1
|
Ren X, Hei Z, Ji K, Yan Y, Tian C, Wei Y, Sun Y. The Minute Virus of Canines (MVC) Activates the RhoA/ROCK1/MLC2 Signal Transduction Pathway Resulting in the Dissociation of Tight Junctions and Facilitating Occludin-Mediated Viral Infection. Microorganisms 2025; 13:695. [PMID: 40142587 PMCID: PMC11944487 DOI: 10.3390/microorganisms13030695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/16/2025] [Accepted: 03/17/2025] [Indexed: 03/28/2025] Open
Abstract
The Minute Virus of Canines (MVC), belonging to the genus Bocaparvovirus within the family Parvoviridae, is associated with enteritis and embryonic infection in neonatal canines. Viral attachment to host cells is a critical step in infection, and viral protein 2 (VP2) as an important structural protein of MVC influences host selection and infection severity. Nevertheless, little is known about the interaction between VP2 protein and host cells. In this study, we identified that VP2 directly interacts with the kinase domain of RhoA-associated protein kinase 1 (ROCK1) by using mass spectrometry and immunoprecipitation approach and demonstrated that the RhoA/ROCK1/myosin light chain 2 (MLC2) signaling pathway was activated during the early stage of MVC infection in Walter Reed canine cell/3873D (WRD) cells. Further studies indicated that RhoA/ROCK1-mediated phosphorylation of MLC2 triggers the contraction of the actomyosin ring, disrupts tight junctions, and exposes the tight junction protein Occludin, which facilitates the interaction between VP2 and Occludin. Specific inhibitors of RhoA and ROCK1 restored the MVC-induced intracellular translocation of Occludin and the increase in cell membrane permeability. Moreover, the two inhibitors significantly reduced viral protein expression and genomic copy number. Collectively, our study provides the first evidence that there is a direct interaction between the structural protein VP2 of MVC and ROCK1, and that the tight junction protein Occludin can serve as a potential co-receptor for MVC infection, which may offer new targets for anti-MVC strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yuning Sun
- School of Basic Medical Science, Ningxia Medical University, Yinchuan 750004, China; (X.R.); (Z.H.); (K.J.); (Y.Y.); (C.T.); (Y.W.)
| |
Collapse
|
2
|
Zhang X, Qin S, Huang F, Liu H, Wang J, Chen Z, Hao H, Ding S, Liu L, Yu B, Liu Y, Liu H, Guan W. N4-acetylcytidine coordinates with NP1 and CPSF5 to facilitate alternative RNA processing during the replication of minute virus of canines. Nucleic Acids Res 2025; 53:gkaf229. [PMID: 40167508 PMCID: PMC11959542 DOI: 10.1093/nar/gkaf229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 02/07/2025] [Accepted: 03/12/2025] [Indexed: 04/02/2025] Open
Abstract
RNA modifications play crucial roles in RNA metabolism, structure, and functions. N4-acetylcytidine (ac4C) modifications have been shown to enhance stability and translation efficiency of messenger RNAs and viral RNAs. However, the relationship between ac4C and alternative RNA processing remains unexplored. Here, N-acetyltransferase 10 (NAT10) and its catalyzed ac4C modifications on minute virus of canines (MVC) were shown to regulate viral DNA replication and RNA processing, including both the alternative RNA splicing and polyadenylation. Through acRIP-seq and RedaC:T-seq, functional ac4C-modified residue 3311 was identified and characterized, which affected MVC RNA processing rather than altered the viral RNA stability. Ac4C modification at nt 3311 was revealed to participate in NP1-mediated viral RNA processing without influencing RNA affinity of NP1. Meanwhile, CPSF5 was identified to interact with NP1 and mediate viral RNA processing in an ac4C-dependent manner. Further in vitro assays showed that NP1 recruited CPSF5 to MVC RNAs, and the ac4C modification promoted specific binding of CPSF5 to the target region, which ensured precise alternative MVC RNA processing. This study not only reveals the functions of NAT10 and ac4C but also elucidates the mechanisms by which RNA modifications orchestrate MVC proteins and host factors for efficient viral replication and alternative RNA processing.
Collapse
Affiliation(s)
- Xueyan Zhang
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
- Hubei Jiangxia Laboratory, Wuhan, Hubei 430200, China
| | - Shuangkang Qin
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fang Huang
- Hubei Jiangxia Laboratory, Wuhan, Hubei 430200, China
| | - Haizhou Liu
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
| | - Jun Wang
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
| | - Zhen Chen
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
| | - Haojie Hao
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
- Hubei Jiangxia Laboratory, Wuhan, Hubei 430200, China
| | - Shuang Ding
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
| | - Lishi Liu
- Hubei Jiangxia Laboratory, Wuhan, Hubei 430200, China
| | - Baocheng Yu
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yi Liu
- Hubei Jiangxia Laboratory, Wuhan, Hubei 430200, China
| | - Haibin Liu
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
- Hubei Jiangxia Laboratory, Wuhan, Hubei 430200, China
| | - Wuxiang Guan
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
- Hubei Jiangxia Laboratory, Wuhan, Hubei 430200, China
| |
Collapse
|
3
|
Ren X, Hei Z, Ji K, Guo J, Yan Y, Sun Y. Preparation and specific identification of monoclonal antibodies targeting the VP2 structural protein of minute virus of canines. J Immunol Methods 2025; 538:113833. [PMID: 39961493 DOI: 10.1016/j.jim.2025.113833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 01/12/2025] [Accepted: 02/10/2025] [Indexed: 02/21/2025]
Abstract
The Minute Virus of Canines (MVC), classified under the genus Bocaparvovirus, causes severe respiratory and gastrointestinal symptoms in neonatal canines worldwide. The structural protein VP2 is essential for the attachment, infection, uncoating, and induction of the host immunological response to MVC. This study aimed to prepare a monoclonal antibody (mAb) against VP2 using the hybridoma technique. AlphaFold and CavityPlus bioinformatics analysis revealed that the N-terminal region of VP2 (amino acids 1-300) possesses structural characteristics that make it the most suitable target for effective antibody generation. The recombinant plasmid pET-32a(+)-VP2(N300) with fused Trx and His tags was successfully constructed. After immunizing mice, nine hybridoma cell lines were obtained, namely 1G5, 1G5-1, 1I24, 1I24-1, 2E6-1, 2 N9, 3C12-1, 4 M1, and 4 M1-1. The ascitic antibody titers of all cell lines were above 1:100,000. Western blot analysis of Walter Reed canine cells infected with MVC indicated the selection of three strains of monoclonal antibodies with strong specificity: 1G5, 3C12-1, and 4 M1. These three strains can be employed in immunofluorescence (IF) and immunoprecipitation (IP) tests for detecting VP2 protein. The monoclonal antibody mAb VP2 prepared in this study may serve as a valuable tool for detecting MVC and beneficial for investigating the mechanisms of MVC infection.
Collapse
Affiliation(s)
- Xiang Ren
- School of Basic Medical Sciences, Ningxia Medical University, Shengli Street, Xingqing District, Yinchuan 750004, Ningxia, China
| | - Zhiping Hei
- School of Basic Medical Sciences, Ningxia Medical University, Shengli Street, Xingqing District, Yinchuan 750004, Ningxia, China
| | - Kai Ji
- School of Basic Medical Sciences, Ningxia Medical University, Shengli Street, Xingqing District, Yinchuan 750004, Ningxia, China
| | - Jianhui Guo
- School of Basic Medical Sciences, Ningxia Medical University, Shengli Street, Xingqing District, Yinchuan 750004, Ningxia, China
| | - Yan Yan
- School of Basic Medical Sciences, Ningxia Medical University, Shengli Street, Xingqing District, Yinchuan 750004, Ningxia, China
| | - Yuning Sun
- School of Basic Medical Sciences, Ningxia Medical University, Shengli Street, Xingqing District, Yinchuan 750004, Ningxia, China.
| |
Collapse
|
4
|
Prpić J, Keros T, Božiković M, Kamber M, Jemeršić L. Current Insights into Porcine Bocavirus (PBoV) and Its Impact on the Economy and Public Health. Vet Sci 2024; 11:677. [PMID: 39729017 DOI: 10.3390/vetsci11120677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/11/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024] Open
Abstract
Effective control of animal infectious diseases is crucial for maintaining robust livestock production systems worldwide. Porcine meat constitutes approximately 35-40% of global meat production with the largest producers being China and the European Union (EU). Emerging viral pathogens in swine, like porcine bocavirus (PBoV), have not garnered significant attention, leaving their pathogenic characteristics largely unexplored. This review aims to bridge this knowledge gap by conducting a comprehensive analysis of the existing literature on PBoV. We explore the virus's genome structure, discovery, classification, detection methods, pathogenesis, and its potential public health implications. Additionally, we discuss the distribution and economic impact of PBoV, which includes potential losses due to decreased productivity, increased veterinary costs, and trade restrictions. By highlighting the current state of knowledge, this review seeks to enhance the understanding of PBoV, thereby aiding in its prevention and control, and mitigating its economic impact on the swine industry.
Collapse
Affiliation(s)
- Jelena Prpić
- Croatian Veterinary Institute, Savska Cesta 143, 10000 Zagreb, Croatia
| | | | | | - Magda Kamber
- Croatian Veterinary Institute, Savska Cesta 143, 10000 Zagreb, Croatia
| | - Lorena Jemeršić
- Croatian Veterinary Institute, Savska Cesta 143, 10000 Zagreb, Croatia
| |
Collapse
|
5
|
Gong C, He H, Fu Y, Li B, Yang B, Li J, He X, Han J, Zhang Y, Liu G, Guo Q. Development of a Synthetic VP1 Protein Peptide-Based ELISA to Detect Antibodies Against Porcine Bocavirus Group 3. Viruses 2024; 16:1946. [PMID: 39772251 PMCID: PMC11680204 DOI: 10.3390/v16121946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/11/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Porcine bocavirus (PBoV), classified within the genus Bocaparvovirus, has been reported worldwide. PBoV has been divided into group 1, group 2, and group 3. PBoV group 3 (G3) viruses are the most prevalent in China. Currently, effective serological methods for the detection of antibodies against PBoV G3 are limited. In this study, we developed an indirect ELISA using a synthetic VP1 peptide designed on the basis of the conserved region of the PBoV VP1 protein as a coating antigen. Through matrix titration, the optimal coating concentration of the VP1 peptide (0.5 μg/mL), serum dilution (1:200), and working concentration of the secondary antibody (1:50,000) were determined. The cutoff value of this developed ELISA was set as 0.4239. Further investigations revealed that this developed ELISA had no cross-reactivity with positive serum antibodies against FMDV-O, FMDV-A, PRV, ASFV, SF, PCV2, PEDV, and TGEV. The detection limit of the method was a 1:1600 dilution of standard positive serum against PBoV G3. The coefficients of variation for both the intra- and interassay data were lower than 10%. A total of 1373 serum samples collected from 12 provinces in China between 2022 and 2023 were subjected to indirect ELISA. The results showed that 47.56% of the samples were PBoV G3 positive. These results reveal that peptide-based ELISA is a reliable and cost-effective method for detecting PBoV G3 antibodies. It also facilitates the investigation of the prevalence and distribution of PBoV G3.
Collapse
Affiliation(s)
- Chao Gong
- Xinjiang Key Laboratory of New Drug Study and Creation for Herbivorous Animals (XJ-KLNDSCHA), College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi 830052, China; (C.G.); (H.H.); (J.L.); (X.H.); (J.H.); (Y.Z.)
| | - Hui He
- Xinjiang Key Laboratory of New Drug Study and Creation for Herbivorous Animals (XJ-KLNDSCHA), College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi 830052, China; (C.G.); (H.H.); (J.L.); (X.H.); (J.H.); (Y.Z.)
| | - Yuguang Fu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou University, Lanzhou 730000, China; (Y.F.); (B.L.); (B.Y.)
| | - Baoyu Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou University, Lanzhou 730000, China; (Y.F.); (B.L.); (B.Y.)
| | - Bin Yang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou University, Lanzhou 730000, China; (Y.F.); (B.L.); (B.Y.)
| | - Jianlong Li
- Xinjiang Key Laboratory of New Drug Study and Creation for Herbivorous Animals (XJ-KLNDSCHA), College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi 830052, China; (C.G.); (H.H.); (J.L.); (X.H.); (J.H.); (Y.Z.)
| | - Xiaodong He
- Xinjiang Key Laboratory of New Drug Study and Creation for Herbivorous Animals (XJ-KLNDSCHA), College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi 830052, China; (C.G.); (H.H.); (J.L.); (X.H.); (J.H.); (Y.Z.)
| | - Juncheng Han
- Xinjiang Key Laboratory of New Drug Study and Creation for Herbivorous Animals (XJ-KLNDSCHA), College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi 830052, China; (C.G.); (H.H.); (J.L.); (X.H.); (J.H.); (Y.Z.)
| | - Yi Zhang
- Xinjiang Key Laboratory of New Drug Study and Creation for Herbivorous Animals (XJ-KLNDSCHA), College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi 830052, China; (C.G.); (H.H.); (J.L.); (X.H.); (J.H.); (Y.Z.)
| | - Guangliang Liu
- Xinjiang Key Laboratory of New Drug Study and Creation for Herbivorous Animals (XJ-KLNDSCHA), College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi 830052, China; (C.G.); (H.H.); (J.L.); (X.H.); (J.H.); (Y.Z.)
