1
|
Suzuki H, Noguchi T, Matsugu N, Suzuki A, Kimura S, Onishi M, Kosaka M, Miyazato P, Morita E, Ebina H. Safety and immunogenicity of parvovirus B19 virus-like particle vaccine lacking phospholipase A2 activity. Vaccine 2022; 40:6100-6106. [PMID: 36114131 DOI: 10.1016/j.vaccine.2022.09.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/14/2022] [Accepted: 09/02/2022] [Indexed: 10/14/2022]
Abstract
Parvovirus B19 (B19) belongs to the Erythroparvovirus genus and is known to cause the fifth disease in children. Primary infection of pregnant women is associated with a high risk of hydrops fetalis and stillbirth due to severe fetal anemia. Virus-like particle (VLP) vaccine candidates for B19 have been developed, although none have been approved so far. The B19 phospholipase A2 domain (B19 PLA2), located in the VP1 unique region, is believed to be associated with adverse inflammatory reactions, and previous effective attempts to improve this vaccine modality inserted a mutation to impair the PLA2 activity of VLPs. In this study, we designed VLPs with a deletion mutant of PLA2 (⊿PLA2 B19 VLP), devoid of PLA2 activity, and confirmed their immunogenicity and safe use in vivo. These results were supported by the lack of histological inflammatory reactions at the site of immunization or the production of IL-6 in ⊿PLA2 B19 VLP-immunized mice, that were observed in mice immunized with B19 VLPs. CD4+ T cells from mice vaccinated with VLPs and B19-seropositive human samples were not activated by B19 PLA2 stimulation, suggesting that the B19 PLA2 domain does not constitute a major CD4+ T cell epitope. Most importantly, the ⊿PLA2 B19 VLPs induced neutralizing antibodies against B19, in levels similar to those found in B19-seropositive human samples, indicating that they could be used as a safe and effective vaccine candidate against B19.
Collapse
Affiliation(s)
- Hidehiko Suzuki
- Virus vaccine group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan; The Research Foundation for Microbial Diseases of Osaka University, Suita, Osaka, Japan
| | - Takafumi Noguchi
- Virus vaccine group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan; The Research Foundation for Microbial Diseases of Osaka University, Suita, Osaka, Japan
| | - Noriko Matsugu
- The Research Foundation for Microbial Diseases of Osaka University, Suita, Osaka, Japan
| | - Akio Suzuki
- The Research Foundation for Microbial Diseases of Osaka University, Suita, Osaka, Japan
| | - Sakika Kimura
- Department of Biochemistry and Molecular Biology, Faculty of Agriculture and Life Science, Hirosaki University, Hirosaki, Aomori, Japan
| | - Misa Onishi
- The Research Foundation for Microbial Diseases of Osaka University, Suita, Osaka, Japan
| | - Mitsuyo Kosaka
- Virus vaccine group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan; The Research Foundation for Microbial Diseases of Osaka University, Suita, Osaka, Japan
| | - Paola Miyazato
- Virus vaccine group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan; The Research Foundation for Microbial Diseases of Osaka University, Suita, Osaka, Japan
| | - Eiji Morita
- Department of Biochemistry and Molecular Biology, Faculty of Agriculture and Life Science, Hirosaki University, Hirosaki, Aomori, Japan
| | - Hirotaka Ebina
- Virus vaccine group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan; The Research Foundation for Microbial Diseases of Osaka University, Suita, Osaka, Japan; Virus vaccine group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan.
