1
|
Lee H, Assaraf R, Subramanian S, Goetschius D, Bieri J, DiNunno NM, Leisi R, Bator CM, Hafenstein SL, Ros C. Infectious parvovirus B19 circulates in the blood coated with active host protease inhibitors. Nat Commun 2024; 15:9543. [PMID: 39500886 PMCID: PMC11538491 DOI: 10.1038/s41467-024-53794-1] [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: 07/02/2024] [Accepted: 10/20/2024] [Indexed: 11/08/2024] Open
Abstract
The lack of a permissive cell culture system has limited high-resolution structures of parvovirus B19 (B19V) to virus-like particles (VLPs). In this study, we present the atomic resolution structure (2.2 Å) of authentic B19V purified from a patient blood sample. There are significant differences compared to non-infectious VLPs. Most strikingly, two host protease inhibitors (PIs), inter-alpha-trypsin inhibitor heavy chain 4 (ITIH4) and serpinA3, were identified in complex with the capsids in all patient samples tested. The ITIH4 binds specifically to the icosahedral fivefold axis and serpinA3 occupies the twofold axis. The protein-coated virions remain infectious, and the capsid-associated PIs retain activity; however, upon virion interaction with target cells, the PIs dissociate from the capsid prior to viral entry. Our finding of an infectious virion shielded by bound host serum proteins suggests an evolutionarily favored phenomenon to evade immune surveillance and escape host protease activity.
Collapse
Affiliation(s)
- Hyunwook Lee
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Ruben Assaraf
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, Bern, Switzerland
| | | | - Dan Goetschius
- The Pennsylvania State College of Medicine, Hershey, PA, USA
| | - Jan Bieri
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| | - Nadia M DiNunno
- The Pennsylvania State College of Medicine, Hershey, PA, USA
| | - Remo Leisi
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| | - Carol M Bator
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Susan L Hafenstein
- The Hormel Institute, University of Minnesota, Austin, MN, USA.
- Department of Biochemistry, Biophysics and Molecular Biology, University of Minnesota, Minneapolis, MN, USA.
- Department of Infectious Diseases, Mayo Clinic, Rochester, MN, USA.
| | - Carlos Ros
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland.
| |
Collapse
|
2
|
Mellid-Carballal R, Gutierrez-Gutierrez S, Rivas C, Garcia-Fuentes M. Viral protein-based nanoparticles (part 2): Pharmaceutical applications. Eur J Pharm Sci 2023; 189:106558. [PMID: 37567394 DOI: 10.1016/j.ejps.2023.106558] [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: 02/17/2023] [Revised: 07/10/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023]
Abstract
Viral protein nanoparticles (ViP NPs) such as virus-like particles and virosomes are structures halfway between viruses and synthetic nanoparticles. The biological nature of ViP NPs endows them with the biocompatibility, biodegradability, and functional properties that many synthetic nanoparticles lack. At the same time, the absence of a viral genome avoids the safety concerns of viruses. Such characteristics of ViP NPs offer a myriad of opportunities for theirapplication at several points across disease development: from prophylaxis to diagnosis and treatment. ViP NPs present remarkable immunostimulant properties, and thus the vaccination field has benefited the most from these platforms capable of overcoming the limitations of both traditional and subunit vaccines. This was reflected in the marketing authorization of several VLP- and virosome-based vaccines. Besides, ViP NPs inherit the ability of viruses to deliver their cargo to target cells. Because of that, ViP NPs are promising candidates as vectors for drug and gene delivery, and for diagnostic applications. In this review, we analyze the pharmaceutical applications of ViP NPs, describing the products that are commercially available or under clinical evaluation, but also the advances that scientists are making toward the implementation of ViP NPs in other areas of major pharmaceutical interest.
Collapse
Affiliation(s)
- Rocio Mellid-Carballal
- CiMUS Research Center, Universidad de Santiago de Compostela, Spain; Department of Pharmacology, Pharmacy and Pharmaceutical Technology, Universidad de Santiago de Compostela, Spain
| | - Sara Gutierrez-Gutierrez
- CiMUS Research Center, Universidad de Santiago de Compostela, Spain; Department of Pharmacology, Pharmacy and Pharmaceutical Technology, Universidad de Santiago de Compostela, Spain
| | - Carmen Rivas
- CiMUS Research Center, Universidad de Santiago de Compostela, Spain; Health Research Institute of Santiago de Compostela (IDIS), Universidad de Santiago de Compostela, Spain; Departamento de Biología Molecular y Celular, Centro Nacional de Biotecnología (CNB)-CSIC, Spain
| | - Marcos Garcia-Fuentes
- CiMUS Research Center, Universidad de Santiago de Compostela, Spain; Department of Pharmacology, Pharmacy and Pharmaceutical Technology, Universidad de Santiago de Compostela, Spain; Health Research Institute of Santiago de Compostela (IDIS), Universidad de Santiago de Compostela, Spain.
| |
Collapse
|
3
|
Sánchez-Moguel I, Montiel C, Bustos-Jaimes I. Therapeutic Potential of Engineered Virus-like Particles of Parvovirus B19. Pathogens 2023; 12:1007. [PMID: 37623967 PMCID: PMC10458557 DOI: 10.3390/pathogens12081007] [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: 07/08/2023] [Revised: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 08/26/2023] Open
Abstract
Virus-like particles (VLPs) comprise one or many structural components of virions, except their genetic material. Thus, VLPs keep their structural properties of cellular recognition while being non-infectious. VLPs of Parvovirus B19 (B19V) can be produced by the heterologous expression of their structural proteins VP1 and VP2 in bacteria. These proteins are purified under denaturing conditions, refolded, and assembled into VLPs. Moreover, chimeric forms of VP2 have been constructed to harbor peptides or functional proteins on the surface of the particles without dropping their competence to form VLPs, serving as presenting nanoparticles. The in-vitro assembly approach offers exciting possibilities for the composition of VLPs, as more than one chimeric form of VP2 can be included in the assembly stage, producing multifunctional VLPs. Here, the heterologous expression and in-vitro assembly of B19V structural proteins and their chimeras are reviewed. Considerations for the engineering of the structural proteins of B19V are also discussed. Finally, the construction of multifunctional VLPs and their future potential as innovative medical tools are examined.
Collapse
Affiliation(s)
- Ignacio Sánchez-Moguel
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City 04510, Mexico;
| | - Carmina Montiel
- Departamento de Alimentos y Biotecnología, Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Mexico City 04510, Mexico;
| | - Ismael Bustos-Jaimes
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City 04510, Mexico;
| |
Collapse
|
4
|
Tracking of Human Parvovirus B19 Virus-Like Particles Using Short Peptide Tags Reveals a Membrane-Associated Extracellular Release of These Particles. J Virol 2023; 97:e0163122. [PMID: 36749078 PMCID: PMC9972994 DOI: 10.1128/jvi.01631-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
B19 virus (B19V) is a pathogenic human parvovirus that infects erythroid progenitor cells. Because there are limited in vitro culture systems to propagate this virus, little is known about the molecular mechanisms by which it propagates in cells. In this study, we introduced a HiBiT peptide tag into various loops of VP2 located on the surface of B19V particles and evaluated their ability to form virus-like particles (VLPs). Three independent sites were identified as permissive sites for peptide tag insertion without affecting VLP formation. When the HiBiT tag was introduced into B19V clones (pB19-M20) and transfected into a semipermissive erythroleukemia cell line (UT7/Epo-S1), HiBiT-dependent luciferase activities (HiBiT activities) increased depending on helicase activity of viral NS1. Furthermore, we used a GFP11 tag-split system to visualize VLPs in the GFP1-10-expressing live cells. Time-lapse imaging of green fluorescent protein (GFP)-labeled VLPs revealed that nuclear VLPs were translocated into the cytoplasm only after cell division, suggesting that the breakdown of the nuclear envelope during mitosis contributes to VLP nuclear export. Moreover, HiBiT activities of culture supernatants were dependent on the presence of a detergent, and the released VLPs were associated with extracellular vesicles, as observed under electron microscopy. Treatment with an antimitotic agent (nocodazole) enhanced the release of VLPs. These results suggest that the virions accumulated in the cytoplasm are constitutively released from the cell as membrane-coated vesicles. These properties are likely responsible for viral escape from host immune responses and enhance membrane fusion-mediated transmission. IMPORTANCE Parvovirus particles are expected to be applied as nanoparticles in drug delivery systems. However, little is known about how nuclear-assembled B19 virus (B19V) virions are released from host cells. This study provides evidence of mitosis-dependent nuclear export of B19V and extracellular vesicle-mediated virion release. Moreover, this study provides methods for modifying particle surfaces with various exogenous factors and contributes to the development of fine nanoparticles with novel valuable functions. The pB19-M20 plasmid expressing HiBiT-tagged VP2 is a novel tool to easily quantify VP2 expression. Furthermore, this system can be applied in high-throughput screening of reagents that affect VP2 expression, which might be associated with viral propagation.
Collapse
|
5
|
Ning K, Zou W, Xu P, Cheng F, Zhang EY, Zhang-Chen A, Kleiboeker S, Qiu J. Identification of AXL as a co-receptor for human parvovirus B19 infection of human erythroid progenitors. SCIENCE ADVANCES 2023; 9:eade0869. [PMID: 36630517 PMCID: PMC9833669 DOI: 10.1126/sciadv.ade0869] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 12/09/2022] [Indexed: 05/31/2023]
Abstract
Parvovirus B19 (B19V) infects human erythroid progenitor cells (EPCs) and causes several hematological disorders and fetal hydrops. Amino acid (aa) 5-68 of minor capsid protein VP1 (VP1u5-68aa) is the minimal receptor binding domain for B19V to enter EPCs. Here, we carried out a genome-wide CRISPR-Cas9 guide RNA screen and identified tyrosine protein kinase receptor UFO (AXL) as a proteinaceous receptor for B19V infection of EPCs. AXL gene silencing in ex vivo expanded EPCs remarkably decreased B19V internalization and replication. Additions of the recombinant AXL extracellular domain or a polyclonal antibody against it upon infection efficiently inhibited B19V infection of ex vivo expanded EPCs. Moreover, B19V VP1u interacted with the recombinant AXL extracellular domain in vitro at a relatively high affinity (KD = 103 nM). Collectively, we provide evidence that AXL is a co-receptor for B19V infection of EPCs.