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou University, Lanzhou 730000, China; (Y.F.); (B.L.); (B.Y.)
| | - Qingyong Guo
- Xinjiang Key Laboratory of New Drug Study and Creation for Herbivorous Animals (XJ-KLNDSCHA), College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi 830052, China; (C.G.); (H.H.); (J.L.); (X.H.); (J.H.); (Y.Z.)
| |
Collapse
|
6
|
Qin S, Chen H, Tian C, Chen Z, Zuo L, Zhang X, Hao H, Huang F, Liu H, Sun X, Guan W. NS1-mediated DNMT1 degradation regulates human bocavirus 1 replication and RNA processing. PLoS Pathog 2024; 20:e1012682. [PMID: 39541416 PMCID: PMC11594422 DOI: 10.1371/journal.ppat.1012682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/26/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024] Open
Abstract
Methylation of the DNA genome plays an important role in viral gene inactivation. However, the role of DNA methylation in human bocavirus (HBoV) remains unclear. In this study, the HBoV1 genomic DNA was found extensively methylated at the CHG and CHH sites. Inhibiting DNA methylation with 5-aza-2'-deoxycytidine (DAC) altered the methylation status and reduced viral DNA production, while enhanced the RNA splicing at D1 and D3 sites and the polyadenylation at the proximal polyadenylation site, (pA)p. Knockdown of DNA methyltransferase 1 (DNMT1) had the same effect on viral DNA synthesis and RNA processing as the DAC treatment, indicating that DNMT1 is the major host methyltransferase involved in viral DNA methylation. In addition, the nonstructural protein NS1 promoted DNMT1 degradation through the ubiquitin-proteasome pathway to regulate viral replication and RNA processing. Collectively, the results suggest that DNA methylation and DNMT1 facilitate HBoV replication and are essential for appropriate NS1 localization in the nucleus. DNMT1 degradation through NS1 promotes the virus RNA processing, leading to viral protein expression.
Collapse
Affiliation(s)
- Shuangkang Qin
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Honghe Chen
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Chuchu Tian
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Zhen Chen
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Li Zuo
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xueyan Zhang
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Haojie Hao
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
- Hubei JiangXia Laboratory, Wuhan, Hubei, China
| | - Fang Huang
- Hubei JiangXia Laboratory, Wuhan, Hubei, China
| | - Haibin Liu
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
- Hubei JiangXia Laboratory, Wuhan, Hubei, China
| | - Xiulian Sun
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Wuxiang Guan
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
- Hubei JiangXia Laboratory, Wuhan, Hubei, China
| |
Collapse
|
7
|
Aksu Kuz C, Ning K, Hao S, Cheng F, Qiu J. Role of the membrane-associated accessory protein (MAAP) in adeno-associated virus (AAV) infection. J Virol 2024; 98:e0063324. [PMID: 38775479 PMCID: PMC11237668 DOI: 10.1128/jvi.00633-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 04/28/2024] [Indexed: 06/14/2024] Open
Abstract
Adeno-associated viruses (AAVs) package a single-stranded (ss) DNA genome of 4.7 kb in their capsid of ~20 nm in diameter. AAV replication requires co-infection of a helper virus, such as adenovirus. During the optimization of recombinant AAV production, a small viral nonstructural protein, membrane-associated accessory protein (MAAP), was identified. However, the function of the MAAP in the context of AAV infection remains unknown. Here, we investigated the expression strategy and function of the MAAP during infection of both AAV2 and AAV5 in human embryonic kidney (HEK)293 cells. We found that AAV2 MAAP2 and AAV5 MAAP5 are expressed from the capsid gene (cap)-transcribing mRNA spliced from the donor to the second splice site that encodes VP2 and VP3. Thus, this AAV cap gene transcribes a multicistronic mRNA that can be translated to four viral proteins, MAAP, VP2, AAP, and VP3 in order. In AAV2 infection, MAAP2 predominantly localized in the cytoplasm, alongside the capsid, near the nuclear and plasma membranes, but a fraction of MAAP2 exhibited nuclear localization. In AAV5 infection, MAAP5 revealed a distinct pattern, predominantly localizing within the nucleus. In the cells infected with an MAAP knockout mutant of AAV2 or AAV5, both viral DNA replication and virus replication increased, whereas virus egress decreased, and the decrease in virus egress can be restored by providing MAAP in trans. In summary, MAAP, a novel AAV nonstructural protein translated from a multicistronic viral cap mRNA, not only facilitates cellular egress of AAV but also likely negatively affects viral DNA replication during infection. IMPORTANCE Recombinant adeno-associated virus (rAAV) has been used as a gene delivery vector in clinical gene therapy. In current gene therapies employing rAAV, a high dose of the vector is required. Consequently, there is a high demand for efficient and high-purity vector production systems. In this study, we demonstrated that membrane-associated accessory protein (MAAP), a small viral nonstructural protein, is translated from the same viral mRNA transcript encoding VP2 and VP3. In AAV-infected cells, apart from its prevalent expression in the cytoplasm with localization near the plasma and nuclear membranes, the MAAP also exhibits notable localization within the nucleus. During AAV infection, MAAP expression increases the cellular egress of progeny virions and decreases viral DNA replication and progeny virion production. Thus, the choice of MAAP expression has pros and cons during AAV infection, which could provide a guide to rAAV production.
Collapse
Affiliation(s)
- Cagla Aksu Kuz
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Kang Ning
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Siyuan Hao
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Fang Cheng
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Jianming Qiu
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
8
|
Wubshet AK, Li GX, Li Q, Dai JF, Ding YZ, Zhou L, Qu M, Wang Y, Ma Z, Werid GM, Abera BH, Kebede AT, Sun Y, Yin X, Liu Y, Jie Z. Stability and integrity of self-assembled bovine parvovirus virus‑like particles (BPV‑VLPs) of VP2 and combination of VP1VP2 assisted by baculovirus-insect cell expression: a potential logistical platform for vaccine deployment. Virol J 2024; 21:87. [PMID: 38641833 PMCID: PMC11027344 DOI: 10.1186/s12985-024-02322-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/20/2024] [Indexed: 04/21/2024] Open
Abstract
BACKGROUND Bovine parvovirus (BPV) is an autonomous DNA virus with a smaller molecular size and subtle differences in its structural proteins, unlike other animal parvoviruses. More importantly, this virus has the potential to produce visible to silent economic catastrophes in the livestock business, despite receiving very little attention. Parvoviral virus-like particles (VLPs) as vaccines and as logistical platforms for vaccine deployment are well studied. However, no single experimental report on the role of VP1 in the assembly and stability of BPV-VLPs is available. Furthermore, the self-assembly, integrity and stability of the VLPs of recombinant BPV VP2 in comparison to VP1 VP2 Cap proteins using any expression method has not been studied previously. In this study, we experimentally evaluated the self-assembling ability with which BPV virus-like particles (VLPs) could be synthesized from a single structural protein (VP2) and by integrating both VP2 and VP1 amino acid sequences. METHODS In silico and experimental cloning methods were carried out. His-tagged and without-His-tag VP2 and V1VP2-encoding amino acid sequences were cloned and inserted into pFastbacdual, and insect cell-generated recombinant protein was evaluated by SDS‒PAGE and western blot. Period of infectivity and expression level were determined by IFA. The integrity and stability of the BPV VLPs were evaluated by transmission electron microscopy. The secondary structure of the BPV VLPs from both VP2 and V1VP2 was analyzed by circular dichroism. RESULTS Our findings show that VP2 alone was equally expressed and purified into detectable proteins, and the stability at different temperatures and pH values was not appreciably different between the two kinds of VLPs. Furthermore, BPV-VP2 VLPs were praised for their greater purity and integrity than BPV-VP1VP2 VLPs, as indicated by SDS‒PAGE. Therefore, our research demonstrates that the function of VP1 has no bearing on the stability or integrity of BPV-VLPs. CONCLUSIONS In summary, incredible physiochemically stable BPV VP2-derived VLPs have been found to be promising candidates for the development of multivalent vaccines and immunodiagnostic kits against enteric viruses and to carry heterogeneous epitopes for various economically important livestock diseases.
Collapse
Affiliation(s)
- Ashenafi Kiros Wubshet
- State Key Laboratory of Veterinary Etiological Biology, National/OIE Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, People's Republic of China
- Department of Veterinary Basics and Diagnostic Sciences, College of Veterinary Science, Mekelle University, 2084, Mekelle, Tigray, Ethiopia
| | - Guo-Xiu Li
- State Key Laboratory of Veterinary Etiological Biology, National/OIE Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, People's Republic of China
| | - Qian Li
- State Key Laboratory of Veterinary Etiological Biology, National/OIE Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, People's Republic of China
| | - Jun-Fei Dai
- State Key Laboratory of Veterinary Etiological Biology, National/OIE Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, People's Republic of China
| | - Yao-Zhong Ding
- State Key Laboratory of Veterinary Etiological Biology, National/OIE Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, People's Republic of China
| | - Luoyi Zhou
- State Key Laboratory of Veterinary Etiological Biology, National/OIE Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, People's Republic of China
| | - Min Qu
- State Key Laboratory of Veterinary Etiological Biology, National/OIE Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, People's Republic of China
| | - Yang Wang
- State Key Laboratory of Veterinary Etiological Biology, National/OIE Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, People's Republic of China
| | - Zhongyuan Ma
- State Key Laboratory of Veterinary Etiological Biology, National/OIE Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, People's Republic of China
| | - Gebremeskel Mamu Werid
- Davies Livestock Research Centre, School of Animal & Veterinary Sciences, University of Adelaide, Roseworthy Campus, Roseworthy, SA, 5371, Australia
| | - Birhanu Hadush Abera
- Department of Veterinary Basics and Diagnostic Sciences, College of Veterinary Science, Mekelle University, 2084, Mekelle, Tigray, Ethiopia
| | - Asmelash Tassew Kebede
- State Key Laboratory of Veterinary Etiological Biology, National/OIE Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, People's Republic of China
- Department of Animal Science, College of Agriculture and Natural Resources, Raya University, 92, Maychew, Tigray, Ethiopia
| | - Yuefeng Sun
- State Key Laboratory of Veterinary Etiological Biology, National/OIE Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, People's Republic of China
| | - Xiangping Yin
- State Key Laboratory of Veterinary Etiological Biology, National/OIE Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, People's Republic of China
| | - Yongsheng Liu
- State Key Laboratory of Veterinary Etiological Biology, National/OIE Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, People's Republic of China.
- College of Animal Science & Technology (CAST), Hebei Normal University of Science & Technology (HNUST), Qinhuangdao, People's Republic of China.
| | - Zhang Jie
- State Key Laboratory of Veterinary Etiological Biology, National/OIE Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, People's Republic of China.
- College of Animal Science & Technology (CAST), Hebei Normal University of Science & Technology (HNUST), Qinhuangdao, People's Republic of China.
| |
Collapse
|
9
|
Zhang X, Guo J, Xu H, Ding S, Liu L, Chen Z, Yang J, Liu Y, Hao H, Huang F, Qiu J, Guan W, Sun Y, Liu H. NS1-mediated enhancement of MVC transcription and replication promoted by KAT5/H4K12ac. J Virol 2024; 98:e0169523. [PMID: 38349085 PMCID: PMC10949499 DOI: 10.1128/jvi.01695-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/07/2023] [Indexed: 03/20/2024] Open
Abstract
Histone modifications function in both cellular and viral gene expression. However, the roles of acetyltransferases and histone acetylation in parvoviral infection remain poorly understood. In the current study, we found the histone deacetylase (HDAC) inhibitor, trichostatin A (TSA), promoted the replication and transcription of parvovirus minute virus of canines (MVC). Notably, the expression of host acetyltransferases KAT5, GTF3C4, and KAT2A was increased in MVC infection, as well as H4 acetylation (H4K12ac). KAT5 is not only responsible for H4K12ac but also crucial for viral replication and transcription. The viral nonstructural protein NS1 interacted with KAT5 and enhanced its expression. Further study showed that Y44 in KAT5, which may be tyrosine-phosphorylated, is indispensable for NS1-mediated enhancement of KAT5 and efficient MVC replication. The data demonstrated that NS1 interacted with KAT5, which resulted in an enhanced H4K12ac level to promote viral replication and transcription, implying the epigenetic addition of H4K12ac in viral chromatin-like structure by KAT5 is vital for MVC replication.IMPORTANCEParvoviral genomes are chromatinized with host histones. Therefore, histone acetylation and related acetyltransferases are required for the virus to modify histones and open densely packed chromatin structures. This study illustrated that histone acetylation status is important for MVC replication and transcription and revealed a novel mechanism that the viral nonstructural protein NS1 hijacks the host acetyltransferase KAT5 to enhance histone acetylation of H4K12ac, which relies on a potential tyrosine phosphorylation site, Y44 in KAT5. Other parvoviruses share a similar genome organization and coding potential and may adapt a similar strategy for efficient viral replication and transcription.
Collapse
Affiliation(s)
- Xueyan Zhang
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Jianhui Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Huanzhou Xu
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Shuang Ding
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Lishi Liu
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Zhen Chen
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
- Hubei Jiangxia Laboratory, Wuhan, Hubei, China
| | - Jingwen Yang
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
- Hubei Jiangxia Laboratory, Wuhan, Hubei, China
| | - Yi Liu
- Hubei Jiangxia Laboratory, Wuhan, Hubei, China
| | - Haojie Hao
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
- Hubei Jiangxia Laboratory, Wuhan, Hubei, China
| | - Fang Huang
- Hubei Jiangxia Laboratory, Wuhan, Hubei, China
| | - Jianming Qiu
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Wuxiang Guan
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
- Hubei Jiangxia Laboratory, Wuhan, Hubei, China
| | - Yuning Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Haibin Liu
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
- Hubei Jiangxia Laboratory, Wuhan, Hubei, China
| |
Collapse
|
10
|
Zhao Y, Liu W, Li Y, Ma J, Liu T, Cui H, Deng Y, Liao X, Wang Z. Human Bocavirus 1 NP1 acts as an ssDNA-binding protein to help AAV2 DNA replication and cooperates with RPA to regulate AAV2 capsid expression. J Virol 2024; 98:e0151523. [PMID: 38323812 PMCID: PMC10949510 DOI: 10.1128/jvi.01515-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 01/14/2024] [Indexed: 02/08/2024] Open
Abstract
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.
Collapse
Affiliation(s)
- Yanqun Zhao
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, Henan University, Kaifeng, Henan, China
| | - Wei Liu
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, Henan University, Kaifeng, Henan, China
| | - Yanjie Li
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, Henan University, Kaifeng, Henan, China
| | - Jing Ma
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, Henan University, Kaifeng, Henan, China
| | - Ting Liu
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, Henan University, Kaifeng, Henan, China
| | - Huichan Cui
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, Henan University, Kaifeng, Henan, China
| | - Yongheng Deng
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, Henan University, Kaifeng, Henan, China
| | - Xiaochen Liao
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, Henan University, Kaifeng, Henan, China
| | - Zekun Wang
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, Henan University, Kaifeng, Henan, China
| |
Collapse
|
11
|
Capozza P, Buonavoglia A, Pratelli A, Martella V, Decaro N. Old and Novel Enteric Parvoviruses of Dogs. Pathogens 2023; 12:pathogens12050722. [PMID: 37242392 DOI: 10.3390/pathogens12050722] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/10/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
Parvovirus infections have been well known for around 100 years in domestic carnivores. However, the use of molecular assays and metagenomic approaches for virus discovery and characterization has led to the detection of novel parvovirus species and/or variants in dogs. Although some evidence suggests that these emerging canine parvoviruses may act as primary causative agents or as synergistic pathogens in the diseases of domestic carnivores, several aspects regarding epidemiology and virus-host interaction remain to be elucidated.
Collapse
Affiliation(s)
- Paolo Capozza
- Department of Veterinary Medicine, University of Bari Aldo Moro, 70010 Valenzano, Italy
| | - Alessio Buonavoglia
- Department of Biomedical and Neuromotor Sciences, Dental School, Via Zamboni 33, 40126 Bologna, Italy
| | - Annamaria Pratelli
- Department of Veterinary Medicine, University of Bari Aldo Moro, 70010 Valenzano, Italy
| | - Vito Martella
- Department of Veterinary Medicine, University of Bari Aldo Moro, 70010 Valenzano, Italy
| | - Nicola Decaro
- Department of Veterinary Medicine, University of Bari Aldo Moro, 70010 Valenzano, Italy
| |
Collapse
|
12
|
Ning K, Kuz CA, Cheng F, Feng Z, Yan Z, Qiu J. Adeno-Associated Virus Monoinfection Induces a DNA Damage Response and DNA Repair That Contributes to Viral DNA Replication. mBio 2023; 14:e0352822. [PMID: 36719192 PMCID: PMC9973366 DOI: 10.1128/mbio.03528-22] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 01/03/2023] [Indexed: 02/01/2023] Open
Abstract
Adeno-associated virus (AAV) belongs to the Dependoparvovirus genus of the Parvoviridae family. AAV replication relies on a helper virus, such as adenovirus (Ad). Co-infection of AAV and Ad induces a DNA damage response (DDR), although its function in AAV DNA replication remains unknown. In this study, monoinfection of AAV2 in HEK293T cells expressing a minimal set of Ad helper genes was used to investigate the role of the DDR solely induced by AAV. We found that AAV2 DNA replication, but not single stranded (ss)DNA genome accumulation and Rep expression only, induced a robust DDR in HEK293T cells. The induced DDR featured the phosphorylation of replication protein A32 (RPA32), histone variant H2AX (H2A histone family member X), and all 3 phosphatidylinositol 3-kinase-related kinases (PIKKs). We also found that the kinase ataxia telangiectasia and Rad3-related protein (ATR) plays a major role in AAV2 DNA replication and that Y family DNA repair DNA polymerases η (Pol η) and Pol κ contribute to AAV2 DNA replication both in vitro and in HEK293T cells. Knockout of Pol η and Pol κ in HEK293T cells significantly decreased wild-type AAV2 replication and recombinant AAV2 production. Thus, our study has proven that AAV2 DNA replication induces a DDR, which in turn initiates a DNA repairing process that partially contributes to the viral genome amplification in HEK293T cells. IMPORTANCE Recombinant AAV (rAAV) has emerged as one of the preferred delivery vectors for clinical gene therapy. rAAV production in HEK293 cells by transfection of a rAAV transgene plasmid, an AAV Rep and Cap expression packaging plasmid, and an Ad helper plasmid remains the popular method. Here, we demonstrated that the high fidelity Y family DNA repair DNA polymerase, Pol η, and Pol κ, plays a significant role in AAV DNA replication and rAAV production in HEK293T cells. Understanding the AAV DNA replication mechanism in HEK293T cells could provide clues to increase rAAV vector yield produced from the transfection method. We also provide evidence that the ATR-mediated DNA repair process through Pol η and Pol κ is one of the mechanisms to amplify AAV genome, which could explain AAV replication and rAAV ssDNA genome conversion in mitotic quiescent cells.
Collapse
Affiliation(s)
- Kang Ning
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Cagla Aksu Kuz
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Fang Cheng
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Zehua Feng
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa, USA
| | - Ziying Yan
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa, USA
| | - Jianming Qiu
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
13
|
The small nonstructural protein NP1 of human bocavirus 1 directly interacts with Ku70 and RPA70 and facilitates viral DNA replication. PLoS Pathog 2022; 18:e1010578. [PMID: 35653410 PMCID: PMC9197078 DOI: 10.1371/journal.ppat.1010578] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 06/14/2022] [Accepted: 05/10/2022] [Indexed: 11/19/2022] Open
Abstract
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.
Collapse
|
14
|
Jager MC, Tomlinson JE, Lopez-Astacio RA, Parrish CR, Van de Walle GR. Small but mighty: old and new parvoviruses of veterinary significance. Virol J 2021; 18:210. [PMID: 34689822 PMCID: PMC8542416 DOI: 10.1186/s12985-021-01677-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 10/08/2021] [Indexed: 12/14/2022] Open
Abstract
In line with the Latin expression "sed parva forti" meaning "small but mighty," the family Parvoviridae contains many of the smallest known viruses, some of which result in fatal or debilitating infections. In recent years, advances in metagenomic viral discovery techniques have dramatically increased the identification of novel parvoviruses in both diseased and healthy individuals. While some of these discoveries have solved etiologic mysteries of well-described diseases in animals, many of the newly discovered parvoviruses appear to cause mild or no disease, or disease associations remain to be established. With the increased use of animal parvoviruses as vectors for gene therapy and oncolytic treatments in humans, it becomes all the more important to understand the diversity, pathogenic potential, and evolution of this diverse family of viruses. In this review, we discuss parvoviruses infecting vertebrate animals, with a special focus on pathogens of veterinary significance and viruses discovered within the last four years.
Collapse
Affiliation(s)
- Mason C Jager
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Joy E Tomlinson
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Robert A Lopez-Astacio
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Colin R Parrish
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Gerlinde R Van de Walle
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
15
|
Hairpin transfer-independent Parvovirus DNA Replication Produces Infectious Virus. J Virol 2021; 95:e0110821. [PMID: 34346761 DOI: 10.1128/jvi.01108-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Parvoviruses package a linear single-stranded DNA genome with hairpin structures at both ends. It has been thought that terminal hairpin sequences are indispensable for viral DNA replication. Here, we provide evidence that the hairpin-deleted duplex genomes of human bocavirus 1 (HBoV1) replicate in human embryonic kidney (HEK) 293 cells. We propose an alternative model for HBoV1 DNA replication in which the leading strand can initiate strand-displacement without "hairpin-transfer." The transfection of the HBoV1 duplex genomes that retain a minimal replication origin at the right-end (OriR), but with extensive deletions in the right-end hairpin (REH), generated viruses in HEK293 cells at a level 10-20 times lower than the wild-type (WT) duplex genome. Importantly, these viruses that have a genome with various deletions after the OriR, but not the one retaining only the OriR, replicated in polarized human airway epithelia. We discovered that the 18-nt sequence (nt 5,403-5,420) beyond the OriR was sufficient to confer virus replication in polarized human airway epithelia, although its progeny virus production was ∼5 times lower than that of the WT virus. Thus, our study demonstrates that hairpin transfer-independent productive parvovirus DNA replication can occur. Importance Hairpin transfer-independent parvovirus replication was modeled with human bocavirus 1 (HBoV1) duplex genomes whose 5' hairpin structure was ablated by various deletions. In HEK293 cells, these duplex viral genomes with ablated 5'/hairpin sequence replicated efficiently and generated viruses that productively infected polarized human airway epithelium. Thus, for the first time, we reveal a previously unknown phenomenon that the productive parvovirus DNA replication does not depend on the hairpin sequence at REH to initiate "rolling hairpin" DNA replication. Notably, the intermediates of viral DNA replication, as revealed two-dimensional electrophoresis, from transfections of hairpin sequence-deleted duplex genome and full-length genome in HEK293 cells, as well as from virus infection of polarized human airway epithelia are similar. Thus, the establishment of the hairpin transfer-independent parvoviral DNA replication deepens our understanding in viral DNA replication and may have implications in development of parvovirus-based viral vectors with alternative properties.
Collapse
|
16
|
The N-terminal 5-68 amino acids domain of the minor capsid protein VP1 of human parvovirus B19 enters human erythroid progenitors and inhibits B19 infection. J Virol 2021; 95:JVI.00466-21. [PMID: 33952637 PMCID: PMC8223926 DOI: 10.1128/jvi.00466-21] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Parvovirus B19 (B19V) infection causes diseases in humans ranging from the mild erythema infectiosum to severe hematological disorders. The unique region of the minor structural protein VP1 (VP1u) of 227 amino acids harbors strong neutralizing epitopes which elicit dominant immune responses in patients. Recent studies have shown that the VP1u selectively binds to and enters B19V permissive cells through an unknown cellular proteinaceous receptor. In the present study, we demonstrated that purified recombinant VP1u effectively inhibits B19V infection of ex vivo expanded primary human erythroid progenitors. Furthermore, we identified the amino acid sequence 5-68 of the VP1 (VP1u5-68aa) is sufficient to confer the inhibition of B19V infection at a level similar to that of the full-length VP1u. In silico structure prediction suggests that the VP1u5-68aa contains three α-helices. Importantly, we found that the inhibition capability of the minimal domain VP1u5-68aa is independent of its dimerization but is likely dependent on the structure of the three predicated α-helices. As VP1u5-68aa outcompetes the full-length VP1u in entering cells, we believe that VP1u5-68aa functions as a receptor-binding ligand during virus entry. Finally, we determined the effective inhibition potency of VP1u5-68aa in B19V infection of human erythroid progenitors, which has a half maximal effective concentration (EC50) of 67 nM, suggesting an anti-viral peptide candidate to combat B19V infection.IMPORTANCEHuman parvovirus B19 infection causes severe hematological disorders, including transient aplastic crisis, pure red cell aplasia, and hydrops fetalis. A productive B19 infection is highly restricted to human erythroid progenitors in human bone marrow and fetal liver. In the current study, we identified that the N-terminal 5-68 amino acids domain of the minor viral capsid protein VP1 enters ex vivo expanded human erythroid progenitors, which is nearly 5 times more efficient than the full-length VP1 unique region (1-227aa). Importantly, purified recombinant 5-68aa of the VP1 has a high efficiency in inhibition of parvovirus B19 infection of human erythroid progenitors, which has a half maximal effective concentration (EC50) of 67 nM and a low cytotoxicity. The N-terminal 5-68 amino acids holds the potential as an effective antiviral of parvovirus B19 caused hematological disorders, as well as a carrier to deliver proteins to human erythroid progenitors.
Collapse
|
17
|
Shao L, Shen W, Wang S, Qiu J. Recent Advances in Molecular Biology of Human Bocavirus 1 and Its Applications. Front Microbiol 2021; 12:696604. [PMID: 34220786 PMCID: PMC8242256 DOI: 10.3389/fmicb.2021.696604] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 05/12/2021] [Indexed: 11/25/2022] Open
Abstract
Human bocavirus 1 (HBoV1) was discovered in human nasopharyngeal specimens in 2005. It is an autonomous human parvovirus and causes acute respiratory tract infections in young children. HBoV1 infects well differentiated or polarized human airway epithelial cells in vitro. Unique among all parvoviruses, HBoV1 expresses 6 non-structural proteins, NS1, NS1-70, NS2, NS3, NS4, and NP1, and a viral non-coding RNA (BocaSR), and three structural proteins VP1, VP2, and VP3. The BocaSR is the first identified RNA polymerase III (Pol III) transcribed viral non-coding RNA in small DNA viruses. It plays an important role in regulation of viral gene expression and a direct role in viral DNA replication in the nucleus. HBoV1 genome replication in the polarized/non-dividing airway epithelial cells depends on the DNA damage and DNA repair pathways and involves error-free Y-family DNA repair DNA polymerase (Pol) η and Pol κ. Importantly, HBoV1 is a helper virus for the replication of dependoparvovirus, adeno-associated virus (AAV), in polarized human airway epithelial cells, and HBoV1 gene products support wild-type AAV replication and recombinant AAV (rAAV) production in human embryonic kidney (HEK) 293 cells. More importantly, the HBoV1 capsid is able to pseudopackage an rAAV2 or rHBoV1 genome, producing the rAAV2/HBoV1 or rHBoV1 vector. The HBoV1 capsid based rAAV vector has a high tropism for human airway epithelia. A deeper understanding in HBoV1 replication and gene expression will help find a better way to produce the rAAV vector and to increase the efficacy of gene delivery using the rAAV2/HBoV1 or rHBoV1 vector, in particular, to human airways. This review summarizes the recent advances in gene expression and replication of HBoV1, as well as the use of HBoV1 as a parvoviral vector for gene delivery.