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
|
5
|
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
|
6
|
Leisi R, Di Tommaso C, Kempf C, Ros C. The Receptor-Binding Domain in the VP1u Region of Parvovirus B19. Viruses 2016; 8:61. [PMID: 26927158 PMCID: PMC4810251 DOI: 10.3390/v8030061] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 02/10/2016] [Accepted: 02/17/2016] [Indexed: 11/16/2022] Open
Abstract
Parvovirus B19 (B19V) is known as the human pathogen causing the mild childhood disease erythema infectiosum. B19V shows an extraordinary narrow tissue tropism for erythroid progenitor cells in the bone marrow, which is determined by a highly restricted uptake. We have previously shown that the specific internalization is mediated by the interaction of the viral protein 1 unique region (VP1u) with a yet unknown cellular receptor. To locate the receptor-binding domain (RBD) within the VP1u, we analyzed the effect of truncations and mutations on the internalization capacity of the recombinant protein into UT7/Epo cells. Here we report that the N-terminal amino acids 5-80 of the VP1u are necessary and sufficient for cellular binding and internalization; thus, this N-terminal region represents the RBD required for B19V uptake. Using site-directed mutagenesis, we further identified a cluster of important amino acids playing a critical role in VP1u internalization. In silico predictions and experimental results suggest that the RBD is structured as a rigid fold of three α-helices. Finally, we found that dimerization of the VP1u leads to a considerably enhanced cellular binding and internalization. Taken together, we identified the RBD that mediates B19V uptake and mapped functional and structural motifs within this sequence. The findings reveal insights into the uptake process of B19V, which contribute to understand the pathogenesis of the infection and the neutralization of the virus by the immune system.
Collapse
Affiliation(s)
- Remo Leisi
- Department of Chemistry and Biochemistry, University of Bern, Bern 3012, Switzerland.
| | - Chiarina Di Tommaso
- Department of Chemistry and Biochemistry, University of Bern, Bern 3012, Switzerland.
| | - Christoph Kempf
- Department of Chemistry and Biochemistry, University of Bern, Bern 3012, Switzerland.
- CSL Behring AG, Bern 3014, Switzerland.
| | - Carlos Ros
- Department of Chemistry and Biochemistry, University of Bern, Bern 3012, Switzerland.
- CSL Behring AG, Bern 3014, Switzerland.
| |
Collapse
|
7
|
Affiliation(s)
- Shweta Kailasan
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, Florida 32610
| | - Mavis Agbandje-McKenna
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, Florida 32610
| | - Colin R. Parrish
- Baker Institute for Animal Health and Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York 14853;
| |
Collapse
|
8
|
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
|
9
|
Generation of a parvovirus B19 vaccine candidate. Vaccine 2013; 31:3872-8. [DOI: 10.1016/j.vaccine.2013.06.062] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 05/24/2013] [Accepted: 06/19/2013] [Indexed: 11/22/2022]
|
10
|
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
|
11
|
The determinants for the enzyme activity of human parvovirus B19 phospholipase A2 (PLA2) and its influence on cultured cells. PLoS One 2013; 8:e61440. [PMID: 23596524 PMCID: PMC3626588 DOI: 10.1371/journal.pone.0061440] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Accepted: 03/08/2013] [Indexed: 12/03/2022] Open
Abstract
Human parvovirus B19 (B19V) is the causative agent of erythema infectiosum in humans. B19 infection also causes severe disease manifestations, such as chronic anemia in immunocompromised patients, aplastic crisis in patients with a high turnover rate of red blood cells, and hydrops fetalis in pregnant women. Although a secreted phospholipase A2 (PLA2) motif has been identified in the unique region of the B19V minor capsid protein VP1(VP1u), the determinants for its enzyme activity and its influences on host cells are not well understood. The purpose of this study was to investigate the contribution of the PLA2 motif and other regions of the VP1u to the PLA2 activity, to determine the cellular localization of the VP1u protein, and to examine the effects of VP1u on cellular cytokines. We found that in addition to the critical conserved and non-conserved amino acids within the VP1u PLA2 motif, amino acid residues outside the VP1u PLA2 motif are also important for the PLA2 activity. VP1u and various mutants all revealed a nucleo-cytoplasmic distribution. UT7-Epo cells treated with prokaryotic expressed VP1u or mutant proteins with PLA2 activity released a large amount of free fatty acid (FFA), and the cell morphological change occurred dramatically. However, neither free fatty acid nor cell morphology change occurred for cells treated with the mutants without PLA2 activity. The wild type and the VP1u mutants with the PLA2 activity also activated TNF-α promoter and upregulated the transcription activity of NF-κB in transfected cells. In addition, we found that the amino acids outside the PLA2 domain are critical for the viral PLA2 activity, and that these tested VP1u mutants did not affect the localization of the VP1u protein.
Collapse
|
12
|
Abstract
Parvoviruses package a ssDNA genome. Both nonpathogenic and pathogenic members exist, including those that cause fetal infections, encompassing the entire spectrum of virus phenotypes. Their small genomes and simple coding strategy has enabled functional annotation of many steps in the infectious life cycle. They assemble a multifunctional capsid responsible for cell recognition and the transport of the packaged genome to the nucleus for replication and progeny virus production. It is also the target of the host immune response. Understanding how the capsid structure relates to the function of parvoviruses provides a platform for recombinant engineering of viral gene delivery vectors for the treatment of clinical diseases, and is fundamental for dissecting the viral determinants of pathogenicity. This review focuses on our current understanding of parvovirus capsid structure and function with respect to the infectious life cycle.
Collapse
Affiliation(s)
- Sujata Halder
- Department of Biochemistry & Molecular Biology, Center for Structural Biology, The McKnight Brain Institute, College of Medicine, 1600 SW Archer Road, PO Box 100245, University of Florida, Gainesville, FL 32610, USA
| | - Robert Ng
- Department of Biochemistry & Molecular Biology, Center for Structural Biology, The McKnight Brain Institute, College of Medicine, 1600 SW Archer Road, PO Box 100245, University of Florida, Gainesville, FL 32610, USA
| | - Mavis Agbandje-McKenna
- Department of Biochemistry & Molecular Biology, Center for Structural Biology, The McKnight Brain Institute, College of Medicine, 1600 SW Archer Road, PO Box 100245, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
13
|
Plentz A, Modrow S. Diagnosis, management and possibilities to prevent parvovirus B19 infection in pregnancy. Future Virol 2011. [DOI: 10.2217/fvl.11.120] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Human parvovirus B19 (B19V) infection in pregnancy can cause severe fetal anemia and nonimmune hydrops fetalis, which may be associated with spontaneous abortion and fetal death. Approximately 30–40% of women of child-bearing age are not immune to B19V infection. The risk to fetal life is particularly high if maternal infection occurs during the first 20 weeks of gestation. In this article we intend to give an overview on the molecular biology, epidemiology and management of B19V infection during pregnancy. These data will be combined with an assessment of the clinical situation of the infected fetus and the possibilities for avoiding and/or preventing B19V infection in pregnant women. Currently B19V infection is the causative agent of one of the most frequently occurring infectious complications in pregnancy that endangers fetal life, and so the necessity to develop a preventive vaccine is discussed.
Collapse
Affiliation(s)
- Annelie Plentz
- Institut für Medizinische Mikrobiologie und Hygiene, Universität Regensburg, Franz-Josef-Strauß Allee 11, 93053 Regensburg, Germany
| | | |
Collapse
|
14
|
Molecular characterization of the newly identified human parvovirus 4 in the family Parvoviridae. Virology 2011; 422:59-69. [PMID: 22044541 DOI: 10.1016/j.virol.2011.09.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Revised: 09/15/2011] [Accepted: 09/27/2011] [Indexed: 01/27/2023]
Abstract
Human parvovirus 4 (PARV4) is an emerging human virus, and little is known about the molecular aspects of PARV4 apart from its incomplete genome sequence, which lacks information of the termini. We analyzed the gene expression profile of PARV4 using a nearly full-length HPV4 genome in a replication competent system in 293 cells. We found that PARV4 utilizes two promoters to transcribe non-structural protein- and structural protein-encoding mRNAs, respectively, which were polyadenylated at the right end of the genome. Three major proteins, including the large non-structural protein NS1a, whose mRNA is spliced, and capsid proteins VP1 and VP2, were detected. Additional functional analysis of the NS1a revealed its capability to induce cell cycle arrest at G2/M phase in ex vivo-generated human hematopoietic stem cells. Taken together, our characterization of the molecular features of PARV4 suggests that PARV4 represents a new genus in the family Parvoviridae.
