Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 35
1.
Viruses ; 16(5)2024 05 13.
Article En | MEDLINE | ID: mdl-38793654

Based on several clinical observations it was hypothesized that herpesviruses may influence the replication of human bocaviruses, the second known parvoviruses that have been confirmed as human pathogens. While several cell lines support the growth of HSV-1, HBoV-1 was exclusively cultivated on air-liquid interface cultures, the latter being a rather complicated, slow, and low throughput system. One of the cell lines are T84 cells, which are derived from the lung metastasis of a colorectal tumor. In this study, we provide evidence that T84 also supports HBoV replication when cultivated as monolayers, while simultaneously being permissive for HSV-1. The cell culture model thus would enable co-infection studies of both viruses and is worth being optimized for high throughput studies with HBoV-1. Additionally, the study provides evidence for a supporting effect of HSV-1 on the replication and packaging of HBoV-1 progeny DNA into DNase-resistant viral particles.


Coinfection , Herpesvirus 1, Human , Human bocavirus , Virus Replication , Herpesvirus 1, Human/physiology , Humans , Coinfection/virology , Human bocavirus/physiology , Human bocavirus/genetics , Cell Line , Cell Line, Tumor , Cell Culture Techniques/methods , Herpes Simplex/virology , Parvoviridae Infections/virology , Chlorocebus aethiops , Virus Cultivation/methods
2.
J Virol ; 96(4): e0184021, 2022 02 23.
Article En | MEDLINE | ID: mdl-34878919

Human bocavirus 1 (HBoV1), an autonomous human parvovirus, causes acute respiratory tract infections in young children. HBoV1 infects well-differentiated (polarized) human airway epithelium cultured at an air-liquid interface (HAE-ALI). HBoV1 expresses a large nonstructural protein, NS1, that is essential for viral DNA replication. HBoV1 infection of polarized human airway epithelial cells induces a DNA damage response (DDR) that is critical to viral DNA replication involving DNA repair with error-free Y-family DNA polymerases. HBoV1 NS1 or the isoform NS1-70 per se induces a DDR. In this study, using the second-generation proximity-dependent biotin identification (BioID2) approach, we identified that Ku70 is associated with the NS1-BioID2 pulldown complex through a direct interaction with NS1. Biolayer interferometry (BLI) assay determined a high binding affinity of NS1 with Ku70, which has an equilibrium dissociation constant (KD) value of 0.16 µM and processes the strongest interaction at the C-terminal domain. The association of Ku70 with NS1 was also revealed during HBoV1 infection of HAE-ALI. Knockdown of Ku70 and overexpression of the C-terminal domain of Ku70 significantly decreased HBoV1 replication in HAE-ALI. Thus, our study provides, for the first time, a direct interaction of parvovirus large nonstructural protein NS1 with Ku70. IMPORTANCE Parvovirus infection induces a DNA damage response (DDR) that plays a pivotal role in viral DNA replication. The DDR includes activation of ATM (ataxia telangiectasia mutated), ATR (ATM- and RAD3-related), and DNA-PKcs (DNA-dependent protein kinase catalytic subunit). The large nonstructural protein (NS1) often plays a role in the induction of DDR; however, how the DDR is induced during parvovirus infection or simply by the NS1 is not well studied. Activation of DNA-PKcs has been shown as one of the key DDR pathways in DNA replication of HBoV1. We identified that HBoV1 NS1 directly interacts with Ku70, but not Ku80, of the Ku70/Ku80 heterodimer at high affinity. This interaction is also important for HBoV1 replication in HAE-ALI. We propose that the interaction of NS1 with Ku70 recruits the Ku70/Ku80 complex to the viral DNA replication center, which activates DNA-PKcs and facilitates viral DNA replication.


Human bocavirus/physiology , Ku Autoantigen/metabolism , Respiratory Mucosa/virology , Viral Nonstructural Proteins/metabolism , Virus Replication , DNA Damage , DNA Replication , DNA, Viral/biosynthesis , Genome, Viral , HEK293 Cells , Human bocavirus/metabolism , Humans , Ku Autoantigen/genetics , Protein Binding , Protein Domains , Respiratory Mucosa/metabolism , Viral Nonstructural Proteins/genetics , Viral Replication Compartments/metabolism
3.
PLoS One ; 16(12): e0259443, 2021.
Article En | MEDLINE | ID: mdl-34919553

The present study was done to identify the viral diversity, seasonality and burden associated with childhood acute respiratory tract infection (ARTI) in Sri Lanka. Nasopharyngeal aspirates (NPA) of hospitalized children (1 month-5 years) with ARTI were collected in 2 centers (wet and dry zones) from March 2013 to August 2014. Respiratory viral antigen detection by immunofluorescence assay (IFA) was used to identify the infecting viruses. IFA negative 100 NPA samples were tested for human metapeumovirus (hMPV), human bocavirus and corona viruses by polymerase chain reaction. Of the 443 and 418 NPAs, 37.2% and 39.4% were positive for any of the 8 different respiratory viruses tested from two centers studied. Viral co-infection was detected with respiratory syncytial virus (RSV) in both centers. Peak viral detection was noted in the wet zone from May-July 2013 and 2014 and in the dry zone from December-January 2014 suggesting a local seasonality for viral ARTI. RSV showed a clear seasonality with a direct correlation of monthly RSV infections with rainy days in the wet zone and an inverse correlation with temperature in both centers. The case fatality rate was 2.7% for RSV associated ARTI. The overall disability adjusted life years was 335.9 and for RSV associated ARTI it was 241.8. RSV was the commonly detected respiratory virus with an annual seasonality and distribution in rainy seasons in the dry and wet zones of Sri Lanka. Identifying the virus and seasonality will contribute to employ preventive measures and reduce the empirical use of antibiotics in resource limited settings.


Coronavirus Infections/epidemiology , Paramyxoviridae Infections/epidemiology , Parvoviridae Infections/epidemiology , Respiratory Syncytial Virus Infections/epidemiology , Respiratory Tract Infections/epidemiology , Viral Load , Child, Hospitalized , Child, Preschool , Coinfection , Coronavirus/pathogenicity , Coronavirus/physiology , Coronavirus Infections/mortality , Coronavirus Infections/virology , Disability-Adjusted Life Years/trends , Female , Human bocavirus/pathogenicity , Human bocavirus/physiology , Humans , Incidence , Infant , Male , Metapneumovirus/pathogenicity , Metapneumovirus/physiology , Paramyxoviridae Infections/mortality , Paramyxoviridae Infections/virology , Parvoviridae Infections/mortality , Parvoviridae Infections/virology , Respiratory Syncytial Virus Infections/mortality , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/pathogenicity , Respiratory Syncytial Virus, Human/physiology , Respiratory Tract Infections/mortality , Respiratory Tract Infections/virology , Seasons , Sri Lanka/epidemiology , Survival Analysis
4.
J Virol ; 94(11)2020 05 18.
Article En | MEDLINE | ID: mdl-32213611

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.


Capsid Proteins , Capsid/metabolism , DNA, Viral , Human bocavirus/physiology , Parvoviridae Infections , Virus Assembly , Capsid Proteins/genetics , Capsid Proteins/metabolism , DNA, Viral/genetics , DNA, Viral/metabolism , Genetic Therapy , Genetic Vectors , HEK293 Cells , Humans , Parvoviridae Infections/genetics , Parvoviridae Infections/metabolism , Parvoviridae Infections/pathology , Transduction, Genetic
5.
Emerg Microbes Infect ; 8(1): 1563-1573, 2019.
Article En | MEDLINE | ID: mdl-31672101

The episomal structures of all human bocavirus (HBoV) genotypes have been deciphered, including the circular genome of HBoV2 (HBoV2-C1). To discern the role of the circular HBoV2 genome, three distinct linearized HBoV2-C1 genomes were cloned into pBlueScript SKII(+) to obtain pBlueScript HBoV2 5043-5042 (retaining all secondary structures), pBlueScript-HBoV2 5075-5074 (retaining hairpin number 2 and the 5' terminal structure), and pBlueScript-HBoV2 5220-5219 (retaining only the 5' terminal structure at the 5' -genome end). The recombinant plasmids were separately transfected HEK293 cells, revealing that more HBoV2 DNA had accumulated in the pBlueScript HBoV2 5043-5042-transfected HEK293 cells at 72 h post-transfection, as determined by real-time PCR. However, more mRNA was transcribed by pBlueScript-HBoV2 5075-5074 than by the other constructs, as determined by dot-blot hybridization and RNAscope. No significant differences in NS1-70 protein expression were observed among the three HBoV2 genomic clones. However, electron microscopy showed that HBoV2 virus particles were only present in the pBlueScript HBoV2 5043-5042-transfected HEK293 cells. By using three hetero-recombinant HBoV2 genomic clones in HEK293 transfected cells, only the genome with intact secondary structures produced virus particles, suggesting that retaining these structures in a circular genome is important for HBoV2 DNA replication and virus assembly.


DNA, Circular/genetics , DNA, Viral/genetics , Human bocavirus/genetics , Parvoviridae Infections/virology , RNA, Untranslated/genetics , Recombination, Genetic , Virus Assembly , DNA Replication , DNA, Circular/chemistry , DNA, Circular/metabolism , DNA, Viral/chemistry , DNA, Viral/metabolism , Genome, Viral , Genomics , Genotype , HEK293 Cells , Human bocavirus/chemistry , Human bocavirus/physiology , Humans , Nucleic Acid Conformation , Phylogeny , RNA, Untranslated/chemistry , RNA, Untranslated/metabolism
6.
Clin Lab ; 65(1)2019 Jan 01.
Article En | MEDLINE | ID: mdl-30775887

BACKGROUND: Human bocavirus (HBoV) is known to cause lower respiratory tract infections (LRTI) in children and may result in substantial morbidity and mortality. The aim of this study was to determine HBoV prevalence among hospitalized infants and small children with acute LRTI in Zagreb, Croatia, as well as to evaluate HBoV DNA quantity in samples in relation to the patients' age and co-infection with other respiratory viruses. METHODS: During winter season 2016/2017, a total of 295 children younger than three years of age who were admitted to hospitals with LRTI were tested for the presence of HBoV, respiratory syncytial virus (RSV), adenovirus (ADV), parainfluenza virus (PIV) types 1 to 3, and human metapneumovirus (HMPV). HBoV was detected with a real-time PCR method, and the other viruses were diagnosed using monoclonal antibodies in direct fluorescence assay. RESULTS: Viral etiology was proven in 225/295 (76.3%) of patients. The most commonly diagnosed virus was RSV (59.3%), followed by HBoV (23.1%), PIVs (4.4%), ADV (3.1%), and HMPV (1.4%). HBoV-infected children were older than RSV-infected children; likewise, detection rates of HBoV infection increased with age, while RSV infection rates decreased with age. In 51% of HBoV-positive samples an additional respiratory virus was also detected. There was no difference in HBoV DNA quantity between samples with single virus detection and those with multiple virus detection (p = 0.056), although samples positive only for HBoV showed higher cycle threshold values. There was no difference in HBoV DNA quantity in samples of different age groups (p > 0.05). CONCLUSIONS: Frequent detection of HBoV in small children with LRTI, even in combination with other viruses, highlights its role in the pathogenesis of respiratory disease.


Coinfection/virology , Human bocavirus/physiology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/physiology , Respiratory Tract Infections/virology , Child , Child, Preschool , Coinfection/diagnosis , Coinfection/epidemiology , Croatia/epidemiology , Female , Hospitalization/statistics & numerical data , Humans , Infant , Male , Prevalence , Respiratory Syncytial Virus Infections/epidemiology , Respiratory Tract Infections/diagnosis , Respiratory Tract Infections/epidemiology , Seasons
8.
Arch Virol ; 163(5): 1325-1330, 2018 May.
Article En | MEDLINE | ID: mdl-29392492

The aims of this study were to investigate the human bocavirus (HBoV) frequency and genotypes in hospitalized children <5 years presenting acute respiratory infections (ARI) within the São Paulo metropolitan area. Nasopharyngeal samples from 300 patients, previously screened for common respiratory viruses, were tested by qPCR for the NSP1 and NP-1 genes. The VP1/2 gene in positive samples was then amplified by PCR and sequenced. A total of 49 positive HBoV cases (16.3%; mean Ct value of 34.41) were detected with the mean age being 18.1 months (range 1 month to 5 years) and the median age being 1 year of age. Children aged between 0 and 12 months had higher detection rates of HBoV (69.4%; 34/49; mean Ct = 34.45) than children from other age groups (30.6%; 15/49; mean Ct = 34.34). No significant differences were observed between HBoV Ct levels and clinical illness. The occurrence was more frequently associated with fall (38.8%; 19/49) and spring (36.7%; 18/49). All 12 sequenced isolates were identified as HBoV-1, displaying minor genetic variation compared to the Swedish reference strains ST1 and ST2 (99.1-99.7% nt). The sole identification of HBoV-1 supports the hypothesis that this particular genotype is strongly related to ARI, and contributes to the role of this virus in the aetiology of respiratory diseases.


Human bocavirus/genetics , Human bocavirus/isolation & purification , Parvoviridae Infections/epidemiology , Parvoviridae Infections/virology , Respiratory Tract Infections/virology , Acute Disease/epidemiology , Brazil/epidemiology , Child , Child, Preschool , DNA, Viral/genetics , Epidemiological Monitoring , Female , Genetic Variation , Genotype , Human bocavirus/physiology , Humans , Infant , Male , Nasopharynx/virology , Phylogeny , Real-Time Polymerase Chain Reaction , Viral Proteins/genetics
9.
Arch Virol ; 163(4): 845-853, 2018 Apr.
Article En | MEDLINE | ID: mdl-29327237

Despite increased understanding of how viral infection is involved in asthma exacerbations, it is less clear which viruses are involved and to what extent they contribute to asthma exacerbations. Here, we sought to determine the prevalence of different respiratory viruses during asthma exacerbations. Systematic computerized searches of the literature up to June 2017 without language limitation were performed. The primary focus was on the prevalence of respiratory viruses, including AdV (adenovirus), BoV (bocavirus), CoV (coronavirus), CMV (cytomegalovirus), EnV (enterovirus), HSV (herpes simplex virus), IfV (influenza virus), MpV (metapneumovirus), PiV (parainfluenzavirus), RV (rhinovirus) and RSV (respiratory syncytial virus) during asthma exacerbations. We also examined the prevalence of viral infection stratified by age, geographic region, type of respiratory secretion, and detection method. Sixty articles were included in the final analysis. During asthma exacerbations, the mean prevalence of AdV, BoV, CoV, CMV, EnV, HSV, IfV, MpV, PiV, RV and RSV was 3.8%, 6.9%, 8.4%, 7.2%, 10.1%, 12.3%, 10.0%, 5.3%, 5.6%, 42.1% and 13.6%, respectively. EnV, MPV, RV and RSV were more prevalent in children, whereas AdV, BoV, CoV, IfV and PiV were more frequently present in adults. RV was the major virus detected globally, except in Africa. RV could be detected in both the upper and lower airway. Polymerase chain reaction was the most sensitive method for detecting viral infection. Our findings indicate the need to develop prophylactic polyvalent or polyvirus (including RV, EnV, IfV and RSV) vaccines that produce herd immunity and reduce the healthcare burden associated with virus-induced asthma exacerbations.


Asthma/epidemiology , Respiratory System/virology , Respiratory Tract Infections/epidemiology , Virus Diseases/epidemiology , Adenoviridae/pathogenicity , Adenoviridae/physiology , Africa/epidemiology , Age Factors , Americas/epidemiology , Asia/epidemiology , Asthma/complications , Asthma/virology , Coronavirus/pathogenicity , Coronavirus/physiology , Cytomegalovirus/pathogenicity , Cytomegalovirus/physiology , Enterovirus/pathogenicity , Enterovirus/physiology , Europe/epidemiology , Human bocavirus/pathogenicity , Human bocavirus/physiology , Humans , Metapneumovirus/pathogenicity , Metapneumovirus/physiology , Prevalence , Respiratory Syncytial Virus, Human/pathogenicity , Respiratory Syncytial Virus, Human/physiology , Respiratory Tract Infections/complications , Respiratory Tract Infections/virology , Respirovirus/pathogenicity , Respirovirus/physiology , Rhinovirus/pathogenicity , Rhinovirus/physiology , Simplexvirus/pathogenicity , Simplexvirus/physiology , Virus Diseases/complications , Virus Diseases/virology
10.
J Virol ; 91(24)2017 12 15.
Article En | MEDLINE | ID: mdl-29021400

Human bocavirus 1 (HBoV1) is a human parvovirus that causes acute respiratory tract infections in young children. In this study, we confirmed that, when polarized/well-differentiated human airway epithelia are infected with HBoV1 in vitro, they develop damage characterized by barrier function disruption and cell hypotrophy. Cell death mechanism analyses indicated that the infection induced pyroptotic cell death characterized by caspase-1 activation. Unlike infections with other parvoviruses, HBoV1 infection did not activate the apoptotic or necroptotic cell death pathway. When the NLRP3-ASC-caspase-1 inflammasome-induced pathway was inhibited by short hairpin RNA (shRNA), HBoV1-induced cell death dropped significantly; thus, NLRP3 mediated by ASC appears to be the pattern recognition receptor driving HBoV1 infection-induced pyroptosis. HBoV1 infection induced steady increases in the expression of interleukin 1α (IL-1α) and IL-18. HBoV1 infection was also associated with the marked expression of the antiapoptotic genes BIRC5 and IFI6 When the expression of BIRC5 and/or IFI6 was inhibited by shRNA, the infected cells underwent apoptosis rather than pyroptosis, as indicated by increased cleaved caspase-3 levels and the absence of caspase-1. BIRC5 and/or IFI6 gene inhibition also significantly reduced HBoV1 replication. Thus, HBoV1 infection of human airway epithelial cells activates antiapoptotic proteins that suppress apoptosis and promote pyroptosis. This response may have evolved to confer a replicative advantage, thus allowing HBoV1 to establish a persistent airway epithelial infection. This is the first report of pyroptosis in airway epithelia infected by a respiratory virus.IMPORTANCE Microbial infection of immune cells often induces pyroptosis, which is mediated by a cytosolic protein complex called the inflammasome that senses microbial pathogens and then activates the proinflammatory cytokines IL-1 and IL-18. While virus-infected airway epithelia often activate NLRP3 inflammasomes, studies to date suggest that these viruses kill the airway epithelial cells via the apoptotic or necrotic pathway; involvement of the pyroptosis pathway has not been reported previously. Here, we show for the first time that virus infection of human airway epithelia can also induce pyroptosis. Human bocavirus 1 (HBoV1), a human parvovirus, causes lower respiratory tract infections in young children. This study indicates that HBoV1 kills airway epithelial cells by activating genes that suppress apoptosis and thereby promote pyroptosis. This strategy appears to promote HBoV1 replication and may have evolved to allow HBoV1 to establish persistent infection of human airway epithelia.


Apoptosis , Epithelial Cells/pathology , Human bocavirus/physiology , Pyroptosis , Respiratory Mucosa/pathology , Respiratory Mucosa/virology , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Caspase 1/deficiency , Caspase 1/genetics , Caspase 3/genetics , Caspase 3/metabolism , DNA Replication , Epithelial Cells/virology , Humans , Inflammasomes , Inhibitor of Apoptosis Proteins/deficiency , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Interleukin-18/genetics , Interleukin-1alpha/genetics , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/genetics , RNA, Small Interfering/genetics , Survivin , Virus Replication
11.
Virology ; 510: 273-280, 2017 10.
Article En | MEDLINE | ID: mdl-28777951

Human bocavirus 1 (HBoV1) is a parvovirus associated with pneumonia in infants. It has been detected in different tissues, including colorectal tumors. In this study, we investigated whether Caco-2 cell line, derived from human colon cancer, can be utilized as a model for HBoV1 replication. We demonstrate HBoV1 replication in Caco-2 cultures supplemented with DEAE-dextran after inoculation with respiratory material from infected patients presenting with acute respiratory infection. A viral cycle of rapid development is displayed. However, in spite of HBoV1 DNA 4-fold increment in the supernatants and monolayers by day 1, evidencing that the system allows the virus genome replication after the entry occurred, infectious progeny particles were not produced. These results are consistent with an infection that is limited to a single growth cycle, which can be associated to mutations in the NS1 and VP1/VP2 regions of HBoV1 genome. Further research will contribute to fully elucidate these observations.


Epithelial Cells/virology , Human bocavirus/physiology , Virus Cultivation , Virus Replication , Caco-2 Cells , DNA, Viral/analysis , Humans
12.
Sci Rep ; 7(1): 7407, 2017 08 07.
Article En | MEDLINE | ID: mdl-28785044

Human bocavirus 1 (HBoV1) is an autonomous parvovirus in the Bocaparvovirus genus. The multifunctional nuclear protein NP1 is involved in viral replication. In the present study, we found that the mutations in the C-terminus of NS1 affected NP1 function in viral replication. Knocking out NP1 expression in the recombinant infectious clone, on which the C-terminus of NS1 was mutated based on the clinical samples from nasopharyngeal aspirates, resulted in different degrees of decreased replication. The result suggested that NP1 facilitated the replication of viral genome but was not necessary, which is different from the minute virus of canines, where NP1 is essential for viral replication. Further studies showed that clinical mutations in the NP1 region did not affect viral genome replication, and UP1 promoted viral DNA replication. Our results suggested that the C-terminus of NS1 is important for viral replication and may be a target for regulating the replication of the viral genome.


Human bocavirus/physiology , Mutant Proteins/metabolism , Point Mutation , Viral Nonstructural Proteins/metabolism , Virus Replication , China , Gene Knockout Techniques , HEK293 Cells , Human bocavirus/genetics , Human bocavirus/isolation & purification , Humans , Mutant Proteins/genetics , Mutation, Missense , Parvoviridae Infections/virology , Viral Nonstructural Proteins/genetics
13.
J Virol ; 91(8)2017 04 15.
Article En | MEDLINE | ID: mdl-28122984

Human bocavirus 1 (HBoV1) belongs to the species Primate bocaparvovirus of the genus Bocaparvovirus of the Parvoviridae family. HBoV1 causes acute respiratory tract infections in young children and has a selective tropism for the apical surface of well-differentiated human airway epithelia (HAE). In this study, we identified an additional HBoV1 gene, bocavirus-transcribed small noncoding RNA (BocaSR), within the 3' noncoding region (nucleotides [nt] 5199 to 5338) of the viral genome of positive sense. BocaSR is transcribed by RNA polymerase III (Pol III) from an intragenic promoter at levels similar to that of the capsid protein-coding mRNA and is essential for replication of the viral DNA in both transfected HEK293 and infected HAE cells. Mechanistically, we showed that BocaSR regulates the expression of HBoV1-encoded nonstructural proteins NS1, NS2, NS3, and NP1 but not NS4. BocaSR is similar to the adenovirus-associated type I (VAI) RNA in terms of both nucleotide sequence and secondary structure but differs from it in that its regulation of viral protein expression is independent of RNA-activated protein kinase (PKR) regulation. Notably, BocaSR accumulates in the viral DNA replication centers within the nucleus and likely plays a direct role in replication of the viral DNA. Our findings reveal BocaSR to be a novel viral noncoding RNA that coordinates the expression of viral proteins and regulates replication of viral DNA within the nucleus. Thus, BocaSR may be a target for antiviral therapies for HBoV and may also have utility in the production of recombinant HBoV vectors.IMPORTANCE Human bocavirus 1 (HBoV1) is pathogenic to humans, causing acute respiratory tract infections in young children. In this study, we identified a novel HBoV1 gene that lies in the 3' noncoding region of the viral positive-sense genome and is transcribed by RNA polymerase III into a noncoding RNA of 140 nt. This bocavirus-transcribed small RNA (BocaSR) diverges from both adenovirus-associated (VA) RNAs and Epstein-Barr virus-encoded small RNAs (EBERs) with respect to RNA sequence, representing a third species of this kind of Pol III-dependent viral noncoding RNA and the first noncoding RNA identified in autonomous parvoviruses. Unlike the VA RNAs, BocaSR localizes to the viral DNA replication centers of the nucleus and is essential for expression of viral nonstructural proteins independent of RNA-activated protein kinase R and replication of HBoV1 genomes. The identification of BocaSR and its role in virus DNA replication reveals potential avenues for developing antiviral therapies.


Gene Expression Regulation, Viral , Human bocavirus/physiology , RNA, Small Untranslated/metabolism , Virus Replication , Cells, Cultured , DNA Replication , Humans
14.
J Virol ; 91(3)2017 Feb 01.
Article En | MEDLINE | ID: mdl-27881651

Alternative processing of human bocavirus (HBoV) P5 promoter-transcribed RNA is critical for generating the structural and nonstructural protein-encoding mRNA transcripts. The regulatory mechanism by which HBoV RNA transcripts are polyadenylated at proximal [(pA)p] or distal [(pA)d] polyadenylation sites is still unclear. We constructed a recombinant HBoV infectious clone to study the alternative polyadenylation regulation of HBoV. Surprisingly, in addition to the reported distal polyadenylation site, (pA)d, a novel distal polyadenylation site, (pA)d2, which is located in the right-end hairpin (REH), was identified during infectious clone transfection or recombinant virus infection. (pA)d2 does not contain typical hexanucleotide polyadenylation signal, upstream elements (USE), or downstream elements (DSE) according to sequence analysis. Further study showed that HBoV nonstructural protein NS1, REH, and cis elements of (pA)d were necessary and sufficient for efficient polyadenylation at (pA)d2. The distance and sequences between (pA)d and (pA)d2 also played a key role in the regulation of polyadenylation at (pA)d2. Finally, we demonstrated that efficient polyadenylation at (pA)d2 resulted in increased HBoV capsid mRNA transcripts and protein translation. Thus, our study revealed that all the bocaviruses have distal poly(A) signals on the right-end palindromic terminus, and alternative polyadenylation at the HBoV 3' end regulates its capsid expression. IMPORTANCE: The distal polyadenylation site, (pA)d, of HBoV is located about 400 nucleotides (nt) from the right-end palindromic terminus, which is different from those of bovine parvovirus (BPV) and canine minute virus (MVC) in the same genus whose distal polyadenylation is located in the right-end stem-loop structure. A novel polyadenylation site, (pA)d2, was identified in the right-end hairpin of HBoV during infectious clone transfection or recombinant virus infection. Sequence analysis showed that (pA)d2 does not contain typical polyadenylation signals, and the last 42 nt form a stem-loop structure which is almost identical to that of MVC. Further study showed that NS1, REH, and cis elements of (pA)d are required for efficient polyadenylation at (pA)d2. Polyadenylation at (pA)d2 enhances capsid expression. Our study demonstrates alternative polyadenylation at the 3' end of HBoV and suggests an additional mechanism by which capsid expression is regulated.


Capsid Proteins/genetics , Gene Expression Regulation, Viral , Human bocavirus/physiology , Polyadenylation , Transcription, Genetic , Alternative Splicing , Base Sequence , Cell Line , Humans , Mutation , Poly A , RNA, Messenger/genetics , RNA, Messenger/metabolism , Regulatory Sequences, Nucleic Acid , Terminal Repeat Sequences
15.
Allergol. immunopatol ; 44(5): 410-414, sept.-oct. 2016. tab
Article En | IBECS | ID: ibc-155851

BACKGROUND: Human bocavirus (HBoV) was recently discovered and identified as an important cause of respiratory infection in young children. However, the relationship between HBoV-bronchiolitis and the development of recurrent wheezing has not yet been established. OBJECTIVE: We designed this study in order to describe the mid-term outcome, regarding the development of recurrent wheezing and asthma of HBoV-bronchiolitis patients and to compare it with RSV-bronchiolitis infants. METHODS: We studied 80 children (10 with HBoV and 70 with RSV infection), currently aged ≥4 years and previously hospitalised during the seasons 2004-2009 due to acute bronchiolitis. Epidemiological and clinical data were collected through structured clinical interviews at the follow-up visit. Spirometry and skin prick tests to common food and inhaled allergens were performed. RESULTS: All HBoV-patients developed recurrent wheezing and half of them had asthma at age 5-7 years. Almost 30% required hospital admission for recurrent wheezing. Asthma (odds ratio (OR)=1.28) and current asthma (OR=2.18) were significantly more frequent in children with HBoV-bronchiolitis than in RSV-bronchiolitis. FEV1 values were 99.2±4.8 in HBoV-group vs.103±11 in RSV-group, p: 0.09. No differences were found with respect to allergic rhinitis, atopic dermatitis, food allergy, proportion of positive prick tests, and family history of atopy or asthma. CONCLUSIONS: Severe HBoV-bronchiolitis in infancy was strongly associated with asthma at 5-7 years


No disponible


Humans , Male , Female , Respiratory Sounds/diagnosis , Respiratory Sounds/immunology , Recurrence , Asthma/complications , Asthma/immunology , Human bocavirus/immunology , Human bocavirus/physiology , Bronchiolitis/complications , Bronchiolitis/diagnosis , Bronchiolitis/immunology , Spirometry/methods , Skin Tests/methods , Skin Tests , Dermatitis, Atopic/complications , Dermatitis, Atopic/immunology , Hypersensitivity, Immediate/immunology
16.
J Virol ; 90(17): 7761-77, 2016 09 01.
Article En | MEDLINE | ID: mdl-27334591