Collapse
Affiliation(s)
- Liting Shao
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Weiran Shen
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Shengqi Wang
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Jianming Qiu
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
18
|
Dudek AM, Porteus MH. Answered and Unanswered Questions in Early-Stage Viral Vector Transduction Biology and Innate Primary Cell Toxicity for Ex-Vivo Gene Editing. Front Immunol 2021; 12:660302. [PMID: 34122418 PMCID: PMC8195279 DOI: 10.3389/fimmu.2021.660302] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 05/04/2021] [Indexed: 01/07/2023] Open
Abstract
Adeno-associated virus is a highly efficient DNA delivery vehicle for genome editing strategies that employ CRISPR/Cas9 and a DNA donor for homology-directed repair. Many groups have used this strategy in development of therapies for blood and immune disorders such as sickle-cell anemia and severe-combined immunodeficiency. However, recent events have called into question the immunogenicity of AAV as a gene therapy vector and the safety profile dictated by the immune response to this vector. The target cells dictating this response and the molecular mechanisms dictating cellular response to AAV are poorly understood. Here, we will investigate the current known AAV capsid and genome interactions with cellular proteins during early stage vector transduction and how these interactions may influence innate cellular responses. We will discuss the current understanding of innate immune activation and DNA damage response to AAV, and the limitations of what is currently known. In particular, we will focus on pathway differences in cell line verses primary cells, with a focus on hematopoietic stem and progenitor cells (HSPCs) in the context of ex-vivo gene editing, and what we can learn from HSPC infection by other parvoviruses. Finally, we will discuss how innate immune and DNA damage response pathway activation in these highly sensitive stem cell populations may impact long-term engraftment and clinical outcomes as these gene-editing strategies move towards the clinic, with the aim to propose pathways relevant for improved hematopoietic stem cell survival and long-term engraftment after AAV-mediated genome editing.
Collapse
Affiliation(s)
- Amanda Mary Dudek
- Department of Pediatrics, Stanford University, Stanford, CA, United States.,Department of Pediatrics, School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Matthew Hebden Porteus
- Department of Pediatrics, Stanford University, Stanford, CA, United States.,Department of Pediatrics, School of Medicine, Stanford University, Palo Alto, CA, United States
| |
Collapse
|
19
|
Abstract
Human bocavirus 1 (HBoV1) and HBoV2-4 infect children and immunocompromised individuals, resulting in respiratory and gastrointestinal infections, respectively. Using cryo-electron microscopy and image reconstruction, the HBoV2 capsid structure was determined to 2.7 Å resolution at pH 7.4 and compared to the previously determined HBoV1, HBoV3, and HBoV4 structures. Consistent with previous findings, surface variable region (VR) III of the capsid protein VP3, proposed as a host tissue-tropism determinant, was structurally similar among the gastrointestinal strains HBoV2-4, but differed from HBoV1 with its tropism for the respiratory tract. Towards understanding the entry and trafficking properties of these viruses, HBoV1 and HBoV2 were further analyzed as species representatives of the two HBoV tropisms. Their cell surface glycan-binding characteristics were analyzed, and capsid structures determined to 2.5-2.7 Å resolution at pH 5.5 and 2.6, conditions normally encountered during infection. The data showed that glycans with terminal sialic acid, galactose, GlcNAc or heparan sulfate moieties do not facilitate HBoV1 or HBoV2 cellular attachment. With respect to trafficking, conformational changes common to both viruses were observed at low pH conditions localized to the VP N-terminus under the 5-fold channel, in the surface loops VR-I and VR-V and specific side-chain residues such as cysteines and histidines. The 5-fold conformational movements provide insight into the potential mechanism of VP N-terminal dynamics during HBoV infection and side-chain modifications highlight pH-sensitive regions of the capsid.IMPORTANCE Human bocaviruses (HBoVs) are associated with disease in humans. However, the lack of an animal model and a versatile cell culture system to study their life cycle limits the ability to develop specific treatments or vaccines. This study presents the structure of HBoV2, at 2.7 Å resolution, determined for comparison to the existing HBoV1, HBoV3, and HBoV4 structures, to enable the molecular characterization of strain and genus-specific capsid features contributing to tissue tropism and antigenicity. Furthermore, HBoV1 and HBoV2 structures determined under acidic conditions provide insight into capsid changes associated with endosomal and gastrointestinal acidification. Structural rearrangements of the capsid VP N-terminus, at the base of the 5-fold channel, demonstrate a disordering of a "basket" motif as pH decreases. These observations begin to unravel the molecular mechanism of HBoV infection and provide information for control strategies.
Collapse
|
20
|
Zheng LL, Cui JT, Qiao H, Li XS, Li XK, Chen HY. Detection and genetic characteristics of porcine bocavirus in central China. Arch Virol 2021; 166:451-460. [PMID: 33392822 DOI: 10.1007/s00705-020-04879-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 09/30/2020] [Indexed: 10/22/2022]
Abstract
To investigate the epidemic profile and genetic diversity of porcine bocavirus (PBoV), 281 clinical samples, including 236 intestinal tissue samples and 45 fecal samples were collected from diarrheic piglets on 37 different pig farms in central China, and two SYBR Green I-based quantitative PCR assays were developed to detect PBoV1/2 and PBoV3/4/5, respectively. One hundred forty-eight (52.67%) of the 281 clinical samples were positive for PBoV1/2, 117 (41.63%) were positive for PBoV3/4/5, 55 (19.57%) were positive for both PBoV1/2 and PBoV3/4/5, and 86.49% (32/37) of the pig farms were positive for PBoV. Overall, the prevalence of PBoV was 74.73% (210/281) in central China. Subsequently, nearly full-length genomic sequences of two PBoV strains (designated CH/HNZM and PBoV-TY) from two different farms were determined. Phylogenetic analysis demonstrated that the two PBoV strains obtained in this study belonged to the PBoV G2 group and had a close relationship to 10 other PBoV G2 strains but differed genetically from PBoV G1, PBoV G3, and seven other bocaviruses. CH/HNZM and PBoV-TY were closely related to the PBoV strain GD18 (KJ755666), which may be derived from the PBoV strains 0912/2012 (MH558677) and 57AT-HU (KF206160) through recombination. Compared with reference strain ZJD (HM053694)-China, more amino acid variation was found in the NS1 proteins of CH/HNZM and PBoV-TY. These data extend our understanding of the molecular epidemiology and evolution of PBoV.
Collapse
Affiliation(s)
- Lan-Lan Zheng
- Zhengzhou Key Laboratory for Pig Disease Prevention and Control, College of Veterinary Medicine, Henan Agricultural University, Nongye Road 63#, Zhengzhou, 450002, Henan Province, People's Republic of China
| | - Jian-Tao Cui
- Zhengzhou Key Laboratory for Pig Disease Prevention and Control, College of Veterinary Medicine, Henan Agricultural University, Nongye Road 63#, Zhengzhou, 450002, Henan Province, People's Republic of China
| | - Han Qiao
- College of Life Science, South China Agricultural University, Guangzhou, 510642, Guangdong Province, People's Republic of China
| | - Xin-Sheng Li
- Zhengzhou Key Laboratory for Pig Disease Prevention and Control, College of Veterinary Medicine, Henan Agricultural University, Nongye Road 63#, Zhengzhou, 450002, Henan Province, People's Republic of China
| | - Xiao-Kang Li
- College of Animal Science and Technology, Henan University of Science and Technology, 263 Kaiyuan Avenue, Luoyang, 471000, Henan Province, People's Republic of China.
| | - Hong-Ying Chen
- Zhengzhou Key Laboratory for Pig Disease Prevention and Control, College of Veterinary Medicine, Henan Agricultural University, Nongye Road 63#, Zhengzhou, 450002, Henan Province, People's Republic of China. .,College of Veterinary Medicine, Henan Agricultural University, Zhengdong New District Longzi Lake#15, Zhengzhou, 450046, Henan Province, People's Republic of China.
| |
Collapse
|
21
|
Xiao Y, Wang H, Feng L, Pan J, Chen Z, Wang H, Yang S, Shen Q, Wang X, Shan T, Zhang W. Fecal, oral, blood and skin virome of laboratory rabbits. Arch Virol 2020; 165:2847-2856. [PMID: 33034764 PMCID: PMC7546134 DOI: 10.1007/s00705-020-04808-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 08/11/2020] [Indexed: 12/24/2022]
Abstract
Here, we investigated the fecal, oral, blood, and skin virome of 10 laboratory rabbits using a viral metagenomic method. In the oral samples, we detected a novel polyomavirus (RabPyV), and phylogenetic analysis based on the large T antigen, VP1 and VP2 regions indicated that the novel strain might have undergone a recombination event. Recombination analysis based on related genomes confirmed that RabPyV is a multiple recombinant between rodent-like and avian-like polyomaviruses. In fecal samples, three partial or complete genome sequences of viruses belonging to the families Picobirnaviridae, Parvoviridae, Microviridae and Coronaviridae were characterized, and phylogenetic trees were constructed based on the predicted amino acid sequences of viral proteins. This study increases the amount of genetic information on viruses present in laboratory rabbits.
Collapse
Affiliation(s)
- Yuqing Xiao
- Department of Microbiology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Hao Wang
- Department of Clinical Laboratory, Huai'an Hospital, Xuzhou Medical University, Huai'an, 223002, Jiangsu, China
| | - Lei Feng
- Department of Microbiology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Jiamin Pan
- Department of Microbiology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Zeyu Chen
- Department of Microbiology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Huiling Wang
- Department of Microbiology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Shixing Yang
- Department of Microbiology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Quan Shen
- Department of Microbiology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Xiaochun Wang
- Department of Microbiology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Tongling Shan
- Department of Swine Infectious Disease, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China.
| | - Wen Zhang
- Department of Microbiology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China.
| |
Collapse
|
22
|
Hao S, Ning K, Wang X, Wang J, Cheng F, Ganaie SS, Tavis JE, Qiu J. Establishment of a Replicon Reporter of the Emerging Tick-Borne Bourbon Virus and Use It for Evaluation of Antivirals. Front Microbiol 2020; 11:572631. [PMID: 33013808 PMCID: PMC7506111 DOI: 10.3389/fmicb.2020.572631] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 08/21/2020] [Indexed: 01/01/2023] Open
Abstract
Bourbon virus (BRBV) was first isolated from a patient hospitalized at the University of Kansas Hospital in 2014. Since then, several deaths have been reported to be caused by BRBV infection in the Midwest and Southern United States. BRBV is a tick-borne virus that is widely carried by lone star ticks. It belongs to genus Thogotovirus of the Orthomyxoviridae family. Currently, there are no treatments or vaccines available for BRBV or thogotovirus infection caused diseases. In this study, we reconstituted a replicon reporter system, composed of plasmids expressing the RNA-dependent RNA polymerase (RdRP) complex (PA, PB1, and PB2), nucleocapsid (NP) protein, and a reporter gene flanked by the 3′ and 5′ untranslated region (UTR) of the envelope glycoprotein (GP) genome segment. By using the luciferase reporter, we screened a few small molecule compounds of anti-endonuclease that inhibited the nicking activity by parvovirus B19 (B19V) NS1, as well as FDA-approved drugs targeting the RdRP of influenza virus. Our results demonstrated that myricetin, an anti-B19V NS1 nicking inhibitor, efficiently inhibited the RdRP activity of BRBV and virus replication. The IC50 and EC50 of myricetin are 2.22 and 4.6 μM, respectively, in cells. Myricetin had minimal cytotoxicity in cells, and therefore the therapeutic index of the compound is high. In conclusion, the BRBV replicon system is a useful tool to study viral RNA replication and to develop antivirals, and myricetin may hold promise in treatment of BRBV infected patients.
Collapse
Affiliation(s)
- Siyuan Hao
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Kang Ning
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Xiaomei Wang
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Jianke Wang
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Fang Cheng
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Safder S Ganaie
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS, United States
| | - John E Tavis
- Department of Molecular Microbiology and Immunology, Saint Louis University St. Louis, MO, United States
| | - Jianming Qiu
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
23
|
Cellular Cleavage and Polyadenylation Specificity Factor 6 (CPSF6) Mediates Nuclear Import of Human Bocavirus 1 NP1 Protein and Modulates Viral Capsid Protein Expression. J Virol 2020; 94:JVI.01444-19. [PMID: 31666379 DOI: 10.1128/jvi.01444-19] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 10/18/2019] [Indexed: 12/13/2022] Open
Abstract
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.
Collapse
|
24
|
Di Martino B, Di Profio F, Melegari I, Marsilio F. Feline Virome-A Review of Novel Enteric Viruses Detected in Cats. Viruses 2019; 11:v11100908. [PMID: 31575055 PMCID: PMC6832874 DOI: 10.3390/v11100908] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 09/28/2019] [Accepted: 09/28/2019] [Indexed: 12/13/2022] Open
Abstract
Recent advances in the diagnostic and metagenomic investigations of the feline enteric environment have allowed the identification of several novel viruses that have been associated with gastroenteritis in cats. In the last few years, noroviruses, kobuviruses, and novel parvoviruses have been repetitively detected in diarrheic cats as alone or in mixed infections with other pathogens, raising a number of questions, with particular regards to their pathogenic attitude and clinical impact. In the present article, the current available literature on novel potential feline enteric viruses is reviewed, providing a meaningful update on the etiology, epidemiologic, pathogenetic, clinical, and diagnostic aspects of the infections caused by these pathogens.