Collapse
Affiliation(s)
- Kang Ning
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Wei Zou
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Peng Xu
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Fang Cheng
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | | | | | - Steve Kleiboeker
- Department of Research and Development, ViraCor Eurofins Laboratories, Lenexa, KS 66219, USA
| | - Jianming Qiu
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
6
|
Recombinant Virus-like Particles of Human Parvovirus B19 with the Internal Location of VP1 Unique Region Produced by Hansenula polymorpha. Viruses 2022; 14:v14112410. [PMID: 36366508 PMCID: PMC9695803 DOI: 10.3390/v14112410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 10/27/2022] [Accepted: 10/28/2022] [Indexed: 01/31/2023] Open
Abstract
Human parvovirus B19 (HPV B19) is pathogenic to human, which can cause fifth disease, transient aplastic crisis, arthritis, myocarditis, autoimmune disorders, hydrops fetalis, and so on. Currently, no approved vaccines or antiviral drugs are available against HPV B19, and thus the development of effective vaccines is needed. The capsid of HPV B19 is composed of two types of proteins, i.e., the major capsid protein VP2 and the minor protein VP1. Previous experimental studies have shown that the dominant immune responses against HPV B19 are elicited by VP1, especially the unique region on the N-terminus of VP1. It has been found that VP2 alone or VP2 and VP1 together can assemble into virus-like particle (VLP). The VLP structure formed by VP2 has been resolved, however, the location of VP1 in the capsid, especially the location of VP1 unique region with strong immunogenicity, is still not clear. In the present work, using the Hansenula polymorpha expression system developed by our laboratory, two kinds of recombinant HPV B19 VLPs were expressed, i.e., the VLP co-assembled by VP1 and VP2 (VP1/VP2 VLP) and the VLP whose VP1 content was improved (VP1h/VP2 VLP). The expression, purity, and morphology of these two VLPs were characterized, and then their immunogenic properties were investigated and compared with those of the VLP containing VP2 alone (VP2 VLP) previously developed by our group. Furthermore, the location of the VP1 unique region in the VLPs was determined by using the immunogold electron microscopy (IGEM). Our experimental results show that the VP1h/VP2 VLP elicits a stronger neutralization against the HPV B19 than VP2 and VP1/VP2 VLPs, which implies that the increase of VP1 content significantly improves the level of neutralizing antibodies. In addition, the IGEM observations suggest that the unique region of VP1 may be located inside the recombinant VLP. The VLPs recombinantly expressed by our Hansenula polymorpha system may serve as a promising candidate immunogen for HPV B19 vaccine development.
Collapse
|
7
|
Lakshmanan RV, Hull JA, Berry L, Burg M, Bothner B, McKenna R, Agbandje-McKenna M. Structural Dynamics and Activity of B19V VP1u during the pHs of Cell Entry and Endosomal Trafficking. Viruses 2022; 14:1922. [PMID: 36146728 PMCID: PMC9505059 DOI: 10.3390/v14091922] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/28/2022] [Accepted: 08/28/2022] [Indexed: 12/28/2022] Open
Abstract
Parvovirus B19 (B19V) is a human pathogen that is the causative agent of fifth disease in children. It is also known to cause hydrops in fetuses, anemia in AIDS patients, and transient aplastic crisis in patients with sickle cell disease. The unique N-terminus of Viral Protein 1 (VP1u) of parvoviruses, including B19V, exhibits phospholipase A2 (PLA2) activity, which is required for endosomal escape. Presented is the structural dynamics of B19V VP1u under conditions that mimic the pHs of cell entry and endosomal trafficking to the nucleus. Using circular dichroism spectroscopy, the receptor-binding domain of B19V VP1u is shown to exhibit an α-helical fold, whereas the PLA2 domain exhibits a probable molten globule state, both of which are pH invariant. Differential scanning calorimetry performed at endosomal pHs shows that the melting temperature (Tm) of VP1u PLA2 domain is tuned to body temperature (37 °C) at pH 7.4. In addition, PLA2 assays performed at temperatures ranging from 25-45 °C show both a temperature and pH-dependent change in activity. We hypothesize that VP1u PLA2 domain differences in Tm at differing pHs have enabled the virus to "switch on/off" the phospholipase activity during capsid trafficking. Furthermore, we propose the environment of the early endosome as the optimal condition for endosomal escape leading to B19V infection.
Collapse
Affiliation(s)
- Renuk V. Lakshmanan
- Department of Biochemistry and Molecular Biology, Center for Structural Biology, The McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Joshua A. Hull
- Department of Biochemistry and Molecular Biology, Center for Structural Biology, The McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Luke Berry
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT 59717, USA
| | - Matthew Burg
- Department of Biochemistry and Molecular Biology, Center for Structural Biology, The McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Brian Bothner
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT 59717, USA
| | - Robert McKenna
- Department of Biochemistry and Molecular Biology, Center for Structural Biology, The McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Mavis Agbandje-McKenna
- Department of Biochemistry and Molecular Biology, Center for Structural Biology, The McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
8
|
Szwarcwort M, Wax M, Halutz O, Mendelson E, Mor O. Evaluation of Biotrin, Serion and Euroimmun Commercial Assays for the Detection of Parvovirus B19-Specific IgM and IgG Antibodies. Diagn Microbiol Infect Dis 2022; 103:115723. [DOI: 10.1016/j.diagmicrobio.2022.115723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/03/2022] [Accepted: 05/05/2022] [Indexed: 11/03/2022]
|
9
|
Wiestner A, Issaragrisil S, Kaufman DW, Ozawa K, Nakao S, Kajigaya S, Wang J, Wu Z, Binh VTT, Dhawan R, Nair V. COLLABORATIONS, COLLEAGUES AND FRIENDSHIPS: THE HEMATOLOGY BRANCH AND BLOOD DISEASE CENTERS IN ASIA. Semin Hematol 2022; 59:6-12. [DOI: 10.1053/j.seminhematol.2022.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
10
|
Soto-Valerio IA, Cayetano-Cruz M, Valadez-García J, Guadarrama P, Méndez C, Bustos-Jaimes I. In vitro refolding of the structural protein VP1 of parvovirus B19 produces virus-like particles with functional VP1 unique region. Virology 2022; 570:57-66. [DOI: 10.1016/j.virol.2022.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 03/01/2022] [Accepted: 03/24/2022] [Indexed: 10/18/2022]
|
11
|
Penkert RR, Chandramouli S, Dormitzer PR, Settembre EC, Sealy RE, Wong S, Young NS, Sun Y, Tang L, Cotton A, Dowdy J, Hayden RT, Hankins JS, Hurwitz JL. Novel Surrogate Neutralizing Assay Supports Parvovirus B19 Vaccine Development for Children with Sickle Cell Disease. Vaccines (Basel) 2021; 9:vaccines9080860. [PMID: 34451986 PMCID: PMC8402426 DOI: 10.3390/vaccines9080860] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 07/27/2021] [Accepted: 07/27/2021] [Indexed: 11/16/2022] Open
Abstract
Children with sickle cell disease (SCD) suffer life-threatening transient aplastic crisis (TAC) when infected with parvovirus B19. In utero, infection of healthy fetuses may result in anemia, hydrops, and death. Unfortunately, although promising vaccine candidates exist, no product has yet been licensed. One barrier to vaccine development has been the lack of a cost-effective, standardized parvovirus B19 neutralization assay. To fill this void, we evaluated the unique region of VP1 (VP1u), which contains prominent targets of neutralizing antibodies. We discovered an antigenic cross-reactivity between VP1 and VP2 that, at first, thwarted the development of a surrogate neutralization assay. We overcame the cross-reactivity by designing a mutated VP1u (VP1uAT) fragment. A new VP1uAT ELISA yielded results well correlated with neutralization (Spearman’s correlation coefficient = 0.581; p = 0.001), superior to results from a standard clinical diagnostic ELISA or an ELISA with virus-like particles. Virus-specific antibodies from children with TAC, measured by the VP1uAT and neutralization assays, but not other assays, gradually increased from days 0 to 120 post-hospitalization. We propose that this novel and technically simple VP1uAT ELISA might now serve as a surrogate for the neutralization assay to support rapid development of a parvovirus B19 vaccine.
Collapse
Affiliation(s)
- Rhiannon R. Penkert
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (R.R.P.); (R.E.S.)
| | - Sumana Chandramouli
- Novartis Vaccines and Diagnostics, Cambridge, MA 02139, USA; (S.C.); (P.R.D.); (E.C.S.)
| | - Philip R. Dormitzer
- Novartis Vaccines and Diagnostics, Cambridge, MA 02139, USA; (S.C.); (P.R.D.); (E.C.S.)
| | - Ethan C. Settembre
- Novartis Vaccines and Diagnostics, Cambridge, MA 02139, USA; (S.C.); (P.R.D.); (E.C.S.)
| | - Robert E. Sealy
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (R.R.P.); (R.E.S.)
| | - Susan Wong
- Hematology Branch, National Heart, Lung and Blood Institute, Bethesda, MD 20892, USA; (S.W.); (N.S.Y.)
| | - Neal S. Young
- Hematology Branch, National Heart, Lung and Blood Institute, Bethesda, MD 20892, USA; (S.W.); (N.S.Y.)
| | - Yilun Sun
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (Y.S.); (L.T.)
| | - Li Tang
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (Y.S.); (L.T.)
| | - Alyssa Cotton
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (A.C.); (J.D.); (J.S.H.)
| | - Jola Dowdy
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (A.C.); (J.D.); (J.S.H.)
| | - Randall T. Hayden
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA;
| | - Jane S. Hankins
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (A.C.); (J.D.); (J.S.H.)
| | - Julia L. Hurwitz
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (R.R.P.); (R.E.S.)
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Correspondence: ; Tel.: +1-901-595-2464
| |
Collapse
|
12
|
Fusion of parvovirus B19 receptor-binding domain and pneumococcal surface protein A induces protective immunity against parvovirus B19 and Streptococcus pneumoniae. Vaccine 2021; 39:5146-5152. [PMID: 34340860 DOI: 10.1016/j.vaccine.2021.07.046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/15/2021] [Accepted: 07/18/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND Parvovirus B19 (B19) is a well-known cause of fifth disease in children, but infection during pregnancy may cause hydrops fetalis and stillbirth. The receptor-binding domain (RBD) of the VP1 unique capsid plays a pivotal role in infection. Here, we aimed to improve the immunogenicity of an RBD-based vaccine by genetically fusing it with Streptococcus pneumoniae surface protein A (PspA). METHODS Mice were intramuscularly injected with RBD-based vaccines. Antigen-specific antibodies and neutralizing activity against B19 were measured. Protective immunity against S. pneumoniae was evaluated by monitoring the survival of mice nasally challenged with bacteria and determining antigen-specific T cell activation in splenic cells. RESULTS RBD alone failed to generate neutralizing antibodies against B19, but fusion with PspA induced higher levels of neutralizing IgG compared to B19 virus-like particles. Furthermore, a comparable level of PspA-specific IgG was induced by RBD-PspA and PspA alone, which was sufficient to protect mice against pneumococcal infection. Stimulation with PspA, but not RBD, induced cytokine production in splenic cells from mice immunized with RBD-PspA, suggesting that PspA-specific T cells supported immunoglobulin class switching of both RBD- and PspA-specific B cells. CONCLUSIONS RBD-PspA should be an effective bivalent vaccine against B19 and S. pneumoniae infections.
