESUMO

Parvovirus B19 (B19V) is a human pathogen that is the causative agent of fifth disease in children. It is also known to cause hydrops in fetuses, anemia in AIDS patients, and transient aplastic crisis in patients with sickle cell disease. The unique N-terminus of Viral Protein 1 (VP1u) of parvoviruses, including B19V, exhibits phospholipase A2 (PLA2) activity, which is required for endosomal escape. Presented is the structural dynamics of B19V VP1u under conditions that mimic the pHs of cell entry and endosomal trafficking to the nucleus. Using circular dichroism spectroscopy, the receptor-binding domain of B19V VP1u is shown to exhibit an α-helical fold, whereas the PLA2 domain exhibits a probable molten globule state, both of which are pH invariant. Differential scanning calorimetry performed at endosomal pHs shows that the melting temperature (Tm) of VP1u PLA2 domain is tuned to body temperature (37 °C) at pH 7.4. In addition, PLA2 assays performed at temperatures ranging from 25-45 °C show both a temperature and pH-dependent change in activity. We hypothesize that VP1u PLA2 domain differences in Tm at differing pHs have enabled the virus to "switch on/off" the phospholipase activity during capsid trafficking. Furthermore, we propose the environment of the early endosome as the optimal condition for endosomal escape leading to B19V infection.


Assuntos
Parvovirus B19 Humano , Internalização do Vírus , Proteínas do Capsídeo/metabolismo , Criança , Endossomos/metabolismo , Humanos , Parvovirus B19 Humano/metabolismo , Fosfolipases A2/química , Proteínas Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA