Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(25): e2320782121, 2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38875150

RESUMO

Human bocavirus 1 (HBoV1) is a human parvovirus that causes lower respiratory tract infections in young children. It contains a single-stranded (ss) DNA genome of ~5.5 kb that encodes a small noncoding RNA of 140 nucleotides known as bocavirus-encoded small RNA (BocaSR), in addition to viral proteins. Here, we determined the secondary structure of BocaSR in vivo by using DMS-MaPseq. Our findings reveal that BocaSR undergoes N6-methyladenosine (m6A) modification at multiple sites, which is critical for viral DNA replication in both dividing HEK293 cells and nondividing cells of the human airway epithelium. Mechanistically, we found that m6A-modified BocaSR serves as a mediator for recruiting Y-family DNA repair DNA polymerase (Pol) η and Pol κ likely through a direct interaction between BocaSR and the viral DNA replication origin at the right terminus of the viral genome. Thus, this report represents direct involvement of a viral small noncoding RNA in viral DNA replication through m6A modification.


Assuntos
Adenosina , Replicação do DNA , DNA Viral , DNA Polimerase Dirigida por DNA , RNA Viral , Replicação Viral , Humanos , Adenosina/análogos & derivados , Adenosina/metabolismo , Replicação Viral/genética , DNA Polimerase Dirigida por DNA/metabolismo , DNA Polimerase Dirigida por DNA/genética , DNA Viral/genética , DNA Viral/metabolismo , Células HEK293 , RNA Viral/genética , RNA Viral/metabolismo , Bocavirus Humano/genética , Bocavirus Humano/metabolismo , Genoma Viral/genética , Infecções por Parvoviridae/virologia
2.
J Virol ; 98(3): e0151523, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38323812

RESUMO

Adeno-associated virus (AAV) requires co-infection with helper virus for efficient replication. We previously reported that Human Bocavirus 1 (HBoV1) genes, including NP1, NS2, and BocaSR, were critical for AAV2 replication. Here, we first demonstrate the essential roles of the NP1 protein in AAV2 DNA replication and protein expression. We show that NP1 binds to single-strand DNA (ssDNA) at least 30 nucleotides (nt) in length in a sequence-independent manner. Furthermore, NP1 colocalized with the BrdU-labeled AAV2 DNA replication center, and the loss of the ssDNA-binding ability of NP1 by site-directed mutation completely abolished AAV2 DNA replication. We used affinity-tagged NP1 protein to identify host cellular proteins associated with NP1 in cells cotransfected with the HBoV1 helper genes and AAV2 duplex genome. Of the identified proteins, we demonstrate that NP1 directly binds to the DBD-F domain of the RPA70 subunit with a high affinity through the residues 101-121. By reconstituting the heterotrimer protein RPA in vitro using gel filtration, we demonstrate that NP1 physically associates with RPA to form a heterologous complex characterized by typical fast-on/fast-off kinetics. Following a dominant-negative strategy, we found that NP1-RPA complex mainly plays a role in expressing AAV2 capsid protein by enhancing the transcriptional activity of the p40 promoter. Our study revealed a novel mechanism by which HBoV1 NP1 protein supports AAV2 DNA replication and capsid protein expression through its ssDNA-binding ability and direct interaction with RPA, respectively.IMPORTANCERecombinant adeno-associated virus (rAAV) vectors have been extensively used in clinical gene therapy strategies. However, a limitation of these gene therapy strategies is the efficient production of the required vectors, as AAV alone is replication-deficient in the host cells. HBoV1 provides the simplest AAV2 helper genes consisting of NP1, NS2, and BocaSR. An important question regarding the helper function of HBoV1 is whether it provides any direct function that supports AAV2 DNA replication and protein expression. Also of interest is how HBoV1 interplays with potential host factors to constitute a permissive environment for AAV2 replication. Our studies revealed that the multifunctional protein NP1 plays important roles in AAV2 DNA replication via its sequence-independent ssDNA-binding ability and in regulating AAV2 capsid protein expression by physically interacting with host protein RPA. Our findings present theoretical guidance for the future application of the HBoV1 helper genes in the rAAV vector production.