Collapse
|
13
|
Van den Abeele T, Delforge ML, Boel A, Reynders M, Padalko E. Comparison of 4 commercial enzyme immunoassays for serology testing of human parvovirus B19 infection. Diagn Microbiol Infect Dis 2021; 101:115489. [PMID: 34352435 DOI: 10.1016/j.diagmicrobio.2021.115489] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 05/25/2021] [Accepted: 07/11/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Parvovirus B19 is a pathogenic virus often diagnosed by serology, yet little is known about analytical performance of commercial enzyme immunoassays (EIAs). OBJECTIVE To investigate performance of 4 EIAs for parvovirus B19 IgM and IgG: Liaison, Euroimmun, Mikrogen and Virion/Serion. STUDY DESIGN To compare 4 EIAs to Biotrin's ELISA on 168 samples and determine consensus score for discordant samples using Mikrogen's confirmatory line assay. RESULTS Two thirds of results for IgM/IgG were identical for all 4 EIAs and Biotrin. Liaison shows the highest IgM sensitivity, but has low specificity. Euroimmun lacks IgM sensitivity. Mikrogen had a good overall performance, but had the lowest IgG specificity. Virion/Serion had variable performance with a low IgM specificity and the most borderline and cross-reactive results. CONCLUSIONS Liaison and Mikrogen have similar performance to Biotrin's ELISA. Euroimmun lacks sensitivity and Virion/Serion produced many borderline and cross-reactive results.
Collapse
Affiliation(s)
- Tim Van den Abeele
- Laboratory of Medical Microbiology, Ghent University Hospital, Gent, Belgium
| | - Marie-Luce Delforge
- National Reference Center for Congenital Infections, Erasme University Hospital, Brussels, Belgium
| | - An Boel
- Department of Microbiology, OLV Hospital, Aalst, Belgium
| | - Marijke Reynders
- Department of Medical Microbiology, Sint-Jan Hospital, Bruges, Belgium
| | - Elizaveta Padalko
- Laboratory of Medical Microbiology, Ghent University Hospital, Gent, Belgium.
| |
Collapse
|
14
|
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: 2.3] [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
|
15
|
Bombyx mori Pupae Efficiently Produce Recombinant AAV2/HBoV1 Vectors with a Bombyx mori Nuclear Polyhedrosis Virus Expression System. Viruses 2021; 13:v13040704. [PMID: 33919645 PMCID: PMC8073075 DOI: 10.3390/v13040704] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/09/2021] [Accepted: 04/16/2021] [Indexed: 01/07/2023] Open
Abstract
Recombinant adeno-associated virus (AAV) vectors have broad application prospects in the field of gene therapy. The establishment of low-cost and large-scale manufacturing is now the general agenda for industry. The baculovirus-insect cell/larva expression system has great potential for these applications due to its scalability and predictable biosafety. To establish a more efficient production system, Bombyx mori pupae were used as a new platform and infected with recombinant Bombyx mori nuclear polyhedrosis virus (BmNPV). The production of a chimeric recombinant adeno-associated virus (rAAV) serotype 2/human bocavirus type-1 (HBoV1) vector was used to evaluate the efficiency of this new baculovirus expression vector (BEV)–insect expression system. For this purpose, we constructed two recombinant BmNPVs, which were named rBmNPV/AAV2Rep-HBoV1Cap and rBmNPV/AAV2ITR-eGFP. The yields of rAAV2/HBoV1 derived from the rBmNPV/AAV2Rep-HBoV1Cap and rBmNPV/AAV2ITR-eGFP co-infected BmN cells exceeded 2 × 104 vector genomes (VG) per cell. The rBmNPV/AAV2Rep-HBoV1Cap and rBmNPV/AAV2ITR-eGFP can express stably for at least five passages. Significantly, rAAV2/HBoV1 could be efficiently generated from BmNPV-infected silkworm larvae and pupae at average yields of 2.52 × 1012 VG/larva and 4.6 × 1012 VG/pupa, respectively. However, the vectors produced from the larvae and pupae had a high percentage of empty particles, which suggests that further optimization is required for this platform in the future. Our work shows that silkworm pupae, as an efficient bioreactor, have great potential for application in the production of gene therapy vectors.
Collapse
|
16
|
Hyman P, Trubl G, Abedon ST. Virus-Like Particle: Evolving Meanings in Different Disciplines. PHAGE (NEW ROCHELLE, N.Y.) 2021; 2:11-15. [PMID: 36148434 PMCID: PMC9041479 DOI: 10.1089/phage.2020.0026] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Virus-like particle (VLP) is a term that has been in use for about 80 years. Usually, VLP has meant a particle that is like a virus, generally by appearance, but without either proven or actual virus functionality. Initially VLP referred to particles seen in electron microscope images of tissues. More recently, VLP has come to mean other things to other researchers. A key divergence has been use of VLP in association with vaccine and biotechnology applications versus use of VLP in enumeration of viruses in environmental samples. To these viral ecologists, a VLP is a particle that is virus sized, has nucleic acid, and could be a functional virus. But to vaccine developers and biotechnology researchers a VLP instead is a viral structure that intentionally lacks a viral genome. In this study, we look at the history of use of VLP, following changes in meaning as the technology to study VLPs changed.
Collapse
Affiliation(s)
- Paul Hyman
- Department of Biology and Toxicology, Ashland University, Ashland, Ohio, USA
| | - Gareth Trubl
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, California, USA
| | - Stephen T. Abedon
- Department of Microbiology, The Ohio State University, Mansfield, Ohio, USA
| |
Collapse
|
17
|
Liu J, Sun M, Cho KB, Gao X, Guo B. A CRISPR-Cas9 repressor for epigenetic silencing of KRAS. Pharmacol Res 2021; 164:105304. [PMID: 33202255 PMCID: PMC8422974 DOI: 10.1016/j.phrs.2020.105304] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 10/20/2020] [Accepted: 11/10/2020] [Indexed: 01/10/2023]
Abstract
KRAS is one of the most frequently mutated oncogenes in cancers. Currently no direct and effective anti-KRAS therapies are available. Using the powerful CRISPR-Cas9 technology to target the mutant KRAS promoter, we designed an epigenetic repressor to silence KRAS through epigenome editing. Catalytically dead Cas9 (dCas9) functioned as a DNA binding device, which was fused with a transcriptional repressor histone deacetylase 1 (HDAC1). We designed a panel of three CRISPR RNAs (crRNAs) covering 1500-bp range of the KRAS promoter and identified that crRNA1 and crRNA2 efficiently silenced KRAS. The suppression of K-Ras resulted in a significant inhibition of cell growth, suppression of colony formation in soft agar and induction of cell death in cancer cells with KRAS mutations. In addition, the chromatin immunoprecipitation (ChIP) assay demonstrated dCas9-HDAC1 modified histone acetylation on the KRAS promoter. Furthermore, transfection of dCas9-HDAC1 protein and gRNA ribonucleoprotein complex also inhibited K-Ras and suppressed cell proliferation. In summary, we have developed a new strategy that combines CRISPR-Cas9 technology with HDAC1 epigenetic silencing to target cancers driven by KRAS mutations.
Collapse
Affiliation(s)
- Jingwen Liu
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX, 77204, United States
| | - Meiyan Sun
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX, 77204, United States; College of Laboratory Medicine, Jilin Medical University, Jilin Province, 132013, China.
| | - Kwang Bog Cho
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX, 77204, United States
| | - Xiang Gao
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX, 77204, United States
| | - Bin Guo
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX, 77204, United States.
| |
Collapse
|
18
|
Penkert RR, Hankins JS, Young NS, Hurwitz JL. Vaccine Design Informed by Virus-Induced Immunity. Viral Immunol 2020; 33:342-350. [PMID: 32366204 PMCID: PMC7247049 DOI: 10.1089/vim.2019.0138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
When an individual is exposed to a viral pathogen for the first time, the adaptive immune system is naive and cannot prevent virus replication. The consequence may be severe disease. At the same time, the host may rapidly generate a pathogen-specific immune response that will prevent disease if the virus is encountered again. Parvovirus B19 provides one such example. Children with sickle cell disease can experience life-threatening transient aplastic crisis when first exposed to parvovirus B19, but an effective immune response confers lifelong protection. We briefly examine the induction and benefits of virus-induced immunity. We focus on three human viruses for which there are no licensed vaccines (respiratory syncytial virus, human immunodeficiency virus type 1, and parvovirus B19) and consider how virus-induced immunity may inform successful vaccine design.
Collapse
Affiliation(s)
- Rhiannon R. Penkert
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Jane S. Hankins
- Pathology Department, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Neal S. Young
- Hematology Branch, National Heart, Lung and Blood Institute, Bethesda, Maryland, USA
| | - Julia L. Hurwitz
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
19
|
Emmanuel SN, Mietzsch M, Tseng YS, Smith JK, Agbandje-McKenna M. Parvovirus Capsid-Antibody Complex Structures Reveal Conservation of Antigenic Epitopes Across the Family. Viral Immunol 2020; 34:3-17. [PMID: 32315582 DOI: 10.1089/vim.2020.0022] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The parvoviruses are small nonenveloped single stranded DNA viruses that constitute members that range from apathogenic to pathogenic in humans and animals. The infection with a parvovirus results in the generation of antibodies against the viral capsid by the host immune system to eliminate the virus and to prevent re-infection. For members currently either being developed as delivery vectors for gene therapy applications or as oncolytic biologics for tumor therapy, efforts are aimed at combating the detrimental effects of pre-existing or post-treatment antibodies that can eliminate therapeutic benefits. Therefore, understanding antigenic epitopes of parvoviruses can provide crucial information for the development of vaccination applications and engineering novel capsids able to escape antibody recognition. This review aims to capture the information for the binding regions of ∼30 capsid-antibody complex structures of different parvovirus capsids determined to date by cryo-electron microscopy and three-dimensional image reconstruction. The comparison of all complex structures revealed the conservation of antigenic regions among parvoviruses from different genera despite low sequence identity and indicates that the available data can be used across the family for vaccine development and capsid engineering.
Collapse
Affiliation(s)
- Shanan N Emmanuel
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Mario Mietzsch
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Yu Shan Tseng
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - James Kennon Smith
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Mavis Agbandje-McKenna
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
20
|
Subramanian S, Maurer AC, Bator CM, Makhov AM, Conway JF, Turner KB, Marden JH, Vandenberghe LH, Hafenstein SL. Filling Adeno-Associated Virus Capsids: Estimating Success by Cryo-Electron Microscopy. Hum Gene Ther 2019; 30:1449-1460. [PMID: 31530236 DOI: 10.1089/hum.2019.041] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Adeno-associated viruses (AAVs) have been employed successfully as gene therapy vectors in treating various genetic diseases for almost two decades. However, transgene packaging is usually imperfect, and developing a rapid and accurate method for measuring the proportion of DNA encapsidation is an important step for improving the downstream process of large scale vector production. In this study, we used two-dimensional class averages and three-dimensional classes, intermediate outputs in the single particle cryo-electron microscopy (cryo-EM) image reconstruction pipeline, to determine the proportion of DNA-packaged and empty capsid populations. Two different preparations of AAV3 were analyzed to estimate the minimum number of particles required to be sampled by cryo-EM in order for robust calculation of the proportion of the full versus empty capsids in any given sample. Cost analysis applied to the minimum amount of data required for a valid ratio suggests that cryo-EM is an effective approach to analyze vector preparations.
Collapse
Affiliation(s)
- Suriyasri Subramanian
- Department of Medicine, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Anna C Maurer
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute, Massachusetts Eye and Ear, Boston, Massachusetts.,Department of Ophthalmology, Harvard Medical School, Ocular Genomics Institute, Boston, Massachusetts.,The Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Carol M Bator
- Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania
| | - Alexander M Makhov
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - James F Conway
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Kevin B Turner
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - James H Marden
- Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania.,Department of Biology, Pennsylvania State University, University Park, Pennsylvania
| | - Luk H Vandenberghe
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute, Massachusetts Eye and Ear, Boston, Massachusetts.,Department of Ophthalmology, Harvard Medical School, Ocular Genomics Institute, Boston, Massachusetts.,The Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Susan L Hafenstein
- Department of Medicine, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania.,Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania.,Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, Pennsylvania
| |
Collapse
|
21
|
Abstract
Parvoviruses are structurally simple viruses with linear single-stranded DNA genomes and nonenveloped icosahedral capsids. They infect a wide range of animals from insects to humans. Parvovirus B19 is a long-known human pathogen, whereas adeno-associated viruses are nonpathogenic. Since 2005, many parvoviruses have been discovered in human-derived samples: bocaviruses 1-4, parvovirus 4, bufavirus, tusavirus, and cutavirus. Some human parvoviruses have already been shown to cause disease during acute infection, some are associated with chronic diseases, and others still remain to be proven clinically relevant-or harmless commensals, a distinction not as apparent as it might seem. One initially human-labeled parvovirus might not even be a human virus, whereas another was originally overlooked due to inadequate diagnostics. The intention of this review is to follow the rocky road of emerging human parvoviruses from discovery of a DNA sequence to current and future clinical status, highlighting the perils along the way.
Collapse
|
22
|
Das P, Chatterjee K, Chattopadhyay NR, Choudhuri T. Evolutionary aspects of Parvovirus B-19V associated diseases and their pathogenesis patterns with an emphasis on vaccine development. Virusdisease 2019; 30:32-42. [PMID: 31143830 PMCID: PMC6517593 DOI: 10.1007/s13337-019-00525-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 03/15/2019] [Indexed: 12/16/2022] Open
Abstract
Parvovirus B-19, a single human pathogenic member of the Parvoviridae family with it's small ssDNA and non-enveloped structure, is known to cause diseases in erythroid progenitor cells. But a wide range of clinical association of this virus with cells of non-erythroid origins has recently been discovered and many of those are being investigated for such association. Higher substitution rates in due course of evolution has been suggested for this cellular tropism and persistence. In this report, we have summarized the different disease manifestations of B-19 virus and have tried to find out a pattern of pathogenesis. Finally, we have focused on the vaccination strategies available against B-19 virus to correlate these with the mechanisms involved in various diseases caused by this virus.
Collapse
Affiliation(s)
- Piyanki Das
- Department of Biotechnology, Visva Bharati Santiniketan, Siksha Bhavana, Bolpur, West Bengal 731235 India
| | - Koustav Chatterjee
- Department of Biotechnology, Visva Bharati Santiniketan, Siksha Bhavana, Bolpur, West Bengal 731235 India
| | - Nabanita Roy Chattopadhyay
- Department of Biotechnology, Visva Bharati Santiniketan, Siksha Bhavana, Bolpur, West Bengal 731235 India
| | - Tathagata Choudhuri
- Department of Biotechnology, Visva Bharati Santiniketan, Siksha Bhavana, Bolpur, West Bengal 731235 India
| |
Collapse
|
23
|
Brenner N, Butt J, Bomfim IL, Tabatabai J, Pawlita M, Schnitzler P, Waterboer T. Validation of monoplex assays detecting antibodies against Corynebacterium diphtheriae and Clostridium tetani toxins, rubella virus and parvovirus B19 for incorporation into Multiplex Serology. Methods 2019; 158:44-53. [PMID: 30703462 DOI: 10.1016/j.ymeth.2019.01.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 12/27/2018] [Accepted: 01/23/2019] [Indexed: 01/12/2023] Open
Abstract
Serological assays detecting antibodies in serum or plasma samples are useful and versatile instruments to investigate an individual's infection and vaccination history, e.g. for clinical diagnosis, personal risk evaluation, and seroepidemiological studies. Multiplex Serology is a suspension bead array-based high-throughput methodology for simultaneous measurement of antibodies against multiple pathogens in a single reaction vessel, thus economizing sample volume, measurement time, and costs. We developed and validated bead-based pathogen-specific Monoplex Serology assays, i.e. assays including only antigens for the respective pathogen, to detect antibodies against Corynebacterium diphtheriae and Clostridium tetani toxins, rubella virus and parvovirus B19. The developed assays expand the portfolio of existing pathogen-specific bead-based serology assays and can be efficiently incorporated into larger Multiplex Serology panels. The newly developed Monoplex Serology assays consist of only one antigen per infectious agent, expressed as Glutathione S-transferase-fusion proteins in E. coli. Specificity, sensitivity and Cohen's kappa statistics in comparison with routine clinical diagnostic assays were calculated for serum dilutions 1:100 and 1:1000. All pathogen-specific assays were successfully validated at both serum dilutions with the exception of rubella Monoplex Serology which showed impaired sensitivity (57.6%) at dilution 1:1000. Specificities of successfully validated Monoplex Serology assays ranged from 85.6% to 100.0% (median: 91.7%), and sensitivities from 81.3% to 95.8% (median: 90.9%); agreement with the reference assays ranged from substantial to almost perfect (kappa: 0.66-0.86, median: 0.78). Statistical performance and slim assay design enable efficient incorporation of the developed assays into Multiplex Serology.
Collapse
Affiliation(s)
- Nicole Brenner
- Infections and Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, Heidelberg, Germany.
| | - Julia Butt
- Infections and Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany.
| | - Izaura Lima Bomfim
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Julia Tabatabai
- Center for Infectious Diseases, Virology, University Hospital of Heidelberg, Heidelberg, Germany.
| | - Michael Pawlita
- Infections and Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany.
| | - Paul Schnitzler
- Center for Infectious Diseases, Virology, University Hospital of Heidelberg, Heidelberg, Germany.
| | - Tim Waterboer
- Infections and Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany.
| |
Collapse
|
24
|
|
25
|
Bustos-Jaimes I, Soto-Román RA, Gutiérrez-Landa IA, Valadez-García J, Segovia-Trinidad CL. Construction of protein-functionalized virus-like particles of parvovirus B19. J Biotechnol 2017; 263:55-63. [PMID: 28935566 DOI: 10.1016/j.jbiotec.2017.09.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 08/25/2017] [Accepted: 09/17/2017] [Indexed: 11/17/2022]
Abstract
Decoration of virus-like particles (VLPs) expands the repertory of functions these particles can display. In the last years, VLPs have successfully been used as scaffolds to present different molecules, frequently through the specific reaction of chemical groups on the surface of the particles, or by protein engineering when the presentation of peptides or proteins is the primary goal. VLPs of parvovirus B19 (B19V), have been previously produced in vitro and its stability and ability to assemble into hybrid particles composed of wild-type and chimeric proteins evidenced their potential as research tools. Herein, we report the presentation of functional proteins on the surface of B19V VLPs, through the fusion of the gene coding for the heterologous protein within the gene coding for the structural protein VP2. Two model proteins were used for the construction of chimeras, a lipase from Bacillus pumilus (BplA) and the enhanced green fluorescent protein (EGFP). Both chimeras were folded and successfully assembled in vitro into VLPs. While the BplA chimera exhibited esterase activity, the chimera of EGFP showed no fluorescence. We replaced the EGFP by its fast-folding derivative "super folder GFP" (sfGFP) flanked by larger linkers to increase its movement freedom, which resulted in fluorescent protein able to assemble fluorescent VLPs. These results expand the toolbox for VLP decoration as well as for the construction of new nanobiomaterials.
Collapse
Affiliation(s)
- Ismael Bustos-Jaimes
- Department of Biochemistry, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico.
| | - Ricardo Arturo Soto-Román
- Department of Biochemistry, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico
| | | | - Josefina Valadez-García
- Department of Biochemistry, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico
| | | |
Collapse
|
26
|
Rahpeyma M, Samarbaf-Zadeh A, Makvandi M, Ghadiri AA, Dowall SD, Fotouhi F. Expression and characterization of codon-optimized Crimean-Congo hemorrhagic fever virus Gn glycoprotein in insect cells. Arch Virol 2017; 162:1951-1962. [PMID: 28316015 DOI: 10.1007/s00705-017-3315-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 02/22/2017] [Indexed: 11/28/2022]
Abstract
Crimean-Congo hemorrhagic fever virus (CCHFV) is a major cause of tick-borne viral hemorrhagic disease in the world. Despite of its importance as a deadly pathogen, there is currently no licensed vaccine against CCHF disease. The attachment glycoprotein of CCHFV (Gn) is a potentially important target for protective antiviral immune responses. To characterize the expression of recombinant CCHFV Gn in an insect-cell-based system, we developed a gene expression system expressing the full-length coding sequence under a polyhedron promoter in Sf9 cells using recombinant baculovirus. Recombinant Gn was purified by affinity chromatography, and the immunoreactivity of the protein was evaluated using sera from patients with confirmed CCHF infection. Codon-optimized Gn was successfully expressed, and the product had the expected molecular weight for CCHFV Gn glycoprotein of 37 kDa. In time course studies, the optimum expression of Gn occurred between 36 and 48 hours postinfection. The immunoreactivity of the recombinant protein in Western blot assay against human sera was positive and was similar to the results obtained with the anti-V5 tag antibody. Additionally, mice were subjected to subcutaneous injection with recombinant Gn, and the cellular and humoral immune response was monitored. The results showed that recombinant Gn protein was highly immunogenic and could elicit high titers of antigen-specific antibodies. Induction of the inflammatory cytokine interferon-gamma and the regulatory cytokine IL-10 was also detected. In conclusion, a recombinant baculovirus harboring CCHFV Gn was constructed and expressed in Sf9 host cells for the first time, and it was demonstrated that this approach is a suitable expression system for producing immunogenic CCHFV Gn protein without any biosafety concerns.
Collapse
Affiliation(s)
- Mehdi Rahpeyma
- Infectious and Tropical Diseases Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Virology, WHO Collaborating Center for Reference and Research on Rabies, Pasteur Institute of Iran, Pasteur Institute, Tehran, Iran
| | - Alireza Samarbaf-Zadeh
- Infectious and Tropical Diseases Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Manoochehr Makvandi
- Department of Virology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ata A Ghadiri
- Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Stuart D Dowall
- National Infection Service, Public Health England, Porton Down, Wiltshire, UK
| | - Fatemeh Fotouhi
- Department of Influenza and Other Respiratory Viruses, Pasteur Institute of Iran, Pasteur Institute, Tehran, Iran.
| |
Collapse
|
27
|
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
|
28
|
Väisänen E, Paloniemi M, Kuisma I, Lithovius V, Kumar A, Franssila R, Ahmed K, Delwart E, Vesikari T, Hedman K, Söderlund-Venermo M. Epidemiology of two human protoparvoviruses, bufavirus and tusavirus. Sci Rep 2016; 6:39267. [PMID: 27966636 PMCID: PMC5155296 DOI: 10.1038/srep39267] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 11/21/2016] [Indexed: 01/19/2023] Open
Abstract
Two human parvoviruses were recently discovered by metagenomics in Africa, bufavirus (BuV) in 2012 and tusavirus (TuV) in 2014. These viruses have been studied exclusively by PCR in stool and detected only in patients with diarrhoea, although at low prevalence. Three genotypes of BuV have been identified. We detected, by in-house EIA, BuV1-3 IgG antibodies in 7/228 children (3.1%) and 10/180 adults (5.6%), whereas TuV IgG was found in one child (0.4%). All children and 91% of the adults were Finnish, yet interestingly 3/6 adults of Indian origin were BuV-IgG positive. By competition EIA, no cross-reactivity between the BuVs was detected, indicating that the BuV genotypes represent distinct serotypes. Furthermore, we analysed by BuV qPCR stool and nasal swab samples from 955 children with gastroenteritis, respiratory illness, or both, and found BuV DNA in three stools (0.3%) and for the first time in a nasal swab (0.1%). This is the first study documenting the presence of BuV and TuV antibodies in humans. Although the seroprevalences of both viruses were low in Finland, our results indicate that BuV infections might be widespread in Asia. The BuV-specific humoral immune responses appeared to be strong and long-lasting, pointing to systemic infection in humans.
Collapse
Affiliation(s)
- Elina Väisänen
- Department of Virology, University of Helsinki, Helsinki 00290, Finland
| | - Minna Paloniemi
- Vaccine Research Center, University of Tampere, Tampere 33520, Finland.,Fimlab laboratories ltd, Tampere 33520, Finland
| | - Inka Kuisma
- Department of Virology, University of Helsinki, Helsinki 00290, Finland
| | - Väinö Lithovius
- Department of Virology, University of Helsinki, Helsinki 00290, Finland
| | - Arun Kumar
- Department of Virology, University of Helsinki, Helsinki 00290, Finland.,Health Sciences North Research Institute, Sudbury, ON P3E 5J1, Canada
| | - Rauli Franssila
- Department of Virology, University of Helsinki, Helsinki 00290, Finland
| | - Kamruddin Ahmed
- Department of Pathobiology and Medical Diagnostics, Faculty of Medicine, Universiti Malaysia Sabah, Kota Kinabalu 88400, Sabah, Malaysia
| | - Eric Delwart
- Blood Systems Research Institute, San Francisco, CA 94118, USA.,Department of Laboratory Medicine, University of California at San Francisco, San Francisco, CA 94118, USA
| | - Timo Vesikari
- Vaccine Research Center, University of Tampere, Tampere 33520, Finland
| | - Klaus Hedman
- Department of Virology, University of Helsinki, Helsinki 00290, Finland.,Helsinki University Hospital, HUSLAB, Helsinki 00290, Finland
| | | |
Collapse
|
29
|
Del Carmen Morán-García A, Rivera-Toledo E, Echeverría O, Vázquez-Nin G, Gómez B, Bustos-Jaimes I. Peptide presentation on primate erythroparvovirus 1 virus-like particles: In vitro assembly, stability and immunological properties. Virus Res 2016; 224:12-8. [PMID: 27523978 DOI: 10.1016/j.virusres.2016.08.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 08/07/2016] [Accepted: 08/09/2016] [Indexed: 10/21/2022]
Abstract
Virus-like particles (VLPs) have demonstrated to be valuable scaffolds for the display of heterologous peptides for vaccine development and other specific interactions. VLPs of primate erythroparvovirus 1, generally referred as parvovirus B19 (B19V), have already been produced in-vivo and in-vitro from the recombinant VP2 protein of this virus. In this study, chimeric forms of B19V VP2 were constructed, and their ability to assemble into VLPs was evaluated. Chimeras were composed of the VP2 protein fused, at its N-terminus, with two peptides derived from the fusion glycoprotein (F) of the respiratory syncytial virus (RSV). The chimeric proteins self-assembled into VLPs morphologically similar to B19V virions. Stability of these VLPs was analyzed under denaturation conditions with guanidinium chloride (GdnHCl). Our results indicate that the presence of the heterologous fragments increased the stability of VLPs assembled by any of the VP2 chimeras. Specific proteolysis assays shown that a fraction of the N-termini of the chimeric proteins is located on the outer surface of the VLPs. Immunogenicity of VLPs against RSV was evaluated and the results indicate that the particles can elicit a humoral immune response, although these antibodies did not cross-react with RSV in ELISA tests. These results provide novel insights into the localization of the N-termini of B19V VP2 protein after in vitro assembly into VLPs, and point them to be attractive sites to display peptides or proteins without compromise the assembly or stability of VLPs.
Collapse
Affiliation(s)
- Areli Del Carmen Morán-García
- Department of Biochemistry, Faculty of Medicine, National Autonomous University of Mexico (UNAM), Mexico City 04510, Mexico
| | - Evelyn Rivera-Toledo
- Department of Microbiology and Parasitology, Faculty of Medicine, UNAM, Mexico City 04510, Mexico
| | - Olga Echeverría
- Department of Cell Biology, Faculty of Sciences, UNAM, Mexico City 04510, Mexico
| | - Gerardo Vázquez-Nin
- Department of Cell Biology, Faculty of Sciences, UNAM, Mexico City 04510, Mexico
| | - Beatriz Gómez
- Department of Microbiology and Parasitology, Faculty of Medicine, UNAM, Mexico City 04510, Mexico
| | - Ismael Bustos-Jaimes
- Department of Biochemistry, Faculty of Medicine, National Autonomous University of Mexico (UNAM), Mexico City 04510, Mexico.
| |
Collapse
|
30
|
Sánchez-Rodríguez SP, Morán-García ADC, Bolonduro O, Dordick JS, Bustos-Jaimes I. Enhanced assembly and colloidal stabilization of primate erythroparvovirus 1 virus-like particles for improved surface engineering. Acta Biomater 2016; 35:206-14. [PMID: 26911883 DOI: 10.1016/j.actbio.2016.02.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 01/27/2016] [Accepted: 02/17/2016] [Indexed: 11/18/2022]
Abstract
Virus-like particles (VLPs) are the product of the self-assembly, either in vivo or in vitro, of structural components of viral capsids. These particles are excellent scaffolds for surface display of biomolecules that can be used in vaccine development and tissue-specific drug delivery. Surface engineering of VLPs requires structural stability and chemical reactivity. Herein, we report the enhanced assembly, colloidal stabilization and fluorescent labeling of primate erythroparvovirus 1 (PE1V), generally referred to as parvovirus B19. In vitro assembly of the VP2 protein of PE1V produces VLPs, which are prone to flocculate and hence undergo limited chemical modification by thiol-specific reagents like the fluorogenic monobromobimane (mBBr). We determined that the addition of 0.2M l-arginine during the assembly process produced an increased yield of soluble VLPs with good dispersion stability. Fluorescent labeling of VLPs suspended in phosphate buffered saline (PBS) added with 0.2M l-Arg was achieved in significantly shorter times than the flocculated VLPs assembled in only PBS buffer. Finally, to demonstrate the potential application of this approach, mBBr-labeled VLPs were successfully used to tag human hepatoma HepG2 cells. This new method for assembly and labeling PE1V VLPs eases its applications and provides insights on the manipulation of this biomaterial for further developments. STATEMENT OF SIGNIFICANCE Application of virus-derived biomaterials sometimes requires surface modification for diverse purposes, including enhanced cell-specific interaction, the inclusion of luminescent probes for bioimaging, or the incorporation of catalytic properties for the production of enzyme nanocarriers. In this research, we reported for the first time the colloidal stabilization of the primate erythroparvovirus 1 (PE1V) virus-like particles (VLPs). Also, we report the chemical modification of the natural Cys residues located on the surface of these VLPs with a fluorescent probe, as well as its application for tagging hepatoma cells in vitro. Keeping in mind that PE1V is a human pathogen, virus-host interactions already exist in human cells, and they can be exploited for therapeutic and research aims. This study will impact on the speed in which the scientific community will be able to manipulate PE1V VLPs for diverse purposes. Additionally, this study may provide insights on the colloidal properties of these VLPs as well as in the effect of different protein additives used for protein stabilization.
Collapse
Affiliation(s)
- Sandra Paola Sánchez-Rodríguez
- Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA; Department of Biochemistry, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico
| | - Areli del Carmen Morán-García
- Department of Biochemistry, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico
| | - Olurotimi Bolonduro
- Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Jonathan S Dordick
- Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA; Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA; Department of Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Ismael Bustos-Jaimes
- Department of Biochemistry, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico.
| |
Collapse
|
31
|
Yeast-generated virus-like particles as antigens for detection of human bocavirus 1–4 specific antibodies in human serum. Appl Microbiol Biotechnol 2016; 100:4935-46. [DOI: 10.1007/s00253-016-7336-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 01/14/2016] [Accepted: 01/17/2016] [Indexed: 12/22/2022]
|
32
|
López-Sagaseta J, Malito E, Rappuoli R, Bottomley MJ. Self-assembling protein nanoparticles in the design of vaccines. Comput Struct Biotechnol J 2015; 14:58-68. [PMID: 26862374 PMCID: PMC4706605 DOI: 10.1016/j.csbj.2015.11.001] [Citation(s) in RCA: 249] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 11/10/2015] [Indexed: 01/09/2023] Open
Abstract
For over 100 years, vaccines have been one of the most effective medical interventions for reducing infectious disease, and are estimated to save millions of lives globally each year. Nevertheless, many diseases are not yet preventable by vaccination. This large unmet medical need demands further research and the development of novel vaccines with high efficacy and safety. Compared to the 19th and early 20th century vaccines that were made of killed, inactivated, or live-attenuated pathogens, modern vaccines containing isolated, highly purified antigenic protein subunits are safer but tend to induce lower levels of protective immunity. One strategy to overcome the latter is to design antigen nanoparticles: assemblies of polypeptides that present multiple copies of subunit antigens in well-ordered arrays with defined orientations that can potentially mimic the repetitiveness, geometry, size, and shape of the natural host-pathogen surface interactions. Such nanoparticles offer a collective strength of multiple binding sites (avidity) and can provide improved antigen stability and immunogenicity. Several exciting advances have emerged lately, including preclinical evidence that this strategy may be applicable for the development of innovative new vaccines, for example, protecting against influenza, human immunodeficiency virus, and respiratory syncytial virus. Here, we provide a concise review of a critical selection of data that demonstrate the potential of this field. In addition, we highlight how the use of self-assembling protein nanoparticles can be effectively combined with the emerging discipline of structural vaccinology for maximum impact in the rational design of vaccine antigens.
Collapse
Affiliation(s)
| | - Enrico Malito
- GlaxoSmithKline Vaccines S.r.l., Via Fiorentina 1, 53100 Siena, Italy
| | - Rino Rappuoli
- GlaxoSmithKline Vaccines S.r.l., Via Fiorentina 1, 53100 Siena, Italy
| | | |
Collapse
|
33
|
Qi Y, Fan J, Huang W, Zhao C, Wang Y, Kong FT, Kong W, Jiang C. Expression and characterization of hepatitis E virus-like particles and non-virus-like particles from insect cells. Biotechnol Appl Biochem 2015; 63:362-70. [PMID: 25824972 DOI: 10.1002/bab.1379] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 03/27/2015] [Indexed: 02/05/2023]
Abstract
The hepatitis E virus (HEV) capsid antigen expressed in insect cell has been proposed as a candidate subunit vaccine for the prevention of hepatitis E. However, the expression and purification of HEV virus-like particles (VLPs) from insect cells have not been explored. We aimed to optimize the procedure to obtain HEV VLPs. In this study, two conformations of the HEV capsid proteins were expressed in insect cells, VLPs and non-VLPs, and they were purified separately. The physicochemical properties and the humoral immune responses induced by the two forms were analyzed and compared. We found that HEV VLPs were more immunogenic in mice than HEV non-VLPs. Therefore, we optimized the conditions that yielded high VLPs expression in insect cell cultures and developed an efficient purification method. The results suggest that the distinction and isolation of VLPs from non-VLPs are essential to generate a more immunogenic vaccine.
Collapse
Affiliation(s)
- Ying Qi
- School of Life Science, Jinlin University, Changchun, People's Republic of China.,National Engineering Laboratory for AIDS Vaccine, Jilin University, Changchun, People's Republic of China.,Division of HIV/AIDS and Sexually-Transmitted Virus Vaccines of the National Institutes for Food and. Drug Control, Beijing, People's Republic of China
| | - Jinping Fan
- Division of HIV/AIDS and Sexually-Transmitted Virus Vaccines of the National Institutes for Food and. Drug Control, Beijing, People's Republic of China
| | - Weijin Huang
- Division of HIV/AIDS and Sexually-Transmitted Virus Vaccines of the National Institutes for Food and. Drug Control, Beijing, People's Republic of China
| | - Chenyan Zhao
- Division of HIV/AIDS and Sexually-Transmitted Virus Vaccines of the National Institutes for Food and. Drug Control, Beijing, People's Republic of China
| | - Youchun Wang
- Division of HIV/AIDS and Sexually-Transmitted Virus Vaccines of the National Institutes for Food and. Drug Control, Beijing, People's Republic of China
| | - Franklin T Kong
- Ann Arbor Pioneer High School, 601 West Stadium Blvd, Ann Arbor
| | - Wei Kong
- School of Life Science, Jinlin University, Changchun, People's Republic of China.,National Engineering Laboratory for AIDS Vaccine, Jilin University, Changchun, People's Republic of China
| | - Chunlai Jiang
- School of Life Science, Jinlin University, Changchun, People's Republic of China.,National Engineering Laboratory for AIDS Vaccine, Jilin University, Changchun, People's Republic of China
| |
Collapse
|
34
|
Gil-Ranedo J, Hernando E, Riolobos L, Domínguez C, Kann M, Almendral JM. The Mammalian Cell Cycle Regulates Parvovirus Nuclear Capsid Assembly. PLoS Pathog 2015; 11:e1004920. [PMID: 26067441 PMCID: PMC4466232 DOI: 10.1371/journal.ppat.1004920] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 04/28/2015] [Indexed: 12/02/2022] Open
Abstract
It is unknown whether the mammalian cell cycle could impact the assembly of viruses maturing in the nucleus. We addressed this question using MVM, a reference member of the icosahedral ssDNA nuclear parvoviruses, which requires cell proliferation to infect by mechanisms partly understood. Constitutively expressed MVM capsid subunits (VPs) accumulated in the cytoplasm of mouse and human fibroblasts synchronized at G0, G1, and G1/S transition. Upon arrest release, VPs translocated to the nucleus as cells entered S phase, at efficiencies relying on cell origin and arrest method, and immediately assembled into capsids. In synchronously infected cells, the consecutive virus life cycle steps (gene expression, proteins nuclear translocation, capsid assembly, genome replication and encapsidation) proceeded tightly coupled to cell cycle progression from G0/G1 through S into G2 phase. However, a DNA synthesis stress caused by thymidine irreversibly disrupted virus life cycle, as VPs became increasingly retained in the cytoplasm hours post-stress, forming empty capsids in mouse fibroblasts, thereby impairing encapsidation of the nuclear viral DNA replicative intermediates. Synchronously infected cells subjected to density-arrest signals while traversing early S phase also blocked VPs transport, resulting in a similar misplaced cytoplasmic capsid assembly in mouse fibroblasts. In contrast, thymidine and density arrest signals deregulating virus assembly neither perturbed nuclear translocation of the NS1 protein nor viral genome replication occurring under S/G2 cycle arrest. An underlying mechanism of cell cycle control was identified in the nuclear translocation of phosphorylated VPs trimeric assembly intermediates, which accessed a non-conserved route distinct from the importin α2/β1 and transportin pathways. The exquisite cell cycle-dependence of parvovirus nuclear capsid assembly conforms a novel paradigm of time and functional coupling between cellular and virus life cycles. This junction may determine the characteristic parvovirus tropism for proliferative and cancer cells, and its disturbance could critically contribute to persistence in host tissues. Cellular and viral life cycles are connected through multiple, though poorly understood, mechanisms. Parvoviruses infect humans and a broad spectrum of animals, causing a variety of diseases, but they are also used in experimental cancer therapy and serve as vectors for gene therapy. Parvoviruses can only multiply in proliferating cells providing essential replicative and transcriptional functions. However, it is unknown whether the cell cycle regulatory machinery may also control parvovirus assembly. We found that the nuclear translocation of parvovirus MVM capsid subunits (VPs) was highly dependent on physiological cell cycle regulations in mammalian fibroblasts, including: quiescence, progression through G1/S boundary, DNA synthesis, and cell to cell contacts. VPs nuclear translocation was significantly more sensitive to cell cycle controls than viral genome replication and gene expression. The results support nuclear capsid assembly as the major driving process of parvoviruses biological hallmarks, such as pathogenesis in proliferative tissues and tropism for cancer cells. In addition, disturbing the tight coupling of parvovirus assembly with the cell cycle may determine viral persistence in quiescent and post-mitotic host tissues. These findings may contribute to understand cellular regulations on the assembly of other nuclear eukaryotic viruses, and to develop cell cycle-based avenues for antiviral therapy.
Collapse
Affiliation(s)
- Jon Gil-Ranedo
- Centro de Biología Molecular "Severo Ochoa" (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Cantoblanco, Madrid, Spain
| | - Eva Hernando
- Centro de Biología Molecular "Severo Ochoa" (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Cantoblanco, Madrid, Spain
| | - Laura Riolobos
- Centro de Biología Molecular "Severo Ochoa" (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Cantoblanco, Madrid, Spain
| | - Carlos Domínguez
- Centro de Biología Molecular "Severo Ochoa" (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Cantoblanco, Madrid, Spain
| | - Michael Kann
- University of Bordeaux, Microbiologie Fondamentale et Pathogénicité, UMR 5234, Bordeaux, France
- CNRS, Microbiologie Fondamentale et Pathogénicité, UMR 5234, Bordeaux, France
- Centre Hospitalier Universitaire de Bordeaux, Service de Virologie, Bordeaux, France
| | - José M. Almendral
- Centro de Biología Molecular "Severo Ochoa" (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Cantoblanco, Madrid, Spain
- * E-mail:
| |
Collapse
|
35
|
Santillán-Uribe JS, Valadez-García J, Morán-García ADC, Santillán-Uribe HC, Bustos-Jaimes I. Peptide display on a surface loop of human parvovirus B19 VP2: Assembly and characterization of virus-like particles. Virus Res 2015; 201:1-7. [DOI: 10.1016/j.virusres.2015.02.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 02/09/2015] [Accepted: 02/10/2015] [Indexed: 11/16/2022]
|
36
|
Sánchez-Rodríguez SP, Enrriquez-Avila JV, Soto-Fajardo JM, Peña-Montes C, Bustos-Jaimes I. In Vitro Encapsulation of Heterologous dsDNA Into Human Parvovirus B19 Virus-Like Particles. Mol Biotechnol 2014; 57:309-17. [DOI: 10.1007/s12033-014-9823-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
37
|
Lavrentyeva IN, Antipova AY. HUMAN PARVOVIRUS В19: VIRUS CHARACTERISTICS, DISTRIBUTION AND DIAGNOSTICS OF PARVOVIRUS INFECTION. ACTA ACUST UNITED AC 2014. [DOI: 10.15789/2220-7619-2013-4-311-322] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
38
|
Zhang WP, Yang H, Chen H, Zhu HR, Lei Q, Song YH, Dai ZM, Sun JS, Jiang LL, Nie ZG. Gene expression analysis of potential genes and pathways involved in the pathogenic mechanisms of parvovirus B19 in human colorectal cancer. Oncol Lett 2014; 8:523-532. [PMID: 25013465 PMCID: PMC4081382 DOI: 10.3892/ol.2014.2151] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 04/10/2014] [Indexed: 11/24/2022] Open
Abstract
In order to investigate the pathogenic mechanisms of parvovirus B19 in human colorectal cancer, plasmids containing the VP1 or VP2 viral capsid proteins or the NS1 non-structural proteins of parvovirus B19 were constructed and transfected into primary human colorectal epithelial cells and LoVo cells. Differential gene expression was detected using a human genome expression array. Functional gene annotation analyses were performed using Database for Annotation, Visualization and Integrated Discovery v6.7 software. Gene ontology (GO) analyses revealed that VP1-related functions included the immune response, immune system process, defense response and the response to stimulus, while NS1-associated functions were found to include organelle fission, nuclear division, mitosis, the M-phase of the mitotic cell cycle, the mitotic cell cycle, M-phase, cell cycle phase, cell cycle process and cell division. Pathway expression analysis revealed that VP1-associated pathways included cell adhesion molecules, antigen processing and presentation, cytokines and the inflammatory response. Moreover, NS1-associated pathways included the cell cycle, pathways in cancer, colorectal cancer, the wnt signaling pathway and focal adhesion. Among the differential genes detected in the present study, 12 genes were found to participate in general cancer pathways and six genes were observed to participate in colorectal cancer pathways. NS1 is a key molecule in the pathogenic mechanism of parvovirus B19 in colorectal cancer. Several GO categories, pathways and genes were selected and may be the key targets through which parvovirus B19 participates in colorectal cancer pathogenesis.
Collapse
Affiliation(s)
- Wei-Ping Zhang
- Department of Gastroenterology and Hepatology, Urumqi Military General Hospital, Urumqi, Xinjiang 830000, P.R. China
| | - Hua Yang
- Department of Outpatients, Urumqi Military General Hospital, Urumqi, Xinjiang 830000, P.R. China
| | - Hong Chen
- Department of Blood Transfusion, Urumqi Military General Hospital, Urumqi, Xinjiang 830000, P.R. China
| | - Hai-Rong Zhu
- Department of Medical Administration, Urumqi Military General Hospital, Urumqi, Xinjiang 830000, P.R. China
| | - Quan Lei
- Department of Medical Administration, Urumqi Military General Hospital, Urumqi, Xinjiang 830000, P.R. China
| | - Yun-Hong Song
- Department of Outpatients, Urumqi Military General Hospital, Urumqi, Xinjiang 830000, P.R. China
| | - Zhong-Ming Dai
- Department of Gastroenterology and Hepatology, Urumqi Military General Hospital, Urumqi, Xinjiang 830000, P.R. China
| | - Jing-Shan Sun
- Department of Gastroenterology and Hepatology, Urumqi Military General Hospital, Urumqi, Xinjiang 830000, P.R. China
| | - Li-Li Jiang
- Department of Gastroenterology and Hepatology, Urumqi Military General Hospital, Urumqi, Xinjiang 830000, P.R. China
| | - Zhan-Guo Nie
- Department of Gastroenterology and Hepatology, Urumqi Military General Hospital, Urumqi, Xinjiang 830000, P.R. China
| |
Collapse
|
39
|
Molino JVD, Viana Marques DDA, Júnior AP, Mazzola PG, Gatti MSV. Different types of aqueous two-phase systems for biomolecule and bioparticle extraction and purification. Biotechnol Prog 2013; 29:1343-53. [DOI: 10.1002/btpr.1792] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 06/28/2013] [Indexed: 12/19/2022]
Affiliation(s)
- João Vitor Dutra Molino
- Dept. of Biochemical and Pharmaceutical Technology; Faculty of Pharmaceutical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 580, Block 16, Cidade Universitária; São Paulo 05508-000 Brazil
| | - Daniela de Araújo Viana Marques
- Dept. of Biochemical and Pharmaceutical Technology; Faculty of Pharmaceutical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 580, Block 16, Cidade Universitária; São Paulo 05508-000 Brazil
| | - Adalberto Pessoa Júnior
- Dept. of Biochemical and Pharmaceutical Technology; Faculty of Pharmaceutical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 580, Block 16, Cidade Universitária; São Paulo 05508-000 Brazil
| | - Priscila Gava Mazzola
- Dept. of Clinical Patology; Faculty of Medical Sciences; University of Campinas, Rua: Tessália Vieira de Camargo, 126, Cidade Universitária "Zeferino Vaz, Campinas; São Paulo 13083-887 Brazil
| | - Maria Silvia Viccari Gatti
- Genetics; Evolution and Bioagents Dept.; Biology Institute; University of Campinas, Rua: Monteiro Lobato, 255, Cidade Universitária “Zeferino Vaz,” Campinas; São Paulo 13083-862 Brazil
| |
Collapse
|
40
|
Fernandes F, Teixeira AP, Carinhas N, Carrondo MJT, Alves PM. Insect cells as a production platform of complex virus-like particles. Expert Rev Vaccines 2013; 12:225-36. [PMID: 23414412 DOI: 10.1586/erv.12.153] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Virus-like particles (VLPs) are multiprotein structures that resemble the conformation of native viruses but lack a viral genome, potentiating their application as safer and cheaper vaccines. The production of VLPs has been strongly linked with the use of insect cells and the baculovirus expression vector system, especially those particles composed of two or more structural viral proteins. In fact, this expression platform has been extensively improved over the years to address the challenges of coexpression of multiple proteins and their proper assembly into complexes in the same cell. In this article, the role of insect cell technology in the development and production of complex VLPs is overviewed; recent achievements, current bottlenecks and future trends are also highlighted.
Collapse
Affiliation(s)
- Fabiana Fernandes
- ITQB-Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | | | | | | | | |
Collapse
|
41
|
Hsu TC, Tsai CC, Chiu CC, Hsu JD, Tzang BS. Exacerbating effects of human parvovirus B19 NS1 on liver fibrosis in NZB/W F1 mice. PLoS One 2013; 8:e68393. [PMID: 23840852 PMCID: PMC3695896 DOI: 10.1371/journal.pone.0068393] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 05/29/2013] [Indexed: 12/27/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disorder with unknown etiology that impacts various organs including liver. Recently, human parvovirus B19 (B19) is recognized to exacerbate SLE. However, the effects of B19 on liver in SLE are still unclear. Herein we aimed to investigate the effects of B19 on liver in NZB/W F1 mice by injecting subcutaneously with PBS, recombinant B19 NS1, VP1u or VP2, respectively. Our experimental results revealed that B19 NS1 protein significantly enhanced the TGF-β/Smad fibrotic signaling by increasing the expressions of TGF-β, Smad2/3, phosphorylated Smad2/3, Smad4 and Sp1. The consequent fibrosis-related proteins, PAI-1 and α-SMA, were also significantly induced in livers of NZB/W F1 mice receiving B19 NS1 protein. Accordingly, markedly increased collagen deposition was also observed in livers of NZB/W F1 mice receiving B19 NS1 protein. However, no significant difference was observed in livers of NZB/W F1 mice receiving B19 VP1u or VP2 as compared to the controls. These findings indicate that B19 NS1 plays a crucial role in exacerbating liver fibrosis in NZB/W F1 mice through enhancing the TGF-â/Smad fibrotic signaling.
Collapse
Affiliation(s)
- Tsai-Ching Hsu
- Institute of Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
| | - Chun-Chou Tsai
- Institute of Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
| | - Chun-Ching Chiu
- Institute of Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
- Department of Neurology and Department of Medical Intensive Care Unit, Chunghua Christian Hospital, Chunghua, Taiwan
| | - Jeng-Dong Hsu
- Department of Pathology, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Pathology, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Bor-Show Tzang
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
- Department of Biochemistry, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Institute of Biochemistry and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
- * E-mail:
| |
Collapse
|
42
|
Abstract
Parvovirus B19 is a widespread human pathogenic virus, member of the Erythrovirus genus in the Parvoviridae family. Infection can be associated with an ample range of pathologies and clinical manifestations, whose characteristics and outcomes depend on the interplay between the pathogenetic potential of the virus, its adaptation to different cellular environments, and the physiological and immune status of the infected individuals. The scope of this review is the advances in knowledge on the biological characteristics of the virus and of virus-host relationships; in particular, the interactions of the virus with different cellular environments in terms of tropism and ability to achieve a productive replicative cycle, or, on the contrary, to establish persistence; the consequences of infection in terms of interference with the cell physiology; the process of recognition of the virus by the innate or adaptive immune system, hence the role of the immune system in controlling the infection or in the development of clinical manifestations. Linked to these issues is the continuous effort to develop better diagnostic algorithms and methods and the need for development of prophylactic and therapeutic options for B19V infections.
Collapse
Affiliation(s)
- Giorgio Gallinella
- Department of Pharmacy and Biotechnology, University of Bologna, and Microbiology, S.Orsola-Malpighi Hospital, Via Massarenti 9, 40138 Bologna, Italy
| |
Collapse
|
43
|
Tsai CC, Chiu CC, Hsu JD, Hsu HS, Tzang BS, Hsu TC. Human parvovirus B19 NS1 protein aggravates liver injury in NZB/W F1 mice. PLoS One 2013; 8:e59724. [PMID: 23555760 PMCID: PMC3605340 DOI: 10.1371/journal.pone.0059724] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Accepted: 02/21/2013] [Indexed: 01/28/2023] Open
Abstract
Human parvovirus B19 (B19) has been associated with a variety of diseases. However, the influence of B19 viral proteins on hepatic injury in SLE is still obscure. To elucidate the effects of B19 viral proteins on livers in SLE, recombinant B19 NS1, VP1u or VP2 proteins were injected subcutaneously into NZB/W F1 mice, respectively. Significant expressions of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) were detected in NZB/W F1 mice receiving B19 NS1 as compared to those mice receiving PBS. Markedly hepatocyte disarray and lymphocyte infiltration were observed in livers from NZB/WF 1 mice receiving B19 NS1 as compared to those mice receiving PBS. Additionally, significant increases of Tumor Necrosis Factor -α (TNF-α), TNF-α receptor, IκB kinase -α (IKK-α), nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor (IκB) and nuclear factor-kappa B (NF-κB) were detected in livers from NZB/W F1 mice receiving B19 NS1 as compared to those mice receiving PBS. Accordingly, significant increases of matrix metalloproteinase-9 (MMP9) and U-plasminogen activator (uPA) were also detected in livers from NZB/W F1 mice receiving B19 NS1 as compared to those mice receiving PBS. Contrarily, no significant variation on livers from NZB/W F1 mice receiving B19 VP1u or VP2 was observed as compared to those mice receiving PBS. These findings firstly demonstrated the aggravated effects of B19 NS1 but not VP1u or VP2 protein on hepatic injury and provide a clue in understanding the role of B19 NS1 on hepatic injury in SLE.