Collapse
Affiliation(s)
- Barbara Di Martino
- Laboratory of Infectious Diseases, Faculty of Veterinary Medicine, University of Teramo, 64100 Teramo, Italy.
| | - Federica Di Profio
- Laboratory of Infectious Diseases, Faculty of Veterinary Medicine, University of Teramo, 64100 Teramo, Italy.
| | - Irene Melegari
- Laboratory of Infectious Diseases, Faculty of Veterinary Medicine, University of Teramo, 64100 Teramo, Italy.
| | - Fulvio Marsilio
- Laboratory of Infectious Diseases, Faculty of Veterinary Medicine, University of Teramo, 64100 Teramo, Italy.
| |
Collapse
|
25
|
Shi QK, Zhang JL, Gu WY, Hou LS, Yuan GF, Chen SJ, Fan JH, Zuo YZ. Seroprevalence of porcine bocavirus in pigs in north-central China using a recombinant-NP1-protein-based indirect ELISA. Arch Virol 2019; 164:2351-2354. [PMID: 31222429 DOI: 10.1007/s00705-019-04325-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Accepted: 05/24/2019] [Indexed: 10/26/2022]
Abstract
Porcine bocavirus (PBoV), which belongs the genus Bocaparvovirus, has been identified throughout the world. However, serological methods for detecting anti-PBoV antibodies are presently limited. In the present study, an indirect enzyme-linked immunosorbent assay (PBoV-rNP1 ELISA) based on a recombinant form of nucleoprotein 1 (NP1) of PBoV was established for investigating the seroprevalence of PBoV in 2025 serum specimens collected in north-central China from 2016 to 2018, and 42.3% of the samples tested positive for anti-PBoV IgG antibodies, indicating that the seroprevalence of PBoV is high in pig populations in China.
Collapse
Affiliation(s)
- Qian-Kai Shi
- College of Veterinary Medicine, Hebei Agricultural University, Lucky Street, Baoding, 071001, People's Republic of China
| | - Jian-Lou Zhang
- College of Veterinary Medicine, Hebei Agricultural University, Lucky Street, Baoding, 071001, People's Republic of China
| | - Wen-Yuan Gu
- College of Veterinary Medicine, Hebei Agricultural University, Lucky Street, Baoding, 071001, People's Republic of China.,Animal Diseases Control Center of Hebei, Shijiazhuang, 050053, People's Republic of China
| | - Lin-Shan Hou
- College of Veterinary Medicine, Hebei Agricultural University, Lucky Street, Baoding, 071001, People's Republic of China
| | - Guang-Fu Yuan
- College of Veterinary Medicine, Hebei Agricultural University, Lucky Street, Baoding, 071001, People's Republic of China
| | - Shao-Jie Chen
- College of Veterinary Medicine, Hebei Agricultural University, Lucky Street, Baoding, 071001, People's Republic of China
| | - Jing-Hui Fan
- College of Veterinary Medicine, Hebei Agricultural University, Lucky Street, Baoding, 071001, People's Republic of China.
| | - Yu-Zhu Zuo
- College of Veterinary Medicine, Hebei Agricultural University, Lucky Street, Baoding, 071001, People's Republic of China.
| |
Collapse
|
26
|
Establishment of a rescue system for porcine parvovirus using a seamless cloning method. Arch Virol 2019; 164:1459-1467. [PMID: 30880347 DOI: 10.1007/s00705-019-04209-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 03/02/2019] [Indexed: 11/27/2022]
Abstract
In this study, we describe a novel and rapid method for the construction of a full-length infectious clone (pPPV). The constructed clone contained an engineered EcoRv site that served as a genetic marker and was shown to be infectious when transfected into a monolayer of PK-15 cells. The rescued virus (rPPV) of the infectious clone was found to be indistinguishable from wild-type virus BQ in terms of its biological properties. The generation of this PPV infectious clone provides a potentially powerful tool with which to elucidate the molecular pathogenesis of PPV.
Collapse
|
27
|
Ding Y, Li N, Sun J, Zhang L, Guo J, Hao X, Sun Y. Oxymatrine Inhibits Bocavirus MVC Replication, Reduces Viral Gene Expression and Decreases Apoptosis Induced by Viral Infection. Virol Sin 2019; 34:78-87. [PMID: 30805776 DOI: 10.1007/s12250-019-00088-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 12/13/2018] [Indexed: 12/20/2022] Open
Abstract
Oxymatrine (OMT), as the main active component of Sophoraflavescens, exhibits a variety of pharmacological properties, including anti-oxidative, anti-inflammatory, anti-tumor, and anti-viral activities, and currently is extensively employed to treat viral hepatitis; however, its effects on parvovirus infection have yet to be reported. In the present study, we investigated the effects of OMT on cell viability, virus DNA replication, viral gene expression, cell cycle, and apoptosis in Walter Reed canine cells/3873D infected with minute virus of canines (MVC). OMT, at concentrations below 4 mmol/L(no cellular toxicity), was found to inhibit MVC DNA replication and reduce viral gene expression at both mRNA and protein levels, which was associated with the inhibition of cell cycle S-phase arrest in early-stage of MVC infection. Furthermore, OMT significantly increased cell viability, decreased MVC-infected cell apoptosis, and reduced the expression of activated caspase 3. Our results suggest that OMT has potential application in combating parvovirus infection.
Collapse
Affiliation(s)
- Yanqin Ding
- Department of Biochemistry and Molecular Biology, Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Science, Ningxia Medical University, Yinchuan, 750004, China
| | - Na Li
- Department of Biochemistry and Molecular Biology, Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Science, Ningxia Medical University, Yinchuan, 750004, China
| | - Jinhan Sun
- School of Clinical Medicine, Hainan Medical University, Haikou, 571199, China
| | - Linran Zhang
- Department of Biochemistry and Molecular Biology, Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Science, Ningxia Medical University, Yinchuan, 750004, China
| | - Jianhui Guo
- Department of Biochemistry and Molecular Biology, Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Science, Ningxia Medical University, Yinchuan, 750004, China
| | - Xueqi Hao
- Department of Respiratory and Critical Care, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, 750004, China
| | - Yuning Sun
- Department of Biochemistry and Molecular Biology, Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Science, Ningxia Medical University, Yinchuan, 750004, China.
| |
Collapse
|
28
|
Yan Z, Zou W, Feng Z, Shen W, Park SY, Deng X, Qiu J, Engelhardt JF. Establishment of a High-Yield Recombinant Adeno-Associated Virus/Human Bocavirus Vector Production System Independent of Bocavirus Nonstructural Proteins. Hum Gene Ther 2019; 30:556-570. [PMID: 30398383 DOI: 10.1089/hum.2018.173] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The genome of recombinant adeno-associated virus 2 (rAAV2) remains a promising candidate for gene therapy for cystic fibrosis (CF) lung disease, but due to limitations in the packaging capacity and the tropism of this virus with respect to the airways, strategies have evolved for packaging an rAAV2 genome (up to 5.8 kb) into the capsid of human bocavirus 1 (HBoV1) to produce a chimeric rAAV2/HBoV1 vector. Although a replication-incompetent HBoV1 genome has been established as a trans helper for capsid complementation, this system remains suboptimal with respect to virion yield. Here, a streamlined production system is described based on knowledge of the involvement of HBoV1 nonstructural (NS) proteins NS1, NS2, NS3, NS4, and NP1 in the process of virion production. The analyses reveal that NS1 and NS2 negatively impact virion production, NP1 is required to prevent premature termination of transcription of the cap mRNA from the native genome, and silent mutations within the polyadenylation sites of the cap coding sequence can eliminate this requirement for NP1. It is further shown that preventing the expression of all NS proteins significantly increases virion yield. Whereas the expression of capsid proteins VP1, VP2, and VP3 from a codon-optimized cap mRNA was highly efficient, optimal virion assembly, and thus potency, required enhanced VP1 expression, entailing a separate VP1 expression cassette. The final NS protein-free production system uses three-plasmid co-transfection of HEK293 cells, with one trans helper plasmid encoding VP1 and the AAV2 Rep proteins, and another encoding VP2-3 and components from adenovirus. This system yielded >16-fold more virions than the prototypic system, without reducing transduction potency. This increase in virion production is expected to facilitate greatly both research on the biology of rAAV2/HBoV1 and preclinical studies testing the effectiveness of this vector for gene therapy of CF lung disease in large animal models.
Collapse
Affiliation(s)
- Ziying Yan
- 1 Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa.,2 Center for Gene Therapy, University of Iowa, Iowa City, Iowa
| | - Wei Zou
- 3 Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas
| | - Zehua Feng
- 1 Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa
| | - Weiran Shen
- 3 Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas
| | - Soo Yeun Park
- 1 Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa
| | - Xuefeng Deng
- 3 Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas
| | - Jianming Qiu
- 3 Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas
| | - John F Engelhardt
- 1 Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa.,2 Center for Gene Therapy, University of Iowa, Iowa City, Iowa
| |
Collapse
|
29
|
Zou W, Xiong M, Deng X, Engelhardt JF, Yan Z, Qiu J. A Comprehensive RNA-seq Analysis of Human Bocavirus 1 Transcripts in Infected Human Airway Epithelium. Viruses 2019; 11:v11010033. [PMID: 30621044 PMCID: PMC6357044 DOI: 10.3390/v11010033] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 12/30/2018] [Accepted: 01/02/2019] [Indexed: 02/07/2023] Open
Abstract
Human bocavirus 1 (HBoV1) infects well-differentiated (polarized) human airway epithelium (HAE) cultured at an air-liquid interface (ALI). In the present study, we applied next-generation RNA sequencing to investigate the genome-wide transcription profile of HBoV1, including viral mRNA and small RNA transcripts, in HBoV1-infected HAE cells. We identified novel transcription start and termination sites and confirmed the previously identified splicing events. Importantly, an additional proximal polyadenylation site (pA)p2 and a new distal polyadenylation site (pA)dREH lying on the right-hand hairpin (REH) of the HBoV1 genome were identified in processing viral pre-mRNA. Of note, all viral nonstructural proteins-encoding mRNA transcripts use both the proximal polyadenylation sites [(pA)p1 and (pA)p2] and distal polyadenylation sites [(pA)d1 and (pA)dREH] for termination. However, capsid proteins-encoding transcripts only use the distal polyadenylation sites. While the (pA)p1 and (pA)p2 sites were utilized at roughly equal efficiency for proximal polyadenylation of HBoV1 mRNA transcripts, the (pA)d1 site was more preferred for distal polyadenylation. Additionally, small RNA-seq analysis confirmed there is only one viral noncoding RNA (BocaSR) transcribed from nt 5199⁻5340 of the HBoV1 genome. Thus, our study provides a systematic and unbiased transcription profile, including both mRNA and small RNA transcripts, of HBoV1 in HBoV1-infected HAE-ALI cultures.
Collapse
Affiliation(s)
- Wei Zou
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Min Xiong
- The Children's Mercy Hospital, School of Medicine, University of Missouri Kansas City, Kansas City, MO 64108, USA.
| | - Xuefeng Deng
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - John F Engelhardt
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242, USA.
| | - Ziying Yan
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52242, USA.
| | - Jianming Qiu
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| |
Collapse
|
30
|
Minute Virus of Canines NP1 Protein Interacts with the Cellular Factor CPSF6 To Regulate Viral Alternative RNA Processing. J Virol 2019; 93:JVI.01530-18. [PMID: 30355695 DOI: 10.1128/jvi.01530-18] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 10/21/2018] [Indexed: 12/21/2022] Open
Abstract
The NP1 protein of minute virus of canines (MVC) governs production of the viral capsid proteins via its role in pre-mRNA processing. NP1 suppresses polyadenylation and cleavage at its internal site, termed the proximal polyadenylation (pA)p site, to allow accumulation of RNAs that extend into the capsid gene, and it enhances splicing of the upstream adjacent third intron, which is necessary to properly enter the capsid protein open reading frame. We find the (pA)p region to be complex. It contains redundant classical cis-acting signals necessary for the cleavage and polyadenylation reaction and splicing of the adjacent upstream third intron, as well as regions outside the classical motifs that are necessary for responding to NP1. NP1, but not processing mutants of NP1, bound to MVC RNA directly. The cellular RNA processing factor CPSF6 interacted with NP1 in transfected cells and participated with NP1 to modulate its effects. These experiments further characterize the role of NP1 in parvovirus gene expression.IMPORTANCE The Parvovirinae are small nonenveloped icosahedral viruses that are important pathogens in many animal species, including humans. Unlike other parvoviruses, the bocavirus genus controls expression of its capsid proteins via alternative RNA processing, by both suppressing polyadenylation at an internal site, termed the proximal polyadenylation (pA)p site, and by facilitating splicing of an upstream adjacent intron. This regulation is mediated by a small genus-specific protein, NP1. Understanding the cis-acting targets of NP1, as well as the cellular factors with which it interacts, is necessary to more clearly understand this unique mode of parvovirus gene expression.
Collapse
|
31
|
A novel recombinant genome of minute virus of canines in China. Arch Virol 2018; 164:861-865. [PMID: 30465112 DOI: 10.1007/s00705-018-4104-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 11/04/2018] [Indexed: 01/12/2023]
Abstract
In this study, we determined almost all of the genome sequence of minute virus of canines (MVC) strain CDK47/2017 and performed phylogenetic analysis with this isolate and other MVC isolates. The genome of CDK47/2017 has the following characteristics: 1) The amino acid sequence of the NS1 protein is similar to that of the novel strain 15D009/KT241026.1, which has 17 identical amino acid changes and two identical amino acid insertions compared with other known MVC strains. These two strains clustered in a unique branch in an NSI-based phylogenetic tree. 2) Phylogenetic analysis based on the NP1 protein showed that strain CDK47/2017clustered in an independent branch with strains 15D009/KT241026.1 and HM-6/AB158475.1, both of which has 10 identical amino acid changes compared with other known MVC strains. 3) Eight unique amino acid substitutions of the CDK47/2017 capsid protein resulted in it forming a unique branch in the phylogenetic tree. 4) Recombination events were identified between the 3' end of the NS1 gene and 5' end of NP1 gene. Together, these characteristics suggest that strain CDK47/2017 represents a novel MVC strain that is distinct from all known MVC strains with sequences in the GenBank database. This contributes to a greater understanding of the genetic evolution of MVC.
Collapse
|
32
|
Fan Y, Sanyal S, Bruzzone R. Breaking Bad: How Viruses Subvert the Cell Cycle. Front Cell Infect Microbiol 2018; 8:396. [PMID: 30510918 PMCID: PMC6252338 DOI: 10.3389/fcimb.2018.00396] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 10/22/2018] [Indexed: 01/10/2023] Open
Abstract
Interactions between the host and viruses during the course of their co-evolution have not only shaped cellular function and the immune system, but also the counter measures employed by viruses. Relatively small genomes and high replication rates allow viruses to accumulate mutations and continuously present the host with new challenges. It is therefore, no surprise that they either escape detection or modulate host physiology, often by redirecting normal cellular pathways to their own advantage. Viruses utilize a diverse array of strategies and molecular targets to subvert host cellular processes, while evading detection. These include cell-cycle regulation, major histocompatibility complex-restricted antigen presentation, intracellular protein transport, apoptosis, cytokine-mediated signaling, and humoral immune responses. Moreover, viruses routinely manipulate the host cell cycle to create a favorable environment for replication, largely by deregulating cell cycle checkpoints. This review focuses on our current understanding of the molecular aspects of cell cycle regulation that are often targeted by viruses. Further study of their interactions should provide fundamental insights into cell cycle regulation and improve our ability to exploit these viruses.
Collapse
Affiliation(s)
- Ying Fan
- HKU-Pasteur Research Pole, LKS Faculty of Medicine, School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong.,MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Sumana Sanyal
- HKU-Pasteur Research Pole, LKS Faculty of Medicine, School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong.,LKS Faculty of Medicine, School of Biomedical Sciences, The University of Hong Kong, Hong Kong, Hong Kong
| | - Roberto Bruzzone
- HKU-Pasteur Research Pole, LKS Faculty of Medicine, School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong.,Department of Cell Biology and Infection, Institut Pasteur, Paris, France
| |
Collapse
|
33
|
The Human Bocavirus 1 NP1 Protein Is a Multifunctional Regulator of Viral RNA Processing. J Virol 2018; 92:JVI.01187-18. [PMID: 30135129 DOI: 10.1128/jvi.01187-18] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 08/16/2018] [Indexed: 12/12/2022] Open
Abstract
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.
Collapse
|
34
|
Xiong YQ, You FF, Chen XJ, Chen YX, Wen YQ, Chen Q. Detection and phylogenetic analysis of porcine bocaviruses carried by murine rodents and house shrews in China. Transbound Emerg Dis 2018; 66:259-267. [PMID: 30179306 DOI: 10.1111/tbed.13011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 03/25/2018] [Accepted: 07/07/2018] [Indexed: 01/29/2023]
Abstract
Bocaparvovirus infections of humans and both wild and domestic animals have been widely reported around the world. In this study, we detected and genetically characterized porcine bocavirus (PBoV) carried by murine rodents (Rattus norvegicus, Rattus tanezumi, and Rattus losea) and house shrews (Suncus murinus) in China. Between May 2015 and May 2017, 496 murine rodents and 23 house shrews were captured in four Chinese provinces. Nested polymerase chain reaction was used to investigate the prevalence of PBoV in throat swab, faecal and serum samples. A total of 7.5% (39/519) throat swab samples, 60.5% (309/511) faecal samples, and 22.9% (52/227) serum samples were PBoV-positive. The prevalence among R. norvegicus and R. tanezumi was higher than that among R. losea and house shrews. PBoV-positive samples were found in all four provinces. Phylogenetic analysis based on partial viral capsid protein 1/2 (VP1/VP2) showed that sequences obtained in this study formed a novel group (PBoV G4). In addition, five near full-length PBoV genomes (4,715-4,798 nt) were acquired. These genomes encoded two non-structural proteins, NS1 (1,908 nt in four genomes and 1,923 nt in the remaining genome) and NP1 (600 nt), and the structural proteins, VP1/VP2 (1,851 nt). Phylogenetic analysis showed that PBoV G4 is distinct from rodent, human, and other bocaviruses. In conclusion, PBoV G4 prevalence was high among two common murine rodents in China, and the pathogenecity of PBoV G4 need to be further clarified.
Collapse
Affiliation(s)
- Yi-Quan Xiong
- Department of Epidemiology, School of Public Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, Southern Medical University, Guangzhou, China
| | - Fang-Fei You
- Department of Epidemiology, School of Public Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, Southern Medical University, Guangzhou, China
| | - Xue-Jiao Chen
- Department of Epidemiology, School of Public Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, Southern Medical University, Guangzhou, China
| | - Yan-Xia Chen
- Department of Epidemiology, School of Public Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, Southern Medical University, Guangzhou, China
| | - Yu-Qi Wen
- Department of Epidemiology, School of Public Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, Southern Medical University, Guangzhou, China
| | - Qing Chen
- Department of Epidemiology, School of Public Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, Southern Medical University, Guangzhou, China
| |
Collapse
|
35
|
Yi S, Niu J, Wang H, Dong G, Zhao Y, Dong H, Guo Y, Wang K, Hu G. Detection and genetic characterization of feline bocavirus in Northeast China. Virol J 2018; 15:125. [PMID: 30089507 PMCID: PMC6083571 DOI: 10.1186/s12985-018-1034-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 07/30/2018] [Indexed: 01/15/2023] Open
Abstract
Background Bocaviruses have been reported to cause respiratory tract infection and gastroenteritis in most animal species. In cats, different genotype bocaviruses have been identified in USA, Japan, Hong Kong and Portugal. However, the clear relationship between the clinical symptoms and FBoV infection is unknown, and the prevalence of FBoV and the distribution of FBoV genotypes in China are still unclear. Results In this study, 197 fecal samples from cats with diarrhea (n = 105) and normal cats (n = 92) were collected in different regions between January 2016 and November 2017 and investigated using PCR targeting different FBoV genotypes. Screening results showed that 51 of 197 samples (25.9%) were positive for FBoV, and a higher positive rate was observed in cats with diarrhea (33.3%, 35/105) than in normal cats (17.4%, 16/92). Of these FBoV-positive samples, 35 were identified as FBoV-1, 12 as FBoV-2 and 4 as coinfection of FBoV-1 and FBoV-2. A phylogenetic analysis based on partial NS1 gene indicated that 24 sequences from randomly selected FBoV-positive samples were divided into 2 different FBoV groups: FBoV-1 and FBoV-2. Furthermore, 6 strains were randomly selected, and the complete genome was sequenced and analyzed. These strains exhibited the typical genome organization of bocavirus and were closely related to FBoV. Two FBoV-2 identified strains shared high homologies with FBoV-2 reference strains based on the complete genome and entire encoding gene, but lower identities were exhibited in the NP1 and VP1 regions for the other 4 FBoV-1 identified strains compared with FBoV-1 reference strains. Conclusion These findings demonstrate that genetically diverse FBoV-1 and FBoV-2 widely circulate in cats in Northeast China and that FBoV-1 is more prevalent. The high prevalence of FBoV in cats with diarrhea symptoms suggests that FBoV infection may be associated with diarrhea in cats. Electronic supplementary material The online version of this article (10.1186/s12985-018-1034-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shushuai Yi
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, Jilin Province, China
| | - Jiangting Niu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, Jilin Province, China
| | - Hualei Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Military Veterinary Research Institute, Academy of Military Medical Sciences, Changchun, 130122, China
| | - Guoying Dong
- College of Global Change and Earth System Science, Beijing Normal University, Haidian, Beijing, 100875, China
| | - Yanli Zhao
- Library, Jilin Agricultural University, Changchun, 130118, Jilin Province, China
| | - Hao Dong
- College of Life Science and Technology, Jilin Agricultural University, Changchun, 130118, Jilin Province, China.
| | - Yanbing Guo
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, Jilin Province, China.,Jilin Institute of Animal Husbandry and Veterinary Science, Changchun, 130062, Jilin Province, China
| | - Kai Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, Jilin Province, China
| | - Guixue Hu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, Jilin Province, China.
| |
Collapse
|
36
|
de Souza WM, Dennis T, Fumagalli MJ, Araujo J, Sabino-Santos G, Maia FGM, Acrani GO, Carrasco ADOT, Romeiro MF, Modha S, Vieira LC, Ometto T, Queiroz LH, Durigon EL, Nunes MRT, Figueiredo LTM, Gifford RJ. Novel Parvoviruses from Wild and Domestic Animals in Brazil Provide New Insights into Parvovirus Distribution and Diversity. Viruses 2018; 10:E143. [PMID: 29565808 PMCID: PMC5923437 DOI: 10.3390/v10040143] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 03/17/2018] [Accepted: 03/20/2018] [Indexed: 12/11/2022] Open
Abstract
Parvoviruses (family Parvoviridae) are small, single-stranded DNA viruses. Many parvoviral pathogens of medical, veterinary and ecological importance have been identified. In this study, we used high-throughput sequencing (HTS) to investigate the diversity of parvoviruses infecting wild and domestic animals in Brazil. We identified 21 parvovirus sequences (including twelve nearly complete genomes and nine partial genomes) in samples derived from rodents, bats, opossums, birds and cattle in Pernambuco, São Paulo, Paraná and Rio Grande do Sul states. These sequences were investigated using phylogenetic and distance-based approaches and were thereby classified into eight parvovirus species (six of which have not been described previously), representing six distinct genera in the subfamily Parvovirinae. Our findings extend the known biogeographic range of previously characterized parvovirus species and the known host range of three parvovirus genera (Dependovirus, Aveparvovirus and Tetraparvovirus). Moreover, our investigation provides a window into the ecological dynamics of parvovirus infections in vertebrates, revealing that many parvovirus genera contain well-defined sub-lineages that circulate widely throughout the world within particular taxonomic groups of hosts.
Collapse
Affiliation(s)
- William Marciel de Souza
- Virology Research Center, Ribeirão Preto Medical School, University of São Paulo,14049-900 Ribeirão Preto, SP, Brazil.
- MRC-University of Glasgow Centre for Virus Research, Glasgow G61 1QH, UK.
| | - Tristan Dennis
- MRC-University of Glasgow Centre for Virus Research, Glasgow G61 1QH, UK.
| | - Marcílio Jorge Fumagalli
- Virology Research Center, Ribeirão Preto Medical School, University of São Paulo,14049-900 Ribeirão Preto, SP, Brazil.
| | - Jansen Araujo
- Institute of Biomedical Sciences, University of São Paulo, 05508-900 São Paulo, SP, Brazil.
| | - Gilberto Sabino-Santos
- Virology Research Center, Ribeirão Preto Medical School, University of São Paulo,14049-900 Ribeirão Preto, SP, Brazil.
| | - Felipe Gonçalves Motta Maia
- Virology Research Center, Ribeirão Preto Medical School, University of São Paulo,14049-900 Ribeirão Preto, SP, Brazil.
- Institute of Biomedical Sciences, University of São Paulo, 05508-900 São Paulo, SP, Brazil.
| | | | | | - Marilia Farignoli Romeiro
- Virology Research Center, Ribeirão Preto Medical School, University of São Paulo,14049-900 Ribeirão Preto, SP, Brazil.
| | - Sejal Modha
- MRC-University of Glasgow Centre for Virus Research, Glasgow G61 1QH, UK.
| | - Luiz Carlos Vieira
- Virology Research Center, Ribeirão Preto Medical School, University of São Paulo,14049-900 Ribeirão Preto, SP, Brazil.
| | - Tatiana Ometto
- Institute of Biomedical Sciences, University of São Paulo, 05508-900 São Paulo, SP, Brazil.
| | - Luzia Helena Queiroz
- Faculty of Veterinary Medicine, São Paulo State University, Araçatuba, SP 16050-680, Brazil.
| | - Edison Luiz Durigon
- Institute of Biomedical Sciences, University of São Paulo, 05508-900 São Paulo, SP, Brazil.
| | - Márcio Roberto Teixeira Nunes
- Center for Technological Innovations, Evandro Chagas Institute, Ministry of Health, Ananindeua, Pará 67030-000, Pará, Brazil.
| | - Luiz Tadeu Moraes Figueiredo
- Virology Research Center, Ribeirão Preto Medical School, University of São Paulo,14049-900 Ribeirão Preto, SP, Brazil.
| | | |
Collapse
|
37
|
Piewbang C, Jo WK, Puff C, Ludlow M, van der Vries E, Banlunara W, Rungsipipat A, Kruppa J, Jung K, Techangamsuwan S, Baumgärtner W, Osterhaus ADME. Canine Bocavirus Type 2 Infection Associated With Intestinal Lesions. Vet Pathol 2018; 55:434-441. [PMID: 29421972 DOI: 10.1177/0300985818755253] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Bocaviruses are small nonenveloped DNA viruses belonging to the Bocaparvovirus genus of the Parvoviridae family and have been linked to both respiratory and enteric disease in humans and animals. To date, 3 bocaviruses, canine bocaviruses 1 to 3 (CBoV-1-3), have been shown to affect dogs with different disease manifestations reported for infected animals. We used next-generation sequencing to identify a novel strain of canine CBoV-2 (CBoV TH-2016) in a litter of puppies that died in Thailand from acute dyspnea and hemoptysis, for which no causal pathogen could be identified in routine assays. Analysis of the complete coding sequences of CBoV TH-2016 showed that this virus was most closely related to a strain previously identified in South Korea (isolate 14D193), with evidence of genetic recombination in the VP2 gene with related strains from South Korea and Hong Kong. Use of quantitative polymerase chain reaction showed the presence of CBoV TH-2016 in several tissues, suggesting hematogenous virus spread, while only intestinal tissue was found to be positive by in situ hybridization and electron microscopy. Histologic small intestinal lesions associated with CBoV TH-2016 infection were eosinophilic intranuclear inclusion bodies within villous enterocytes without villous atrophy or fusion, similar to those previously considered pathognomonic for CBoV-1 infection. Therefore, this study provides novel insights in the pathogenicity of canine bocavirus infections and suggests that a novel recombinant CBoV-2 may result in atypical findings of CBoV infection. Although the specific cause of death of these puppies remained undetermined, a contributory role of enteric CBoV TH-2016 infection is possible.