Assuntos
Proteínas do Capsídeo , Capsídeo , DNA de Cadeia Simples , DNA Viral , Proteínas de Ligação a DNA , Dependovirus , Bocavirus Humano , Proteínas Virais , Humanos , Capsídeo/metabolismo , Proteínas do Capsídeo/biossíntese , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Dependovirus/genética , Dependovirus/crescimento & desenvolvimento , Dependovirus/metabolismo , DNA de Cadeia Simples/biossíntese , DNA de Cadeia Simples/metabolismo , DNA Viral/biossíntese , DNA Viral/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação Viral da Expressão Gênica , Bocavirus Humano/genética , Bocavirus Humano/metabolismo , Cinética , Mutagênese Sítio-Dirigida , Mutação , Regiões Promotoras Genéticas , Ligação Proteica , Domínios Proteicos , Proteínas Virais/genética , Proteínas Virais/metabolismo , Replicação Viral
3.
Sheng Wu Gong Cheng Xue Bao ; 40(2): 485-495, 2024 Feb 25.
Artigo em Chinês | MEDLINE | ID: mdl-38369835

RESUMO

Human bocaparvovirus 1 (HBoV1) is one of the two parvoviruses that infect humans and cause diseases. Infection with HBoV1 in infants and young children aged 2-5 years can lead to mild or severe acute respiratory diseases, with the most severe cases posing a life-threatening risk. Similar to other parvoviruses, the HBoV1 DNA genome consists of two terminal reverse repeats (ITRs) at its ends, which are necessary for viral genome replication. However, up to now, it has remained a technical challenge to clone the entire ITRs through PCR amplification. In this study, we successfully constructed a full-length infectious clone of HBoV1, termed as pSKHBoV1, by synthesizing and cloning the terminal ITRs in a stepwise manner. After transfecting HEK293 cells with the infectious clone pSKHBoV1, we were able to reconstitute the viral replication cycle. This included the expression of key non-structural proteins, post-transcriptional modification and processing of viral RNA, viral genome replication, and potentially the production of progeny virions containing the defined DNA genome. The successful construction of the infectious clone pSKHBoV1 lays the foundation for future studies on HBoV1 replication and propagation, virus-host interaction, and the development of viral vaccines.


Assuntos
Bocavirus Humano , Criança , Humanos , Pré-Escolar , Células HEK293 , Bocavirus Humano/genética , Bocavirus Humano/metabolismo , Replicação Viral/genética , Células Clonais , DNA
4.
New Microbiol ; 45(4): 331-337, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36538298

RESUMO

Human bocavirus 1 (HBoV1) is an important pathogen causing lower respiratory tract infection. The VP1 unique region (VP1u), consisting of 129 amino acids at the N-terminus of the HBoV1 structural protein VP1, is an important component of virus infection. Bioinformatics analysis predicted that HBoV1 VP1u exhibits two bipartite nuclear localization signals (NLS) and contains four basic regions (BRs). The two potential bipartite NLSs consist of BR2 and 3 and BR3 and 4, respectively. In this study, we inserted the truncated vp1u sequences into a double EGFP fusion expression vector and confirmed the vimentin (VIM)-HBoV1 VP1u interaction by mass spectrometry and immunoprecipitation. The results of our IFA analysis showed that all four VP1u BRs displayed strong nuclear transport functions. We further demonstrated that VP1u interacted with VIM and that they colocalized in the cytoplasm. VP1u expression in the cells enhanced the VIM expression, and the VP1u expression also increased upon VIM overexpression, although it was not affected by VIM knockdown. Upon VIM overexpression, VP1u nucleation was significantly enhanced, but was inhibited by VIM downregulation. These results indicate that the VP1u-VIM interaction could be involved in the nuclear transport of VP1u. VP1u nucleation might further affect HBoV1 replication and infection. This study could potentially help clarify the function of VP1u by further revealing the HBoV1 nuclear transport mechanism and provide a new approach for elucidating the molecular mechanism of HBoV1 replication.


Assuntos
Bocavirus Humano , Humanos , Transporte Ativo do Núcleo Celular , Bocavirus Humano/metabolismo , Vimentina/genética , Vimentina/metabolismo
5.
PLoS Pathog ; 18(6): e1010578, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35653410

RESUMO

Human bocavirus 1 (HBoV1), a member of the genus Bocaparvovirus of the family Parvoviridae, causes acute respiratory tract infections in young children. Well-differentiated pseudostratified human airway epithelium cultured at an air-liquid interface (HAE-ALI) is an ideal in vitro culture model to study HBoV1 infection. Unique to other parvoviruses, bocaparvoviruses express a small nonstructured protein NP1 of ~25 kDa from an open reading frame (ORF) in the center of the viral genome. NP1 plays an important role in viral DNA replication and pre-mRNA processing. In this study, we performed an affinity purification assay to identify HBoV1 NP1-inteacting proteins. We identified that Ku70 and RPA70 directly interact with the NP1 at a high binding affinity, characterized with an equilibrium dissociation constant (KD) of 95 nM and 122 nM, respectively. Furthermore, we mapped the key NP1-interacting domains of Ku70 at aa266-439 and of RPA70 at aa181-422. Following a dominant negative strategy, we revealed that the interactions of Ku70 and RPA70 with NP1 play a significant role in HBoV1 DNA replication not only in an in vitro viral DNA replication assay but also in HBoV1-infected HAE-ALI cultures. Collectively, our study revealed a novel mechanism by which HBoV1 NP1 enhances viral DNA replication through its direct interactions with Ku70 and RPA70.


Assuntos
Bocavirus Humano , Infecções por Parvoviridae , Criança , Pré-Escolar , Replicação do DNA , DNA Viral/genética , DNA Viral/metabolismo , Genoma Viral , Bocavirus Humano/genética , Bocavirus Humano/metabolismo , Humanos , Replicação Viral/genética
6.
J Virol ; 96(4): e0184021, 2022 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-34878919

RESUMO

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.


Assuntos
Bocavirus Humano/fisiologia , Autoantígeno Ku/metabolismo , Mucosa Respiratória/virologia , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , Dano ao DNA , Replicação do DNA , DNA Viral/biossíntese , Genoma Viral , Células HEK293 , Bocavirus Humano/metabolismo , Humanos , Autoantígeno Ku/genética , Ligação Proteica , Domínios Proteicos , Mucosa Respiratória/metabolismo , Proteínas não Estruturais Virais/genética , Compartimentos de Replicação Viral/metabolismo
7.
Curr Protoc Microbiol ; 58(1): e107, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32639683

RESUMO

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


Assuntos
Células Epiteliais/virologia , Bocavirus Humano/metabolismo , Transfecção/métodos , Vírion/metabolismo , Cultura de Vírus/métodos , Diferenciação Celular , Linhagem Celular , Genoma Viral , Células HEK293 , Humanos , Infecções por Parvoviridae/metabolismo , Plasmídeos , Sistema Respiratório/virologia , Replicação Viral
8.
J Virol ; 94(2)2020 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-31666379

RESUMO

Human bocavirus 1 (HBoV1), which belongs to the genus Bocaparvovirus of the Parvoviridae family, causes acute respiratory tract infections in young children. In vitro, HBoV1 infects polarized primary human airway epithelium (HAE) cultured at an air-liquid interface (HAE-ALI). HBoV1 encodes a small nonstructural protein, nuclear protein 1 (NP1), that plays an essential role in the maturation of capsid protein (VP)-encoding mRNAs and viral DNA replication. In this study, we determined the broad interactome of NP1 using the proximity-dependent biotin identification (BioID) assay combined with mass spectrometry (MS). We confirmed that two host mRNA processing factors, DEAH-box helicase 15 (DHX15) and cleavage and polyadenylation specificity factor 6 (CPSF6; also known as CFIm68), a subunit of the cleavage factor Im complex (CFIm), interact with HBoV1 NP1 independently of any DNA or mRNAs. Knockdown of CPSF6 significantly decreased the expression of capsid protein but not that of DHX15. We further demonstrated that NP1 directly interacts with CPSF6 in vitro and colocalizes within the virus replication centers. Importantly, we revealed a novel role of CPSF6 in the nuclear import of NP1, in addition to the critical role of CPSF6 in NP1-facilitated maturation of VP-encoding mRNAs. Thus, our study suggests that CPSF6 interacts with NP1 to escort NP1 imported into the nucleus for its function in the modulation of viral mRNA processing and viral DNA replication.IMPORTANCE Human bocavirus 1 (HBoV1) is one of the significant pathogens causing acute respiratory tract infections in young children worldwide. HBoV1 encodes a small nonstructural protein (NP1) that plays an important role in the maturation of viral mRNAs encoding capsid proteins as well as in viral DNA replication. Here, we identified a critical host factor, CPSF6, that directly interacts with NP1, mediates the nuclear import of NP1, and plays a role in the maturation of capsid protein-encoding mRNAs in the nucleus. The identification of the direct interaction between viral NP1 and host CPSF6 provides new insights into the mechanism by which a viral small nonstructural protein facilitates the multiple regulation of viral gene expression and replication and reveals a novel target for potent antiviral drug development.


Assuntos
Proteínas do Capsídeo/biossíntese , Núcleo Celular , Regulação Viral da Expressão Gênica , Bocavirus Humano/metabolismo , Proteínas Nucleares/metabolismo , RNA Mensageiro , RNA Viral , Fatores de Poliadenilação e Clivagem de mRNA/metabolismo , Transporte Ativo do Núcleo Celular , Proteínas do Capsídeo/genética , Núcleo Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/virologia , Células HEK293 , Bocavirus Humano/genética , Humanos , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Proteínas Nucleares/genética , RNA Helicases/genética , RNA Helicases/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Viral/genética , RNA Viral/metabolismo , Fatores de Poliadenilação e Clivagem de mRNA/genética
9.
New Microbiol ; 42(3): 161-165, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31157401

RESUMO

Human bocavirus 1 (HBoV1) refers to a human parvovirus causing acute respiratory tract infection in children. Bocaviruses encode an NP1 protein, which has 47% amino acid homology with NP1 of Minute Virus of Canines (MVC) and Bovine Parvovirus (BPV), but not with any protein of other parvoviruses. NP1 was found to induce apoptosis in Hela cells, which does not depend on viral replication and other protein expression. However, whether NP1 induces pulmonary cell death is unclear. In the present study, we investigate the impacts of NP1 on the autophagy and viability of A549 cells by expressing NP1. The plasmid containing NP1 gene was transfected into A549 cells. The apoptosis of A549 was evaluated by apoptosis detection kit and expression of caspase3. Cell viability and cell migration were detected by CCK8 kit and cell scratch test, respectively. The autophagy-related proteins and HMGB1 were detected by Western blot after NP1 expression in transfected cells. The real-time PCR was employed to detect HMGB1 mRNA. The secretory HMGB1 in supernatant of cell culture was measured by ELISA kit. The transient expression of NP1 did not induce apoptosis in A549 cells, but inhibited cell viability and migration. The expression of Beclin1 and LC3 II increased significantly and that of autophagy substrate P62 decreased dramatically upon transfection of NP1. The expression of NP1 reduced both levels of mRNA and protein HMGB1. The NP1 induced A549 autophagy was activated by STAT3 signaling pathway. HBoV1 NP1 induced autophagy in A549 cells by activating phosphorylation of STAT3 signaling pathway and inhibited A549 cell viability. This study provides insight into further elucidating the replication mechanism of HBoV1.


Assuntos
Autofagia , Sobrevivência Celular , Bocavirus Humano , Proteínas Virais , Células A549 , Autofagia/fisiologia , Caspase 3/genética , Caspase 3/metabolismo , Expressão Gênica , Células HeLa , Bocavirus Humano/genética , Bocavirus Humano/metabolismo , Humanos , Pulmão/citologia , Fosforilação , Fatores de Transcrição STAT/metabolismo , Transfecção , Proteínas Virais/genética , Proteínas Virais/metabolismo
10.
PLoS One ; 13(8): e0202667, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30114253

RESUMO

Both human parvovirus B19 (B19V) and human bocavirus (HBoV) are known to be important human pathogens of the Parvoviridae family. Our earlier investigation demonstrated that both B19V-VP1u and HBoV-VP1u have a significantly disruptive effect on tight junctions (TJs) in A549 cells, implying the essential role of parvovirus in airway infection and lung injury. However, no direct evidence that B19V-VP1u and HBoV-VP1u induce lung injury exists. The present study further investigates the induction of lung injury by B19V-VP1u and HBoV-VP1u in naïve Balb/c mice following subcutaneous injection of PBS, recombinant B19V-VP1u or HBoV-VP1u. The experimental results reveal significantly increased activity, protein expression and ratio of matrix metalloproteinase-9 (MMP-9) to MMP-2 in Balb/c mice that received B19V-VP1u or HBoV-VP1u compared to those that received PBS. Significantly higher levels of inflammatory cytokines, including IL-6 and IL-1ß, and greater lymphocyte infiltration in lung tissue sections were detected in mice that received B19V-VP1u or HBoV-VP1u. Additionally, significantly increased levels of phosphorylated p65 (NF-κB) and MAPK signaling proteins were observed in lung tissue of mice that received B19V-VP1u or HBoV-VP1u compared to those of mice that received PBS. These findings demonstrate for the first time that B19V-VP1u and HBoV-VP1u proteins induce lung inflammatory reactions through p65 (NF-κB) and MAPK signaling.


Assuntos
Bocavirus Humano/metabolismo , Lesão Pulmonar/patologia , Parvovirus B19 Humano/metabolismo , Proteínas Virais/metabolismo , Animais , Feminino , Humanos , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/patologia , Lesão Pulmonar/veterinária , Lesão Pulmonar/virologia , Linfócitos/citologia , Linfócitos/imunologia , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Junções Íntimas/metabolismo , Fator de Transcrição RelA/metabolismo , Proteínas Virais/genética
11.
Mol Med Rep ; 18(4): 4072-4078, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30106148

RESUMO

Human parvovirus B19 (B19) and human bocavirus 1 (HBoV) are the only known pathogenic parvoviruses, and are responsible for a variety of diseases in human beings. Mounting evidence indicates a strong association between B19 infection and cardiac disorders including myocarditis, dilated cardiomyopathy and heart failure. However, very limited information about the role of HBoV in cardiac disorders is known. To elucidate the effects of B19 and HBoV on cardiac disorders, we expressed EGFP­conjugate constructs of B19­VP1 unique region (VP1u) and HBoV­VP1u, along with the mutants EGFP­B19­VP1uD175A and EGFP­HBoV­VP1uV12A, in H9c2 cells by stable transfection. The protein expression levels of EGFP, EGFP­B19­VP1u, EGFP­B19­VP1uD175A, EGFP­HBoV­VP1u and EGFP­HBoV­VP1uV12A in H9c2 cells were observed under a fluorescence microscope and confirmed by western blotting. Secreted phospholipase A2 (sPLA2) activity was detected in B19­VP1u and HBoV­VP1u but not B19­VP1uD175A and HBoV­VP1uV12A recombinant proteins. Significantly higher expression levels of MCP2 and IP­10 mRNA were detected in H9c2 cells that were transfected with pEGFP­B19­VP1u, compared with in those cells transfected with pEGFP­HBoV­VP1u, pEGFP­B19­VP1uD175A or pEGFP­HBoV­VP1uV12A. Significantly higher protein levels of IL­1ß and IL­6 were detected in H9c2 cells transfected with pEGFP­B19­VP1u or pEGFP­HBoV­VP1u, compared with in those cells transfected with pEGFP­B19­VP1uD175A or pEGFP­HBoV­VP1uV12A. Notably, significantly higher expression of both TNF­α and NF­κB was observed only in H9c2 cells transfected with pEGFP­B19­VP1u, but not in those cells transfected with pEGFP­HBoV­VP1u, pEGFP­B19­VP1uD175A or pEGFP­HBoV­VP1uV12A. These findings, to our knowledge for the first time, reveal the difference between B19­VP1u and HBoV­VP1u in H9c2 cells and provide insight into the roles of B19­VP1u and HBoV­VP1u in the pathogenesis of cardiac inflammation.


Assuntos
Proteínas do Capsídeo/metabolismo , Bocavirus Humano/metabolismo , Inflamação/patologia , Miócitos Cardíacos/metabolismo , Parvovirus B19 Humano/metabolismo , Animais , Quimiocina CCL8/genética , Quimiocina CCL8/metabolismo , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Citocinas/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Camundongos , NF-kappa B/metabolismo , Fosfolipases A2/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Recombinantes de Fusão/metabolismo
12.
J Virol ; 92(22)2018 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-30135129

RESUMO

Human bocavirus 1 (HBoV1) encodes a genus-specific protein, NP1, which regulates viral alternative pre-mRNA processing. Similar to NP1 of the related bocavirus minute virus of canine (MVC), HBoV1 NP1 suppressed cleavage and polyadenylation of RNAs at the viral internal polyadenylation site (pA)p. HBoV1 (pA)p is a complex region. It contains 5 significant cleavage and polyadenylation sites, and NP1 was found to regulate only the three of these sites that are governed by canonical AAUAAA hexamer signals. HBoV1 NP1 also facilitated splicing of the upstream intron adjacent to (pA)p. Alternative polyadenylation and splicing of the upstream intron were independent of each other, functioned efficiently within an isolated transcription unit, and were responsive independent of NP1. Characterization of HBoV1 NP1 generalizes its function within the genus Bocaparvovirus, uncovers important differences, and provides important comparisons with MVC NP1 for mechanistic and evolutionary considerations.IMPORTANCE The Parvovirinae are small nonenveloped icosahedral viruses that are important pathogens in many animal species, including humans. The NP1 protein of human bocavirus 1 (HBoV1), similar to NP1 of the bocavirus minute virus of canine (MVC), regulates viral alternative RNA processing by both suppressing polyadenylation at an internal site, (pA)p, and facilitating splicing of an upstream adjacent intron. These effects allow both extension into the capsid gene and splicing of the viral pre-mRNA that correctly registers the capsid gene open reading frame. Characterization of HBoV1 NP1 generalizes this central mode of parvovirus gene regulation to another member of the bocavirus genus and uncovers both important similarities and differences in function compared to MVC NP1 that will be important for future comparative studies.


Assuntos
Processamento Alternativo/genética , Proteínas do Capsídeo/genética , Regulação Viral da Expressão Gênica/genética , Bocavirus Humano/genética , RNA Viral/genética , Proteínas não Estruturais Virais/metabolismo , Capsídeo/metabolismo , Proteínas do Capsídeo/biossíntese , Linhagem Celular , Células HEK293 , Bocavirus Humano/metabolismo , Humanos , Poliadenilação , Replicação Viral/genética
13.
Hum Gene Ther ; 28(8): 612-625, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28490200

RESUMO

Human bocavirus type-1 (HBoV1) has a high tropism for the apical membrane of human airway epithelia. The packaging of a recombinant adeno-associated virus 2 (rAAV2) genome into HBoV1 capsid produces a chimeric vector (rAAV2/HBoV1) that also efficiently transduces human airway epithelia. As such, this vector is attractive for use in gene therapies to treat lung diseases such as cystic fibrosis. However, preclinical development of rAAV2/HBoV1 vectors has been hindered by the fact that humans are the only known host for HBoV1 infection. This study reports that rAAV2/HBoV1 vector is capable of efficiently transducing the lungs of both newborn (3- to 7-day-old) and juvenile (29-day-old) ferrets, predominantly in the distal airways. Analyses of in vivo, ex vivo, and in vitro models of the ferret proximal airway demonstrate that infection of this particular region is less effective than it is in humans. Studies of vector binding and endocytosis in polarized ferret proximal airway epithelial cultures revealed that a lack of effective vector endocytosis is the main cause of inefficient transduction in vitro. While transgene expression declined proportionally with growth of the ferrets following infection at 7 days of age, reinfection of ferrets with rAAV2/HBoV1 at 29 days gave rise to approximately 5-fold higher levels of transduction than observed in naive infected 29-day-old animals. The findings presented here lay the foundation for clinical development of HBoV1 capsid-based vectors for lung gene therapy in cystic fibrosis using ferret models.


Assuntos
Proteínas do Capsídeo/metabolismo , Dependovirus/genética , Vetores Genéticos/genética , Bocavirus Humano/metabolismo , Mucosa Respiratória/metabolismo , Transdução Genética , Animais , Proteínas do Capsídeo/genética , Linhagem Celular , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Furões , Expressão Gênica , Genes Reporter , Xenoenxertos , Bocavirus Humano/genética , Humanos , Pulmão/metabolismo , Camundongos , Parvovirinae/genética , Mucosa Respiratória/virologia , Fatores de Tempo , Transgenes
14.
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
15.
J Virol ; 90(9): 4658-4669, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26912614

RESUMO

UNLABELLED: A novel chimeric parvoviral vector, rAAV2/HBoV1, in which the recombinant adeno-associated virus 2 (rAAV2) genome is pseudopackaged by the human bocavirus 1 (HBoV1) capsid, has been shown to be highly efficient in gene delivery to human airway epithelia (Z. Yan et al., Mol Ther 21:2181-2194, 2013,http://dx.doi.org/10.1038/mt.2013.92). In this vector production system, we used an HBoV1 packaging plasmid, pHBoV1NSCap, that harbors HBoV1 nonstructural protein (NS) and capsid protein (Cap) genes. In order to simplify this packaging plasmid, we investigated the involvement of the HBoV1 NS proteins in capsid protein expression. We found that NP1, a small NS protein encoded by the middle open reading frame, is required for the expression of the viral capsid proteins (VP1, VP2, and VP3). We also found that the other NS proteins (NS1, NS2, NS3, and NS4) are not required for the expression of VP proteins. We performed systematic analyses of the HBoV1 mRNAs transcribed from the pHBoV1NSCap packaging plasmid and its derivatives in HEK 293 cells. Mechanistically, we found that NP1 is required for both the splicing and the read-through of the proximal polyadenylation site of the HBoV1 precursor mRNA, essential functions for the maturation of capsid protein-encoding mRNA. Thus, our study provides a unique example of how a small viral nonstructural protein facilitates the multifaceted regulation of capsid gene expression. IMPORTANCE: A novel chimeric parvoviral vector, rAAV2/HBoV1, expressing a full-length cystic fibrosis transmembrane conductance regulator (CFTR) gene, is capable of correcting CFTR-dependent chloride transport in cystic fibrosis human airway epithelium. Previously, an HBoV1 nonstructural and capsid protein-expressing plasmid, pHBoV1NSCap, was used to package the rAAV2/HBoV1 vector, but yields remained low. In this study, we demonstrated that the nonstructural protein NP1 is required for the expression of capsid proteins. However, we found that the other four nonstructural proteins (NS1 to -4) are not required for expression of capsid proteins. By mutating theciselements that function as internal polyadenylation signals in the capsid protein-expressing mRNA, we constructed a simple HBoV1 capsid protein-expressing gene that expresses capsid proteins as efficiently as pHBoV1NSCap does, and at similar ratios, but independently of NP1. Our study provides a foundation to develop a better packaging system for rAAV2/HBoV1 vector production.