Collapse
Affiliation(s)
- Chun-Chou Tsai
- Institute of Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
| | - Chun-Ching Chiu
- Institute of Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
- Department of Neurology and Department of Medical Intensive Care Unit, Chunghua Christian Hospital, Chunghua, Taiwan
| | - Jeng-Dong Hsu
- Department of Pathology, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Pathology, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Huai-Sheng Hsu
- Institute of Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
| | - Bor-Show Tzang
- Department of Biochemistry, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Institute of Biochemistry and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
- * E-mail: (BST); (TCH)
| | - Tsai-Ching Hsu
- Institute of Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
- * E-mail: (BST); (TCH)
| |
Collapse
|
44
|
Dembinski J. Diagnosis of in utero Parvovirus B19 infection and maternal immune response - the relevance of linear epitopes and advanced serologic testing. ACTA ACUST UNITED AC 2013; 4:139-48. [PMID: 23484447 DOI: 10.1517/17530050903452190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
IMPORTANCE OF THE FIELD Parvovirus B19 (B19V) infection in utero causing fetal anemia and non-immune hydrops fetalis (NIHF) is a potentially life-threatening event for the fetus. Postexpositional non-invasive diagnosis is based on maternal IgG/IgM response and detection of viral genome in maternal blood. Serologic testing directs prenatal follow-up. Fetal infection is confirmed by polymerase chain reaction or in situ hybridization in fetal blood and/or amniotic fluid cells. The performance of serologic tests is significant in order to direct pre- and perinatal care at rational use of resources. Timing of diagnostic procedures and knowledge of the time course of infection in pregnant, asymptomatic women are critical. IgM negative testing in the presence of prolonged viremia may complicate individual risk analysis in pregnancy. Recently, advanced IgG avidity assays and epitope-type specific assays (IgG ETS EIA) have been re-evaluated. AREAS COVERED IN THIS REVIEW Epidemiology, clinical relevance and management of B19V infection in pregnancy. A review of the current literature (November 1984 - May 2009) and evaluation of current information on performance and predictive value of molecular and VP1/VP2 antigen-based IgG tests directed at the diagnosis of materno-fetal B19V infection and detection of past immunity. New aspects of B19V-associated fetal disease other than anemia/NIHF are also covered. WHAT THE READER WILL GAIN An overview of immunology and clinical relevance of B19V infection in pregnancy, of the potential value of advanced serologic testing and fields of future research. TAKE HOME MESSAGE In the absence of a commercially available vaccine, serologic tests remain important tools in individual risk analysis of pregnant women exposed to B19V. Sequential application of IgG avidity and IgG ETS EIAs may improve risk stratification and timing of invasive testing in B19V-exposed pregnancies, in particular with IgM-negativity and/or persistent DNAemia. Prospective evaluation of these test systems correlated to fetal outcome in order to reduce fetal morbidity and mortality as well as the overall burden of disease of B19V with regard to fetal malformation may be subject to future research.
Collapse
Affiliation(s)
- Jörg Dembinski
- Clinic of Pediatrics, Klinikum Itzehoe, Academic Teaching Hospital of Universitätsklinikum Hamburg Eppendorf UKE / Universitätsklinikum Schleswig-Holstein UKSH +49 0 4821 772 2201 ; +49 0 4821 772 2209 ;
| |
Collapse
|
45
|
Mihály I, Trethon A, Arányi Z, Lukács A, Kolozsi T, Prinz G, Marosi A, Lovas N, Dobner IS, Prinz G, Szalai Z, Pék T. [Observations on human parvovirus B19 infection diagnosed in 2011]. Orv Hetil 2013. [PMID: 23204301 DOI: 10.1556/oh.2012.29447] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
INTRODUCTION The incidence of human parvovirus B19 infection is unknown. AIM A retrospective analysis of clinical and laboratory findings was carried out in patients diagnosed with human parvovirus B19 infection in 2011 in a virologic laboratory of a single centre in Hungary. METHODS Clinical and laboratory data of patients with proven human parvovirus B19 infection were analysed using in- and out-patient files. RESULTS In 2011, 72 patients proved to have human parvovirus B19 infection with the use of enzyme immunoassay. The clinical diagnoses of these patients were as follows: human parvovirus B19 infection (30.6%), transient aplastic crisis (16.7%), arthritis (8.3%) and acute hepatitis (4.1%). Symptoms of each of the four phases of the infection occurred in various combinations with the exception of the monophase of cheek exanthema. This occurred without the presence of other symptoms in some cases. Leading symptoms and signs were exanthema (in 74.6% of cases), haematological disorders (in 69% of cases), fever (in 54.9% of cases) and arthritis (in 33.8% of cases). Several atypical dermatological symptoms were also observed. Acute arthritis without exanthema was noted in 8 patients. Of the 72 patients with proven human parvovirus B19 infection there were 7 pregnant women, and one of them had hydrops foetalis resulting spontaneous abortion. In 16 patients (22.5%) human parvovirus B19 IgG was undetectable despite an optimal time for testing. CONCLUSION The observations of this study may contribute to a better recognition of clinical symptoms of human parvovirus B19 infection.
Collapse
Affiliation(s)
- Ilona Mihály
- Egyesített Szent István és Szent László Kórház-Rendelőintézet Mikrobiológiai Laboratóriumi Osztály, Virológiai Laboratórium Budapest.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Kushnir N, Streatfield SJ, Yusibov V. Virus-like particles as a highly efficient vaccine platform: diversity of targets and production systems and advances in clinical development. Vaccine 2012; 31:58-83. [PMID: 23142589 PMCID: PMC7115575 DOI: 10.1016/j.vaccine.2012.10.083] [Citation(s) in RCA: 417] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Revised: 10/13/2012] [Accepted: 10/25/2012] [Indexed: 12/16/2022]
Abstract
Virus-like particles (VLPs) are a class of subunit vaccines that differentiate themselves from soluble recombinant antigens by stronger protective immunogenicity associated with the VLP structure. Like parental viruses, VLPs can be either non-enveloped or enveloped, and they can form following expression of one or several viral structural proteins in a recombinant heterologous system. Depending on the complexity of the VLP, it can be produced in either a prokaryotic or eukaryotic expression system using target-encoding recombinant vectors, or in some cases can be assembled in cell-free conditions. To date, a wide variety of VLP-based candidate vaccines targeting various viral, bacterial, parasitic and fungal pathogens, as well as non-infectious diseases, have been produced in different expression systems. Some VLPs have entered clinical development and a few have been licensed and commercialized. This article reviews VLP-based vaccines produced in different systems, their immunogenicity in animal models and their status in clinical development.
Collapse
Affiliation(s)
- Natasha Kushnir
- Fraunhofer USA Center for Molecular Biotechnology, Newark, DE 19711, USA
| | | | | |
Collapse
|
47
|
Tsao EI, Mason MR, Cacciuttolo MA, Bowen SH, Folena-Wasserman G. Production of parvovirus B19 vaccine in insect cells co-infected with double baculoviruses. Biotechnol Bioeng 2012; 49:130-8. [PMID: 18623563 DOI: 10.1002/(sici)1097-0290(19960120)49:2<130::aid-bit2>3.0.co;2-r] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Recombinant human parvovirus B19 virus-like particles (VLPs), a candidate vaccine, were produced using the insect cell (Sf-9)-baculovirus (AcNPV) expression system. The synthesis and assembly of the particles in Sf-9 cells are directed by double infections with one recombinant virus (bacVP1) expressing the parvovirus minor viral protein VP1 and a second virus (bacVP2) expressing the major viral protein VP2. Previous animal studies demonstrated that the polypeptide composition of the VLPs strongly affects the elicitation of virus neutralizing antibodies. The key factor controlling the production of an immunologically potent product in bioreactors was identified to be the multiplicity of infection (MOI) of bacVP1 and bacVP2 used for infection. A probabilistic model, which correlates well with the experimental results, was employed to facilitate the selection of MOIs and to provide a better understanding of the baculovirus co-infection process. A novel production process based on secondary infections was developed to ensure product consistency and to simplify large-scale logistics. The effects of other critical process parameters, such as temperature, dissolved oxygen concentration, lactate concentration, cell concentration at infection, and harvest time, were also investigated. (c) 1996 John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- E I Tsao
- Department of Process Cell Culture and Fermentation, Medimmune, Inc., 35 West Watkins Mill Road, Gaithersburg, Maryland 20878
| | | | | | | | | |
Collapse
|
48
|
Seroprevalence of immunoglobulin G antibody to parvovirus B19 in Ontario. Can J Infect Dis 2012; 7:313-6. [PMID: 22514456 DOI: 10.1155/1996/941356] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/1995] [Accepted: 03/25/1996] [Indexed: 11/18/2022] Open
Abstract
The prevalence of antibody to parvovirus B19 was assessed in two populations. In a group of 494 residents from Ontario and the Maritimes, virus-specific immunoglobulin (Ig) M antibody, a marker of acute infection, was found throughout the year but was most prevalent during the late winter and early spring months. The overall prevalence of IgG antibody in this group was 30.3%. In an effort to examine age-specific prevalence in this population, a second group of sera from 210 pediatric patients at The Hospital for Sick Children, Toronto, Ontario and from Red Cross blood donors was tested for the presence of B19-specific IgG, and of these, 31.4% of the samples were positive. This prevalence varied from 3.3% in the under five-year-old age group to 66.7% in the 35- to 45-year-old age group. Eighty per cent of sera from females of this group were seropositive. This study provides insight into the prevalence of parvovirus B19 IgG antibody in the population.
Collapse
|
49
|
Application of baculovirus as a delivery vehicle for study of transcription and translation mechanism of parvovirus in non-permissive mammalian cells. J Virol Methods 2012; 183:1-7. [DOI: 10.1016/j.jviromet.2012.02.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Revised: 02/19/2012] [Accepted: 02/28/2012] [Indexed: 01/14/2023]
|
50
|
Sokolenko S, George S, Wagner A, Tuladhar A, Andrich JMS, Aucoin MG. Co-expression vs. co-infection using baculovirus expression vectors in insect cell culture: Benefits and drawbacks. Biotechnol Adv 2012; 30:766-81. [PMID: 22297133 PMCID: PMC7132753 DOI: 10.1016/j.biotechadv.2012.01.009] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Revised: 01/13/2012] [Accepted: 01/17/2012] [Indexed: 12/12/2022]
Abstract
The baculovirus expression vector system (BEVS) is a versatile and powerful platform for protein expression in insect cells. With the ability to approach similar post-translational modifications as in mammalian cells, the BEVS offers a number of advantages including high levels of expression as well as an inherent safety during manufacture and of the final product. Many BEVS products include proteins and protein complexes that require expression from more than one gene. This review examines the expression strategies that have been used to this end and focuses on the distinguishing features between those that make use of single polycistronic baculovirus (co-expression) and those that use multiple monocistronic baculoviruses (co-infection). Three major areas in which researchers have been able to take advantage of co-expression/co-infection are addressed, including compound structure-function studies, insect cell functionality augmentation, and VLP production. The core of the review discusses the parameters of interest for co-infection and co-expression with time of infection (TOI) and multiplicity of infection (MOI) highlighted for the former and the choice of promoter for the latter. In addition, an overview of modeling approaches is presented, with a suggested trajectory for future exploration. The review concludes with an examination of the gaps that still remain in co-expression/co-infection knowledge and practice.
Collapse
Affiliation(s)
- Stanislav Sokolenko
- Department of Chemical Engineering, Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, Ontario, Canada N2L 3G1
| | | | | | | | | | | |
Collapse
|