Collapse
|
15
|
Nie X, Zhang G, Xu D, Sun X, Li Z, Li X, Zhang X, He F, Li Y. The VP1-unique region of parvovirus B19 induces myocardial injury in mice. ACTA ACUST UNITED AC 2010; 42:121-8. [PMID: 19883162 DOI: 10.3109/00365540903321580] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
As a common human pathogen, parvovirus B19 (B19V) has been shown to be associated with many heart diseases, such as myocarditis, cardiomyopathy and cardiopericarditis. The virus protein 1-unique region (VP1u) is critical to B19V infectivity, but its role in the pathogenesis of B19V-induced myocardial injury has not been well studied. In this study to investigate the effects ofVP1u on the host myocardium, we first expressed a recombinant VP1u protein in Escherichia coli, produced it on a large scale by high-volume fermentation, and purified it using the AKTA explorer 100 system. Following treatment of mice with the recombinant protein, we then examined changes in the morphology of the cardiac muscles, the titre of anti-VP1u protein antibodies, and a panel of heart functional protein markers. Our results show that VP1u alone is sufficient to elicit pathological and ultrastructural changes in the host myocardium, and to increase the levels of the functional enzymes aspartate aminotransferase (AST), lactate dehydrogenase (LDH), creatine kinase (CK), creatine kinase isoenzyme (CK-MB) and alpha-hydroxybutyric acid dehydrogenase (alpha-HBDH). The changes in myocardial pathology and myocardial zymogram indicate that the VP1u protein of B19V causes myocardial injury, and may largely contribute to the pathogenesis of B19V-induced heart diseases.
Collapse
Affiliation(s)
- Xiaojing Nie
- Department of Paediatrics, Xijing Hospital, Xi'an, Shaanxi, China
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Parrish CR. Structures and functions of parvovirus capsids and the process of cell infection. Curr Top Microbiol Immunol 2010; 343:149-76. [PMID: 20397069 DOI: 10.1007/82_2010_33] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
To infect a cell, the parvovirus or adeno-associated virus (AAV) genome must be delivered from outside the plasma membrane to the nucleus, and in the process, the capsid must follow a series of binding and trafficking steps and also undergo necessary changes that result in exposure or release the ssDNA genome at the appropriate time and place within the cell. The 25 nm parvovirus capsid is comprised of two or three forms of a single protein, and although it is robust and stable, it is still sufficiently flexible to allow the exposure of several internal components at appropriate times during cell infection. The capsid can also accommodate insertion of peptides into surface loops, and capsid proteins from different viral serotypes can be shuffled to create novel functional variants. The capsids of the different viruses bind to one or more cell receptors, and for at least some viruses, the insertion of additional or alternative receptor binding sequences or structures into the capsid can expand or redirect its tropism. The infection process after cell binding involves receptor-mediated endocytosis followed by viral trafficking through the endosomal systems. That endosomal trafficking may be complex and prolonged for hours or be relatively brief. Generally only a small proportion of the particles taken up enter the cytoplasm after altering the endosomal membrane through the activity of a VP1-encoded phospholipase A2 domain that becomes released to the outside of the viral particle. Modifications to the capsid that can occur within the endosome or cytoplasm include structural changes to expose internal components, ubiquination and proteosomal processing, and possible trafficking of particles on molecular motors. It is still not clear how the genomes enter the nucleus, but nuclear pore-dependent entry of particles or permeabilization of nuclear membranes have been proposed. Those processes control the infection, pathogenesis, and host ranges of the autonomous viruses and determine the effectiveness of gene therapy using AAV capsids.
Collapse
Affiliation(s)
- Colin R Parrish
- Baker Institute for Animal Health, Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
17
|
Analysis of nucleotide sequences of human parvovirus B19 genome reveals two different modes of evolution, a gradual alteration and a sudden replacement: a retrospective study in Sapporo, Japan, from 1980 to 2008. J Virol 2009; 83:10975-80. [PMID: 19710152 DOI: 10.1128/jvi.00273-09] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
There have been no long-term systematic analyses of the molecular epidemiology of human parvovirus B19 (B19V). We investigated the variations of nucleotide sequences of B19V strains collected in Sapporo, Japan, from 1980 to 2008. In that period, six outbreaks of erythema infectiosum occurred regularly at 5-year intervals. The B19V strains collected successively, regardless of the outbreak, were analyzed for nucleotide variation in the subgenomic NS1-VP1u junction. The isolated strains can be classified into 10 subgroups. Two patterns of change of endemic strains were observed. One was a dynamic replacement of strains that occurred almost every 10 years, and the other was a gradual change consisting of an accumulation of point mutations.
Collapse
|
18
|
Chehadeh W, Halim MA, Al-Nakib W. Antibody-mediated opsonization of red blood cells in parvovirus B19 infection. Virology 2009; 390:56-63. [PMID: 19450862 DOI: 10.1016/j.virol.2009.04.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2009] [Revised: 04/01/2009] [Accepted: 04/22/2009] [Indexed: 11/28/2022]
Abstract
Red blood cells (RBCs) express abundantly parvovirus B19 receptor, and their role in the dissemination or clearance of B19 infection is unknown. In this study, we report that in early, acute or persistent infection, B19 viremia is mostly associated with RBCs. The capacity of different patients' plasma or IgG to opsonize RBCs collected from patients with early B19 infection, was investigated. The highest opsonization activity was observed with plasma or IgG fractions from patients with past B19 infection. In contrast, IgG samples from patients with acute or persistent infection showed no or little opsonization activity. The depletion of antibodies specific to B19 VP1, but not VP2, from IgG samples, resulted in a significant suppression of opsonization. Furthermore, IgG samples preincubated with heated B19 particles exposing VP1-unique (VP1u) region were unable to opsonize RBCs. These observations clearly suggest a role for anti-VP1u IgG in the opsonization of RBC-bound B19 particles.
Collapse
Affiliation(s)
- Wassim Chehadeh
- Virology Unit, Department of Microbiology, Faculty of Medicine, Kuwait University, PO Box 24923, Safat 13310 Kuwait
| | | | | |
Collapse
|
19
|
Visualization of the externalized VP2 N termini of infectious human parvovirus B19. J Virol 2008; 82:7306-12. [PMID: 18508892 DOI: 10.1128/jvi.00512-08] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The structures of infectious human parvovirus B19 and empty wild-type particles were determined by cryoelectron microscopy (cryoEM) to 7.5-A and 11.3-A resolution, respectively, assuming icosahedral symmetry. Both of these, DNA filled and empty, wild-type particles contain a few copies of the minor capsid protein VP1. Comparison of wild-type B19 with the crystal structure and cryoEM reconstruction of recombinant B19 particles consisting of only the major capsid protein VP2 showed structural differences in the vicinity of the icosahedral fivefold axes. Although the unique N-terminal region of VP1 could not be visualized in the icosahedrally averaged maps, the N terminus of VP2 was shown to be exposed on the viral surface adjacent to the fivefold beta-cylinder. The conserved glycine-rich region is positioned between two neighboring, fivefold-symmetrically related VP subunits and not in the fivefold channel as observed for other parvoviruses.