Assuntos
Proteínas do Capsídeo/genética , Regulação Viral da Expressão Gênica , Bocavirus Humano/genética , Bocavirus Humano/metabolismo , Proteínas não Estruturais Virais/metabolismo , Processamento Alternativo , Sequência de Bases , Linhagem Celular , Códon de Iniciação , DNA Complementar , Técnicas de Inativação de Genes , Genoma Viral , Humanos , Dados de Sequência Molecular , Mutação , Poli A/genética , Sítios de Splice de RNA , RNA Mensageiro/genética , RNA Viral , Transcrição Gênica , Ativação Transcricional
16.
PLoS One ; 10(7): e0132162, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26147465

RESUMO

Human adenovirus (HAdV) cause upper and lower respiratory tract infections. However, there are few large prospective studies focused on HAdVs acute infections requiring hospitalization. From 2005 to 2013 a prospective study was conducted on children admitted with acute respiratory infections. Specimens of nasopharyngeal aspirate were taken for virological study by PCR and clinical data was recorded. HAdV specimens were genotyped. Frequency and clinical course of HAdV infections were compared with RSV, rhinovirus (RV), human bocavirus (HBoV) and influenza in the same population. HAdV was detected in 403 cases of 2371 confirmed viral infections (17.2%) , of which 154 were single virus infections (38%). We genotyped 154 HAdVs. The most frequent genotypes were HAdV-3 (24%), HAdV-6 (21%), and HAdV-5 (20%). A total of 262 children had fever (64.9%); 194 suffered hypoxia (48%), and 147 presented infiltrate in chest x-rays (36.4%). The most frequent diagnoses were recurrent wheezing or asthma (51.7%), bronchiolitis (18.3 %), and pneumonia (11.9%), and 46 (11.4%) episodes required prolonged hospitalization (>7 days) due to the severity. Adenovirus single infections were compared with single infections of 598 RSV, 494 RV, 83 influenza and 78 HBoV. Significant clinical differences were found between HAdV, RSV and RV infections.


Assuntos
Infecções por Adenoviridae/epidemiologia , Infecções por Orthomyxoviridae/epidemiologia , Infecções por Parvoviridae/epidemiologia , Infecções por Picornaviridae/epidemiologia , Infecções por Vírus Respiratório Sincicial/epidemiologia , Adenoviridae/genética , Adenoviridae/metabolismo , Infecções por Adenoviridae/genética , Adolescente , Criança , Pré-Escolar , Feminino , Hospitalização , Bocavirus Humano/genética , Bocavirus Humano/metabolismo , Humanos , Lactente , Masculino , Orthomyxoviridae/genética , Orthomyxoviridae/metabolismo , Infecções por Orthomyxoviridae/genética , Infecções por Parvoviridae/genética , Infecções por Picornaviridae/genética , Infecções por Vírus Respiratório Sincicial/genética , Vírus Sinciciais Respiratórios/genética , Vírus Sinciciais Respiratórios/metabolismo , Estudos Retrospectivos , Rhinovirus/genética , Rhinovirus/metabolismo
17.
Wei Sheng Wu Xue Bao ; 53(7): 737-45, 2013 Jul 04.
Artigo em Chinês | MEDLINE | ID: mdl-24195381

RESUMO

OBJECTIVE: We studied the regulating effect of human bocavirus 1 (HBoV1) nonstructural protein NP1 on the activity of cellular transcription factors and the expression of inflammatory cytokine TNF-alpha and IL-6. METHODS: The modulation of NP1 was measured by the Dual Luciferase Reporter Assay System and the expression of cytokines TNF-alpha and IL-6 was detected by ELISA and Real-time PCR. The luciferase based mammalian two-hybrid system was used to analyze whether the function of NP1 protein aroused from oligomerization. RESULTS: The transcription factors AP-1, STAT3 and STAT1 but not NF-kappaB were up-regulated by NP1, which was evidenced by approximately 2-3-fold increase of the luciferase activity compared to the control vector. Moreover, NP1 increased the TNF-alpha mRNA expression, but not contributed to cytokine IL-6 secretion. We also found that the self-interaction did not exist when NPI was solely expressed in 293T cells. CONCLUSION: This study demonstrates for the first time that NP1 may play important roles in activation of transcription factors and up-regulation of inflammatory cytokines expression, suggesting that NP1 be involved in HBoV1 pathogenesis.