Collapse
Affiliation(s)
- Chutchai Piewbang
- 1 Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Pathumwan, Bangkok, Thailand.,2 Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Hannover, Germany
| | - Wendy K Jo
- 2 Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Hannover, Germany
| | - Christina Puff
- 3 Department of Pathology, University of Veterinary Medicine, Hannover, Germany
| | - Martin Ludlow
- 2 Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Hannover, Germany
| | - Erhard van der Vries
- 2 Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Hannover, Germany
| | - Wijit Banlunara
- 1 Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Pathumwan, Bangkok, Thailand
| | - Anudep Rungsipipat
- 1 Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Pathumwan, Bangkok, Thailand
| | - Jochen Kruppa
- 4 Institute for Animal Breeding and Genetics, University of Veterinary Medicine, Hannover, Germany
| | - Klaus Jung
- 4 Institute for Animal Breeding and Genetics, University of Veterinary Medicine, Hannover, Germany
| | - Somporn Techangamsuwan
- 1 Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Pathumwan, Bangkok, Thailand.,5 STAR Diagnosis and Monitoring of Animal Pathogen, Faculty of Veterinary Science, Chulalongkorn University, Pathumwan, Bangkok, Thailand
| | | | - Albert D M E Osterhaus
- 2 Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Hannover, Germany
| |
Collapse
|
38
|
Genetic analysis of three porcine bocaparvoviruses and identification of a natural recombinant breakpoint in NS1. Arch Virol 2017; 163:707-712. [PMID: 29170830 DOI: 10.1007/s00705-017-3606-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 09/13/2017] [Indexed: 10/18/2022]
Abstract
In this study, we obtained the whole genomes of three porcine bocaparvovirus (PBoV) strains (GD6, GD10, and GD23) by polymerase chain reaction. Sequence analysis showed that all three field strains belonged to PBoV group 3 (G3). The phylogenetic trees based on NS1, NP1, and VP1 differed to the extent that these PBoVs were potentially more closely related to bocaparvoviruses known to infect other animals than to other PBoVs. GD6, GD10, and GD23 all included the conserved sequences YLGPF and HDXXY, with known phospholipase A2 activity. Using recombination-detection software we identified a natural recombinant breakpoint in the NS1 region of PBoV G3. The results of this study will further the epidemiological characterization of PBoVs.
Collapse
|
39
|
Human Bocavirus 1 Is a Novel Helper for Adeno-associated Virus Replication. J Virol 2017; 91:JVI.00710-17. [PMID: 28659483 DOI: 10.1128/jvi.00710-17] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 06/22/2017] [Indexed: 12/11/2022] Open
Abstract
Human bocavirus 1 (HBoV1) is an autonomous parvovirus that infects well-differentiated primary human airway epithelia (HAE) in vitro In human embryonic kidney HEK293 cells, the transfection of a duplex HBoV1 genome initiates viral DNA replication and produces progeny virions that are infectious in HAE. HBoV1 takes advantage of signaling pathways in the DNA damage response for efficient genome amplification in both well-differentiated (nondividing) HAE and dividing HEK293 cells. On the other hand, adeno-associated virus 2 (AAV2) is a helper-dependent dependoparvovirus, and productive AAV2 replication requires coinfection with a helper virus (e.g., adenovirus or herpesvirus) or treatment with genotoxic agents. Here, we report that HBoV1 is a novel helper virus for AAV2 replication. Coinfection by HBoV1 and AAV2 rescued AAV2 replication in HAE cells. The helper function of HBoV1 for AAV2 is not limited to HAE cells but also includes HEK293 and HeLa cells. Importantly, the helper function of HBoV1 for AAV2 relies on neither HBoV1 replication nor the DNA damage response. Following transfection of HEK293 cells, the minimal requirements for the replication of the AAV2 duplex DNA genome and the production of progeny virions included the HBoV1 NP1 and NS4 proteins and a newly identified viral long noncoding RNA (BocaSR). However, following infection of HEK293 and HeLa cells with AAV2 virions, HBoV1 NS2 (but not NS4), NP1, and BocaSR were required for AAV2 DNA replication and progeny virion formation. These new methods for packaging the AAV2 genome may be useful for generating recombinant AAV-packaging cell lines and the directed evolution of AAV capsids.IMPORTANCE We first report that an autonomous parvovirus, HBoV1, helps the replication of a dependoparvovirus, AAV2, in differentiated human airway epithelia. We identified the minimal sets of HBoV1 genes required to facilitate the replication of the AAV2 duplex genome and for AAV2 infection. Notably, together with the expression of the NP1 and BocaSR genes, HBoV1 NS2 is required for the productive infection of HEK293 and HeLa cells by AAV2, whereas NS4 is sufficient for viral DNA replication of an AAV2 duplex genome. The identification of HBoV1 as a helper virus for AAV2 replication has implications for the improvement of recombinant AAV production in HEK293 cells and cell types that do not express the adenovirus E1 gene as well as for the rescue of wild-type AAV genomes from tissues during directed evolution in the absence of wild-type adenovirus. A further understanding of the mechanism underlying HBoV1 helper-dependent AAV2 replication may also provide insights into its functions in HBoV1 replication.
Collapse
|
40
|
Zhang J, Bai Y, Zhu B, Hao S, Chen Z, Wang H, Guan W. Mutations in the C-terminus of HBoV NS1 affect the function of NP1. Sci Rep 2017; 7:7407. [PMID: 28785044 PMCID: PMC5547040 DOI: 10.1038/s41598-017-06513-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 06/13/2017] [Indexed: 02/07/2023] Open
Abstract
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.
Collapse
Affiliation(s)
- Junmei Zhang
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, 430071, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yan Bai
- Pediatric department of Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bing Zhu
- Center Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou, 510120, P. R. China
| | - Sujuan Hao
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, 430071, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhen Chen
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, 430071, China
| | - Hanzhong Wang
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, 430071, China
| | - Wuxiang Guan
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, 430071, China.
| |
Collapse
|
41
|
Woo PCY, Lau SKP, Tsoi HW, Patteril NG, Yeung HC, Joseph S, Wong EYM, Muhammed R, Chow FWN, Wernery U, Yuen KY. Two novel dromedary camel bocaparvoviruses from dromedaries in the Middle East with unique genomic features. J Gen Virol 2017; 98:1349-1359. [PMID: 28613145 DOI: 10.1099/jgv.0.000775] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The recent emergence of Middle East respiratory syndrome (MERS) coronavirus and its discovery from dromedary camels has boosted interest in the search for novel viruses in dromedaries. While bocaparvoviruses are known to infect various animals, it was not known that they exist in dromedaries. In this study, we describe the discovery of two novel dromedary camel bocaparvoviruses (DBoVs), DBoV1 and DBoV2, from dromedary faecal samples in Dubai. Among 667 adult dromedaries and 72 dromedary calves, 13.9 % of adult dromedaries and 33.3 % of dromedary calves were positive for DBoV1, while 7.0 % of adult dromedaries and 25.0 % of dromedary calves were positive for DBoV2, as determined by PCR. Sequencing of 21 DBoV1 and 18 DBoV2 genomes and phylogenetic analysis showed that DBoV1 and DBoV2 formed two distinct clusters, with only 32.6-36.3 % amino acid identities between the DBoV1 and DBoV2 strains. Quasispecies were detected in both DBoVs. The amino acid sequences of the NS1 proteins of all the DBoV1 and DBoV2 strains showed <85 % identity to those of all the other bocaparvoviruses, indicating that DBoV1 and DBoV2 are two bocaparvovirus species according to the ICTV criteria. Although the typical genome structure of NS1-NP1-VP1/VP2 was observed in DBoV1 and DBoV2, no phospholipase A2 motif and associated calcium binding site were observed in the predicted VP1 sequences for any of the 18 sequenced DBoV2, and no start codons were found for their VP1. For all 18 DBoV2 genomes, an AT-rich region of variable length and composition was present downstream to NP1. Further studies will be crucial to understand the pathogenic potential of DBoVs in this unique group of animals.
Collapse
Affiliation(s)
- Patrick C Y Woo
- Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong SAR.,State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong SAR.,Department of Microbiology, The University of Hong Kong, Hong Kong SAR.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The University of Hong Kong, Hong Kong SAR.,Carol Yu Centre for Infection, The University of Hong Kong, Hong Kong SAR
| | - Susanna K P Lau
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The University of Hong Kong, Hong Kong SAR.,Department of Microbiology, The University of Hong Kong, Hong Kong SAR.,Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong SAR.,Carol Yu Centre for Infection, The University of Hong Kong, Hong Kong SAR.,State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong SAR
| | - Hoi-Wah Tsoi
- Department of Microbiology, The University of Hong Kong, Hong Kong SAR
| | | | - Hazel C Yeung
- Department of Microbiology, The University of Hong Kong, Hong Kong SAR
| | | | - Emily Y M Wong
- Department of Microbiology, The University of Hong Kong, Hong Kong SAR
| | | | - Franklin W N Chow
- Department of Microbiology, The University of Hong Kong, Hong Kong SAR
| | | | - Kwok-Yung Yuen
- Carol Yu Centre for Infection, The University of Hong Kong, Hong Kong SAR.,State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong SAR.,Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong SAR.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The University of Hong Kong, Hong Kong SAR.,Department of Microbiology, The University of Hong Kong, Hong Kong SAR
| |
Collapse
|
42
|
Conceição‐Neto N, Godinho R, Álvares F, Yinda CK, Deboutte W, Zeller M, Laenen L, Heylen E, Roque S, Petrucci‐Fonseca F, Santos N, Van Ranst M, Mesquita JR, Matthijnssens J. Viral gut metagenomics of sympatric wild and domestic canids, and monitoring of viruses: Insights from an endangered wolf population. Ecol Evol 2017; 7:4135-4146. [PMID: 28649326 PMCID: PMC5478050 DOI: 10.1002/ece3.2991] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 03/15/2017] [Accepted: 03/21/2017] [Indexed: 12/13/2022] Open
Abstract
Animal host-microbe interactions are a relevant concern for wildlife conservation, particularly regarding generalist pathogens, where domestic host species can play a role in the transmission of infectious agents, such as viruses, to wild animals. Knowledge on viral circulation in wild host species is still scarce and can be improved by the recent advent of modern molecular approaches. We aimed to characterize the fecal virome and identify viruses of potential conservation relevance of diarrheic free-ranging wolves and sympatric domestic dogs from Central Portugal, where a small and threatened wolf population persists in a highly anthropogenically modified landscape. Using viral metagenomics, we screened diarrheic stools collected from wolves (n = 8), feral dogs (n = 4), and pet dogs (n = 6), all collected within wolf range. We detected novel highly divergent viruses as well as known viral pathogens with established effects on population dynamics, including canine distemper virus, a novel bocavirus, and canine minute virus. Furthermore, we performed a 4-year survey for the six wolf packs comprising this endangered wolf population, screening 93 fecal samples from 36 genetically identified wolves for canine distemper virus and the novel bocavirus, previously identified using our metagenomics approach. Our novel approach using metagenomics for viral screening in noninvasive samples of wolves and dogs has profound implications on the knowledge of both virology and wildlife diseases, establishing a complementary tool to traditional screening methods for the conservation of threatened species.