UNLABELLED: Parvoviruses are single-stranded DNA viruses that use the palindromic structures at the ends of the viral genome for their replication. The mechanism of parvovirus replication has been studied mostly in the dependoparvovirus adeno-associated virus 2 (AAV2) and the protoparvovirus minute virus of mice (MVM). Here, we used human bocavirus 1 (HBoV1) to understand the replication mechanism of bocaparvovirus. HBoV1 is pathogenic to humans, causing acute respiratory tract infections, especially in young children under 2 years old. By using the duplex replicative form of the HBoV1 genome in human embryonic kidney 293 (HEK293) cells, we identified the HBoV1 minimal replication origin at the right-end hairpin (OriR). Mutagenesis analyses confirmed the putative NS1 binding and nicking sites within the OriR. Of note, unlike the large nonstructural protein (Rep78/68 or NS1) of other parvoviruses, HBoV1 NS1 did not specifically bind OriR in vitro, indicating that other viral and cellular components or the oligomerization of NS1 is required for NS1 binding to the OriR. In vivo studies demonstrated that residues responsible for NS1 binding and nicking are within the origin-binding domain. Further analysis identified that the small nonstructural protein NP1 is required for HBoV1 DNA replication at OriR. NP1 and other viral nonstructural proteins (NS1 to NS4) colocalized within the viral DNA replication centers in both OriR-transfected cells and virus-infected cells, highlighting a direct involvement of NP1 in viral DNA replication at OriR. Overall, our study revealed the characteristics of HBoV1 DNA replication at OriR, suggesting novel characteristics of autonomous parvovirus DNA replication. IMPORTANCE: Human bocavirus 1 (HBoV1) causes acute respiratory tract infections in young children. The duplex HBoV1 genome replicates in HEK293 cells and produces progeny virions that are infectious in well-differentiated airway epithelial cells. A recombinant AAV2 vector pseudotyped with an HBoV1 capsid has been developed to efficiently deliver the cystic fibrosis transmembrane conductance regulator gene to human airway epithelia. Here, we identified both cis-acting elements and trans-acting proteins that are required for HBoV1 DNA replication at the right-end hairpin in HEK293 cells. We localized the minimal replication origin, which contains both NS1 nicking and binding sites, to a 46-nucleotide sequence in the right-end hairpin. The identification of these essential elements of HBoV1 DNA replication acting both in cis and in trans will provide guidance to develop antiviral strategies targeting viral DNA replication at the right-end hairpin and to design next-generation recombinant HBoV1 vectors, a promising tool for gene therapy of lung diseases.


DNA Replication , DNA, Viral/genetics , DNA, Viral/metabolism , Human bocavirus/physiology , Virus Replication , Cell Line , DNA Mutational Analysis , Human bocavirus/genetics , Humans , Protein Binding , Replication Origin , Viral Nonstructural Proteins/metabolism
17.
Arch Virol ; 161(8): 2095-102, 2016 Aug.
Article En | MEDLINE | ID: mdl-27155943

Human bocavirus (HBoV) is a prevalent virus worldwide and is mainly associated with respiratory disorders. Recently, it was detected in several disease conditions, including cancers. Colorectal cancer (CRC) is the third main cause of cancers worldwide. Risk factors that initiate cell transformation include nutritional, hereditary and infectious causes. The aim of the current study was to screen for the presence of HBoV in solid tumors of colorectal cancer and to determine the genotypes of the detected strains. Surgically excised and paraffin-embedded colorectal cancer tissue specimens from 101 male and female patients with and without metastasis were collected over the last four years. Pathological analysis and tumor stages were determined. The presence of HBoV was screened by polymerase chain reaction, and the genotype of the detected HBoV was determined by direct gene sequencing. Most of the examined specimens were adenocarcinoma with mucinous activity in many of them. Twenty-four out of 101 (23.8 %) CRC tissue specimens were found to contain HBoV-1. Low sequence diversity was recorded in the detected strains. The virus was detected in both male and female patients with an age range of 30-75 years. It is proposed that HBoV-1 could play a potential role in the induction of CRC.


Colorectal Neoplasms/virology , Human bocavirus/isolation & purification , Parvoviridae Infections/virology , Adult , Aged , Colon/pathology , Colon/surgery , Colon/virology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/surgery , Female , Human bocavirus/classification , Human bocavirus/genetics , Human bocavirus/physiology , Humans , Male , Middle Aged , Parvoviridae Infections/pathology , Parvoviridae Infections/surgery , Phylogeny
18.
PLoS One ; 11(1): e0147010, 2016.
Article En | MEDLINE | ID: mdl-26807786

Human Bocavirus subtype 1 (HBoV1) is associated with respiratory diseases and may contribute to chronic lung diseases by persisting in the infected host. Here the question was addressed if HBoV infections could contribute to fibrogenesis processes as suggested by previously published clinical observations. Cytokine profiles induced by HBoV infection in CuFi-8 air-liquid interphase cell cultures and in bronchoalveolar lavage fluid (BALF) of 20 HBoV-positive and 12 HBoV-negative patients were analysed by semi-quantitative Western spot blot analyses. Although lots of cytokines were regulated independently of HBoV status, several cytokines associated with lung fibrosis and tumour development, e.g., EGF, VEGF, TARC (CCL17), TNF-α, TNF-ß, TIMP-1, were clearly upregulated in the HBoV-positive cohort. These findings suggest that the development of lung fibrosis might be triggered by HBoV induced cytokine expression.