Assuntos
Bocavirus Humano/metabolismo , Infecções por Parvoviridae/genética , Fatores de Transcrição/genética , Regulação para Cima , Proteínas não Estruturais Virais/metabolismo , Citocinas/genética , Citocinas/metabolismo , Bocavirus Humano/genética , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Infecções por Parvoviridae/metabolismo , Infecções por Parvoviridae/virologia , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/metabolismo , Fatores de Transcrição/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Proteínas não Estruturais Virais/genética
18.
Virology ; 440(1): 75-83, 2013 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-23507451

RESUMO

Human bocavirus type 1 (HBoV1) is a newly identified pathogen associated with human respiratory tract illnesses. Previous studies demonstrated that proteins of HBoV1 failed to cause cell death, which is considered as a possible common feature of bocaviruses. However, our work showed that the NP1 of HBoV1 induced apoptotic cell death in Hela cells in the absence of viral genome replication and expression of other viral proteins. Mitochondria apoptotic pathway was involved in the NP1-induced apoptosis that was confirmed by apoptotic characteristics including morphological changes, DNA fragmentation and caspase activation. We also demonstrated that the cell cycle of NP1-transfected Hela cells was transiently arrested at G2/M phase followed by rapid appearance of apoptosis and that the N terminal domain of NP1 was critical to its nuclear localization and function in apoptosis induction in Hela cells. These findings might provide alternative information for further study of mechanism of HBoV1 pathogenesis.


Assuntos
Apoptose/fisiologia , Pontos de Checagem do Ciclo Celular/fisiologia , Bocavirus Humano/metabolismo , Proteínas não Estruturais Virais/metabolismo , Caspase 3/genética , Caspase 3/metabolismo , Caspase 9/genética , Caspase 9/metabolismo , Regulação Enzimológica da Expressão Gênica , Regulação Viral da Expressão Gênica , Células HeLa , Bocavirus Humano/genética , Humanos
19.
J Gen Virol ; 94(Pt 6): 1335-1342, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23388199

RESUMO

Human bocavirus (HBoV), closely related to canine minute virus (MVC) and bovine parvovirus (BPV), is a new member of the Bocavirus genus within the Parvoviridae family. The non-structural protein NP1 of HBoV is a nuclear localized protein and plays an important role in DNA replication as well as in the evasion of host innate immunity. In the current study, we provide the first evidence that NP1 possesses a non-classical nuclear localization signal (ncNLS) (amino acids 7-50). Embedded within this ncNLS is a classical bipartite nuclear localization signal (cNLS) (amino acids 14-28), capable of transporting a heterologous cytoplasmic protein ß-galactosidase fusion protein (ß-gal-EGFP) to the nucleus via the classical importin α/ß1-mediated pathway. Amino acids 7-50 containing the cNLS and the ncNLS of NP1 or full-length NP1 interact with importin α1, importin ß1 and importin ß1Δ, which lacks the importin α binding domain, indicating that the nuclear import of NP1 is through both conventional importin α/ß1 heterodimer- and non-classical importinß1-mediated pathways. Given that the arrangement of a cNLS embedded within an ncNLS is unusual in viral proteins, our data together reveal a novel molecular mechanism underlying the nuclear import of HBoV NP1, providing a basis for further understanding its biological function.


Assuntos
Núcleo Celular/metabolismo , Bocavirus Humano/genética , Sinais de Localização Nuclear , Infecções por Parvoviridae/virologia , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/metabolismo , Transporte Ativo do Núcleo Celular , Motivos de Aminoácidos , Sequência de Aminoácidos , Linhagem Celular , Núcleo Celular/virologia , Bocavirus Humano/química , Bocavirus Humano/metabolismo , Humanos , Infecções por Parvoviridae/metabolismo , Proteínas não Estruturais Virais/genética , beta Carioferinas/metabolismo
20.
PLoS One ; 7(11): e48980, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23133667

RESUMO

To determine if human bocavirus 2 (HBoV2) has a circular genome similar to the head-to-tail sequence of HBoV1 and the episomal form of HBoV3, 15 HBoV2 positive samples identified from 553 stool specimens from children with acute diarrhea were tested for a head-to-tail sequence using TaqMan-based real-time PCR. A circular genome with a head-to-tail sequence was identified in one (BJQ435) out of 15 samples tested by nested PCR. The complete circular genome of HBoV2-C1 (BJQ435) was 5307 nt in length and was flanked with a 520 nt-long terminal non-coding region (NCR). The secondary structure of HBoV2 -C1 had some differences compared to HBoV3-E1 (JN086998). Our study indicates that the HBoV genome exists in the form of a head-to-tail monomer and provides more information for understanding the HBoV replication mechanism.


Assuntos
Diarreia/fisiopatologia , Diarreia/virologia , Bocavirus Humano/metabolismo , Adolescente , Sequência de Bases , Criança , Pré-Escolar , China , DNA Viral/análise , Fezes , Feminino , Gastroenterite/genética , Gastroenterite/virologia , Genoma Viral , Humanos , Lactente , Masculino , Dados de Sequência Molecular , Prevalência , Estrutura Secundária de Proteína , Reação em Cadeia da Polimerase em Tempo Real/métodos , Homologia de Sequência do Ácido Nucleico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...