Collapse
Affiliation(s)
- Nádia Conceição‐Neto
- Department of Microbiology and ImmunologyLaboratory of Viral MetagenomicsRega Institute for Medical ResearchKU Leuven – University of LeuvenLeuvenBelgium
- Department of Microbiology and ImmunologyLaboratory of Clinical VirologyRega Institute for Medical ResearchKU Leuven – University of LeuvenLeuvenBelgium
| | - Raquel Godinho
- CIBIO/InBIOCentro de Investigação em Biodiversidade e Recursos GenéticosUniversidade do PortoVairãoPortugal
- Departamento de BiologiaFaculdade de CiênciasUniversidade do PortoPortoPortugal
| | - Francisco Álvares
- CIBIO/InBIOCentro de Investigação em Biodiversidade e Recursos GenéticosUniversidade do PortoVairãoPortugal
| | - Claude K. Yinda
- Department of Microbiology and ImmunologyLaboratory of Viral MetagenomicsRega Institute for Medical ResearchKU Leuven – University of LeuvenLeuvenBelgium
- Department of Microbiology and ImmunologyLaboratory of Clinical VirologyRega Institute for Medical ResearchKU Leuven – University of LeuvenLeuvenBelgium
| | - Ward Deboutte
- Department of Microbiology and ImmunologyLaboratory of Viral MetagenomicsRega Institute for Medical ResearchKU Leuven – University of LeuvenLeuvenBelgium
| | - Mark Zeller
- Department of Microbiology and ImmunologyLaboratory of Viral MetagenomicsRega Institute for Medical ResearchKU Leuven – University of LeuvenLeuvenBelgium
| | - Lies Laenen
- Department of Microbiology and ImmunologyLaboratory of Clinical VirologyRega Institute for Medical ResearchKU Leuven – University of LeuvenLeuvenBelgium
| | - Elisabeth Heylen
- Department of Microbiology and ImmunologyLaboratory of Viral MetagenomicsRega Institute for Medical ResearchKU Leuven – University of LeuvenLeuvenBelgium
| | - Sara Roque
- cE3c, Centre for Ecology, Evolution and Environmental ChangesFaculdade de Ciências da Universidade de LisboaLisbonPortugal
- Departamento de Biologia AnimalGrupo LoboFaculdade de Ciências da Universidade de LisboaLisbonPortugal
| | - Francisco Petrucci‐Fonseca
- cE3c, Centre for Ecology, Evolution and Environmental ChangesFaculdade de Ciências da Universidade de LisboaLisbonPortugal
- Departamento de Biologia AnimalGrupo LoboFaculdade de Ciências da Universidade de LisboaLisbonPortugal
| | - Nuno Santos
- CIBIO/InBIOCentro de Investigação em Biodiversidade e Recursos GenéticosUniversidade do PortoVairãoPortugal
| | - Marc Van Ranst
- Department of Microbiology and ImmunologyLaboratory of Clinical VirologyRega Institute for Medical ResearchKU Leuven – University of LeuvenLeuvenBelgium
| | - João R. Mesquita
- CIBIO/InBIOCentro de Investigação em Biodiversidade e Recursos GenéticosUniversidade do PortoVairãoPortugal
- Department of Zootechnics, Rural Engineering and VeterinaryAgrarian Superior School of ViseuViseuPortugal
| | - Jelle Matthijnssens
- Department of Microbiology and ImmunologyLaboratory of Viral MetagenomicsRega Institute for Medical ResearchKU Leuven – University of LeuvenLeuvenBelgium
| |
Collapse
|
43
|
Deng YP, Liu YJ, Yang ZQ, Wang YJ, He BY, Liu P. Human bocavirus induces apoptosis and autophagy in human bronchial epithelial cells. Exp Ther Med 2017; 14:753-758. [PMID: 28672995 DOI: 10.3892/etm.2017.4533] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 12/23/2016] [Indexed: 12/20/2022] Open
Abstract
Human bocavirus (HBoV) is classified in the Bocavirus genus within the Parvoviridae family, first identified from children with respiratory diseases. Previous studies have investigated the stimulating effect of HBoV on cell apoptosis and autophagy. In the present study, human bronchial epithelial cells (HBECs) were utilized to examine the mechanism of HBoV recombination expressing vector (pWHL-1) on the promotion of cell apoptosis and autophagy. The results from the present study indicated that pWHL-1 inhibited the proliferation of HBECs in a time-dependent manner. Additionally, pWHL-1induced apoptosis, as substantiated by an increased apoptotic rate and presence of autophagosomes. Following pWHL-1 transfection, proliferating cell nuclear antigen, caspase-3 and B cell lymphoma 2 (Bcl-2) protein expression levels were decreased, with the exception of Bcl-2 associated × (Bax) protein, which increased. mRNA and protein expression levels of microtubule-associated protein 1A/1B-light chain 3 (LC3) II and autophagy protein 5 were increased in pWHL-1-transfected HBECs, whereas, the mRNA and protein levels of LC3I and sequestosome 1 were decreased. Notably, pWHL-1 also enhanced the activation of p53 and inhibited AKT activation in HBECs. Results from the present study suggest that pWHL-1 induces apoptosis and autophagy, thus providing a novel insight into the effect of HBoV and its uses in respiratory diseases.
Collapse
Affiliation(s)
- You-Ping Deng
- Department of Pediatrics, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Ying-Juan Liu
- Medical Research Center, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Zhan-Qiu Yang
- State Key Laboratory of Virology/Institute of Medical Virology, School of Medicine, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Yan-Jun Wang
- Department of Pediatrics, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Bing-Yan He
- Department of Pediatrics, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Pin Liu
- Department of Pediatrics, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
44
|
Minute Virus of Canines NP1 Protein Governs the Expression of a Subset of Essential Nonstructural Proteins via Its Role in RNA Processing. J Virol 2017; 91:JVI.00260-17. [PMID: 28356522 DOI: 10.1128/jvi.00260-17] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 03/24/2017] [Indexed: 12/24/2022] Open
Abstract
Parvoviruses use a variety of means to control the expression of their compact genomes. The bocaparvovirus minute virus of canines (MVC) encodes a small, genus-specific protein, NP1, which governs access to the viral capsid gene via its role in alternative polyadenylation and alternative splicing of the single MVC pre-mRNA. In addition to NP1, MVC encodes five additional nonstructural proteins (NS) that share an initiation codon at the left end of the genome and which are individually encoded by alternative multiply spliced mRNAs. We found that three of these proteins were encoded by mRNAs that excise the NP1-regulated MVC intron immediately upstream of the internal polyadenylation site, (pA)p, and that generation of these proteins was thus regulated by NP1. Splicing of their progenitor mRNAs joined the amino termini of these proteins to the NP1 open reading frame, and splice site mutations that prevented their expression inhibited virus replication in a host cell-dependent manner. Thus, in addition to controlling capsid gene access, NP1 also controls the expression of three of the five identified NS proteins via its role in governing MVC pre-mRNA splicing.IMPORTANCE The Parvovirinae are small nonenveloped icosahedral viruses that are important pathogens in many animal species, including humans. Minute virus of canine (MVC) is an autonomous parvovirus in the genus Bocaparvovirus It has a single promoter that generates a single pre-mRNA. NP1, a small genus-specific MVC protein, participates in the processing of this pre-mRNA and so controls capsid gene access via its role in alternative internal polyadenylation and splicing. We show that NP1 also controls the expression of three of the five identified NS proteins via its role in governing MVC pre-mRNA splicing. These NS proteins together are required for virus replication in a host cell-dependent manner.
Collapse
|
45
|
Parvovirus Expresses a Small Noncoding RNA That Plays an Essential Role in Virus Replication. J Virol 2017; 91:JVI.02375-16. [PMID: 28122984 DOI: 10.1128/jvi.02375-16] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 01/20/2017] [Indexed: 12/11/2022] Open
Abstract
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.
Collapse
|
46
|
Alternative Polyadenylation of Human Bocavirus at Its 3' End Is Regulated by Multiple Elements and Affects Capsid Expression. J Virol 2017; 91:JVI.02026-16. [PMID: 27881651 PMCID: PMC5244319 DOI: 10.1128/jvi.02026-16] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 11/12/2016] [Indexed: 12/23/2022] Open
Abstract
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.
Collapse
|
47
|
Abstract
Parvovirus B19 (B19V) and human bocavirus 1 (HBoV1), members of the large Parvoviridae family, are human pathogens responsible for a variety of diseases. For B19V in particular, host features determine disease manifestations. These viruses are prevalent worldwide and are culturable in vitro, and serological and molecular assays are available but require careful interpretation of results. Additional human parvoviruses, including HBoV2 to -4, human parvovirus 4 (PARV4), and human bufavirus (BuV) are also reviewed. The full spectrum of parvovirus disease in humans has yet to be established. Candidate recombinant B19V vaccines have been developed but may not be commercially feasible. We review relevant features of the molecular and cellular biology of these viruses, and the human immune response that they elicit, which have allowed a deep understanding of pathophysiology.
Collapse
Affiliation(s)
- Jianming Qiu
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | | | - Neal S Young
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
48
|
DNA Damage Signaling Is Required for Replication of Human Bocavirus 1 DNA in Dividing HEK293 Cells. J Virol 2016; 91:JVI.01831-16. [PMID: 27733644 DOI: 10.1128/jvi.01831-16] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 10/05/2016] [Indexed: 02/07/2023] Open
Abstract
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).
Collapse
|
49
|
Lau SKP, Ahmed SS, Yeung HC, Li KSM, Fan RYY, Cheng TYC, Cai JP, Wang M, Zheng BJ, Wong SSY, Woo PCY, Yuen KY. Identification and interspecies transmission of a novel bocaparvovirus among different bat species in China. J Gen Virol 2016; 97:3345-3358. [PMID: 27902362 DOI: 10.1099/jgv.0.000645] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
We report the discovery of a novel bocaparvovirus, bat bocaparvovirus (BtBoV), in one spleen, four respiratory and 61 alimentary samples from bats of six different species belonging to three families, Hipposideridae, Rhinolophidae and Vespertilionidae. BtBoV showed a higher detection rate in alimentary samples of Rhinolophus sinicus (5.7 %) than those of other bat species (0.43-1.59 %), supporting R. sinicus as the primary reservoir and virus spillover to accidental bat species. BtBoV peaked during the lactating season of R. sinicus, and it was more frequently detected among female than male adult bats (P<0.05), and among lactating than non-lactating female bats (P<0.0001). Positive BtBoV detection was associated with lower body weight in lactating bats (P<0.05). Ten nearly complete BtBoV genomes from three bat species revealed a unique large ORF1 spanning NS1 and NP1 in eight genomes and conserved splicing signals leading to multiple proteins, as well as a unique substitution in the conserved replication initiator motif within NS1. BtBoV was phylogenetically distantly related to known bocaparvoviruses with ≤57.3 % genome identities, supporting BtBoV as a novel species. Ms-BtBoV from Miniopterus schreibersii and Hp-BtBoV from Hipposideros pomona demonstrated 97.2-99.9 % genome identities with Rs-BtBoVs from R. sinicus, supporting infection of different bat species by a single BtBoV species. Rs-BtBoV_str15 represents the first bat parvovirus genome with non-coding regions sequenced, which suggested the presence of head-to-tail genomic concatamers or episomal forms of the genome. This study represents the first to describe interspecies transmission in BoVs. The high detection rates in lactating female and juvenile bats suggest possible vertical transmission of BtBoV.
Collapse
Affiliation(s)
- Susanna K P Lau
- Department of Microbiology, The University of Hong Kong, Hong Kong SAR, PR China.,Carol Yu Centre for Infection, The University of Hong Kong, Hong Kong SAR, PR China.,State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong SAR, PR China.,Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong SAR, PR China
| | - Syed Shakeel Ahmed
- Department of Microbiology, The University of Hong Kong, Hong Kong SAR, PR China
| | - Hazel C Yeung
- Department of Microbiology, The University of Hong Kong, Hong Kong SAR, PR China
| | - Kenneth S M Li
- Department of Microbiology, The University of Hong Kong, Hong Kong SAR, PR China
| | - Rachel Y Y Fan
- Department of Microbiology, The University of Hong Kong, Hong Kong SAR, PR China
| | - Toni Y C Cheng
- Department of Microbiology, The University of Hong Kong, Hong Kong SAR, PR China
| | - Jian-Piao Cai
- Department of Microbiology, The University of Hong Kong, Hong Kong SAR, PR China
| | - Ming Wang
- Guangzhou Center for Disease Control and Prevention, Guangzhou, PR China
| | - Bo-Jian Zheng
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong SAR, PR China.,Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong SAR, PR China.,Carol Yu Centre for Infection, The University of Hong Kong, Hong Kong SAR, PR China.,Department of Microbiology, The University of Hong Kong, Hong Kong SAR, PR China
| | - Samson S Y Wong
- Department of Microbiology, The University of Hong Kong, Hong Kong SAR, PR China.,Carol Yu Centre for Infection, The University of Hong Kong, Hong Kong SAR, PR China.,Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong SAR, PR China.,State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong SAR, PR China
| | - Patrick C Y Woo
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong SAR, PR China.,Department of Microbiology, The University of Hong Kong, Hong Kong SAR, PR China.,Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong SAR, PR China.,Carol Yu Centre for Infection, The University of Hong Kong, Hong Kong SAR, PR China
| | - Kwok-Yung Yuen
- Department of Microbiology, The University of Hong Kong, Hong Kong SAR, PR China.,Carol Yu Centre for Infection, The University of Hong Kong, Hong Kong SAR, PR China.,Research Centre of Infection and Immunology, The University of Hong Kong, Hong Kong SAR, PR China.,State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong SAR, PR China
| |
Collapse
|
50
|
Guido M, Tumolo MR, Verri T, Romano A, Serio F, De Giorgi M, De Donno A, Bagordo F, Zizza A. Human bocavirus: Current knowledge and future challenges. World J Gastroenterol 2016; 22:8684-8697. [PMID: 27818586 PMCID: PMC5075545 DOI: 10.3748/wjg.v22.i39.8684] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 08/30/2016] [Accepted: 09/14/2016] [Indexed: 02/06/2023] Open
Abstract
Human bocavirus (HBoV) is a parvovirus isolated about a decade ago and found worldwide in both respiratory samples, mainly from early life and children of 6-24 mo of age with acute respiratory infection, and in stool samples, from patients with gastroenteritis. Since then, other viruses related to the first HBoV isolate (HBoV1), namely HBoV2, HBoV3 and HBoV4, have been detected principally in human faeces. HBoVs are small non-enveloped single-stranded DNA viruses of about 5300 nucleotides, consisting of three open reading frames encoding the first two the non-structural protein 1 (NS1) and nuclear phosphoprotein (NP1) and the third the viral capsid proteins 1 and 2 (VP1 and VP2). HBoV pathogenicity remains to be fully clarified mainly due to the lack of animal models for the difficulties in replicating the virus in in vitro cell cultures, and the fact that HBoV infection is frequently accompanied by at least another viral and/or bacterial respiratory and/or gastroenteric pathogen infection. Current diagnostic methods to support HBoV detection include polymerase chain reaction, real-time PCR, enzyme-linked immunosorbent assay and enzyme immunoassay using recombinant VP2 or virus-like particle capsid proteins, although sequence-independent amplification techniques combined with next-generation sequencing platforms promise rapid and simultaneous detection of the pathogens in the future. This review presents the current knowledge on HBoV genotypes with emphasis on taxonomy, phylogenetic relationship and genomic analysis, biology, epidemiology, pathogenesis and diagnostic methods. The emerging discussion on HBoVs as true pathogen or innocent bystander is also emphasized.
Collapse
|