Cytokines/metabolism , Human bocavirus/physiology , Lung/virology , Parvoviridae Infections/physiopathology , Pneumonia, Viral/physiopathology , Adult , Aged , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/virology , Bronchoalveolar Lavage Fluid/chemistry , Cells, Cultured , Chronic Disease , Coinfection , Cytokines/biosynthesis , HEK293 Cells , Humans , Lung Diseases, Interstitial/complications , Lung Diseases, Interstitial/physiopathology , Lung Diseases, Interstitial/virology , Middle Aged , Pneumonia, Viral/complications , Pneumonia, Viral/virology , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/physiopathology , Pulmonary Fibrosis/virology , Retrospective Studies , Up-Regulation
19.
PLoS Pathog ; 12(1): e1005399, 2016 Jan.
Article En | MEDLINE | ID: mdl-26765330

Human bocavirus 1 (HBoV1) belongs to the genus Bocaparvovirus of the Parvoviridae family, and is an emerging human pathogenic respiratory virus. In vitro, HBoV1 infects well-differentiated/polarized primary human airway epithelium (HAE) cultured at an air-liquid interface (HAE-ALI). Although it is well known that autonomous parvovirus replication depends on the S phase of the host cells, we demonstrate here that the HBoV1 genome amplifies efficiently in mitotically quiescent airway epithelial cells of HAE-ALI cultures. Analysis of HBoV1 DNA in infected HAE-ALI revealed that HBoV1 amplifies its ssDNA genome following a typical parvovirus rolling-hairpin DNA replication mechanism. Notably, HBoV1 infection of HAE-ALI initiates a DNA damage response (DDR) with activation of all three phosphatidylinositol 3-kinase-related kinases (PI3KKs). We found that the activation of the three PI3KKs is required for HBoV1 genome amplification; and, more importantly, we identified that two Y-family DNA polymerases, Pol η and Pol κ, are involved in HBoV1 genome amplification. Overall, we have provided an example of de novo DNA synthesis (genome amplification) of an autonomous parvovirus in non-dividing cells, which is dependent on the cellular DNA damage and repair pathways.


Human bocavirus/physiology , Parvoviridae Infections/virology , Respiratory Mucosa/virology , Virus Replication/physiology , Cells, Cultured , DNA Damage/physiology , DNA Repair/physiology , Fluorescent Antibody Technique , Humans , Immunoblotting
20.
PLoS One ; 10(8): e0135640, 2015.
Article En | MEDLINE | ID: mdl-26267139

To evaluate the role of human bocavirus (hBoV) as a causative agent of respiratory disease, the importance of the viral load in respiratory disease type and severity and the pathogenicity of the different hBoV species, we studied all hBoV-positive nasopharyngeal samples collected from children who attended an emergency room for a respiratory tract infection during three winters (2009-2010, 2011-2012, and 2013-2014). Human bocavirus was detected using the respiratory virus panel fast assay and real-time PCR. Of the 1,823 nasopharyngeal samples, 104 (5.7%) were positive for hBoV; a similar prevalence was observed in all three periods studied. Among hBoV-infected children, 53.8% were between 1-2 years old, and hBoV was detected alone in 57/104 (54.8%) cases. All of the detected hBoV strains belonged to genotype 1. The median hBoV load was significantly higher in samples containing strains with both the N546H and T590S mutations compared to other samples (p<0.05). Children with a single hBoV-1 infection more frequently had upper respiratory tract infections (URTIs) than those who were co-infected (37.0% vs 17.8%, respectively, p = 0.04). The duration of hospitalization was longer among children with high viral loads than that observed among children with low viral loads (8.0 ±2.2 days vs 5.0 ±1.5 days, respectively, p = 0.03), and the use of aerosol therapy was more frequent among children with high viral loads than among those with low viral loads (77.1% vs 55.7%, respectively, p = 0.04). This study shows that hBoV is a relatively uncommon but stable infectious agent in children and that hBoV1 seems to be the only strain detected in Italy in respiratory samples. From a clinical point of view, hBoV1 seems to have in the majority of healthy children relatively low clinical relevance. Moreover, the viral load influences only the duration of hospitalization and the use of aerosol therapy without any association with the site of the respiratory disease.


Human bocavirus/physiology , Parvoviridae Infections/diagnosis , Respiratory Tract Infections/diagnosis , Child, Preschool , Female , Genotype , Human bocavirus/classification , Human bocavirus/genetics , Humans , Incidence , Male , Parvoviridae Infections/pathology , Parvoviridae Infections/virology , Phylogeny , Respiratory Tract Infections/pathology , Respiratory Tract Infections/virology , Viral Load
...