Collapse
|
20
|
Kantola K, Hedman L, Allander T, Jartti T, Lehtinen P, Ruuskanen O, Hedman K, Söderlund-Venermo M. Serodiagnosis of human bocavirus infection. Clin Infect Dis 2008; 46:540-6. [PMID: 18199037 PMCID: PMC7107971 DOI: 10.1086/526532] [Citation(s) in RCA: 139] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Background. A new human-pathogenic parvovirus, human bocavirus (HBoV), has recently been discovered and associated with respiratory disease in small children. However, many patients have presented with low viral DNA loads, suggesting HBoV persistence and rendering polymerase chain reaction-based diagnosis problematic. Moreover, nothing is known of HBoV immunity. We examined HBoV-specific systemic B cell responses and assessed their diagnostic use in young children with respiratory disease. Patients and methods. Paired serum samples from 117 children with acute wheezing, previously studied for 16 respiratory viruses, were tested by immunoblot assays using 2 recombinant HBoV capsid antigens: the unique part of virus protein 1 and virus protein 2. Results. Virus protein 2 was superior to the unique part of virus protein 1 with respect to immunoreactivity. According to the virus protein 2 assay, 24 (49%) of 49 children who were positive for HBoV according to polymerase chain reaction had immunoglobulin (Ig) M antibodies, 36 (73%) had IgG antibodies, and 29 (59%) exhibited IgM antibodies and/or an increase in IgG antibody level. Of 22 patients with an increase in antibody levels, 20 (91%) had a high load of HBoV DNA in the nasopharynx, supporting the hypothesis that a high HBoV DNA load indicates acute primary infection, whereas a low load seems to be of less clinical significance. In a subgroup of patients who were previously determined to have acute HBoV infection (defined as a high virus load in the nasopharynx, viremia, and absence of other viral infections), 9 (100%) of 9 patients had serological evidence of primary infection. In the control group of 68 children with wheezing who had polymerase chain reaction results negative for HBoV in the nasopharynx, 9 (13%) had IgM antibodies, including 5 who displayed an increase in IgG antibody levels and were viremic. No cross-reactivity with human parvovirus B19 was detected. Conclusions. Respiratory infections due to HBoV are systemic, elicit B cell immune responses, and can be diagnosed serologically. Serological diagnoses correlate with high virus loads in the nasopharynx and with viremia. Serological testing is an accurate tool for disclosing the association of HBoV infection with disease.
Collapse
Affiliation(s)
- Kalle Kantola
- Department of Virology, Haartman Institute, University of Helsinki, Finland
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Filippone C, Zhi N, Wong S, Lu J, Kajigaya S, Gallinella G, Kakkola L, Söderlund-Venermo M, Young NS, Brown KE. VP1u phospholipase activity is critical for infectivity of full-length parvovirus B19 genomic clones. Virology 2008; 374:444-52. [PMID: 18252260 DOI: 10.1016/j.virol.2008.01.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2007] [Revised: 12/14/2007] [Accepted: 01/04/2008] [Indexed: 11/15/2022]
Abstract
Three full-length genomic clones (pB19-M20, pB19-FL and pB19-HG1) of parvovirus B19 were produced in different laboratories. pB19-M20 was shown to produce infectious virus. To determine the differences in infectivity, all three plasmids were tested by transfection and infection assays. All three clones were similar in viral DNA replication, RNA transcription, and viral capsid protein production. However, only pB19-M20 and pB19-HG1 produced infectious virus. Comparison of viral sequences showed no significant differences in ITR or NS regions. In the capsid region, there was a nucleotide sequence difference conferring an amino acid substitution (E176K) in the phospholipase A2-like motif of the VP1-unique (VP1u) region. The recombinant VP1u with the E176K mutation had no catalytic activity as compared with the wild-type. When this mutation was introduced into pB19-M20, infectivity was significantly attenuated, confirming the critical role of this motif. Investigation of the original serum from which pB19-FL was cloned confirmed that the phospholipase mutation was present in the native B19 virus.
Collapse
Affiliation(s)
- Claudia Filippone
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institute of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Drechsler MD, Obermeier I, Döring Y, Lackner KJ, Modrow S, von Landenberg P. Generation of multifunctional murine monoclonal antibodies specifically directed to the VP1unique region protein of human parvovirus B19. Immunobiology 2008; 213:511-7. [PMID: 18514753 DOI: 10.1016/j.imbio.2007.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2007] [Revised: 12/05/2007] [Accepted: 12/13/2007] [Indexed: 11/17/2022]
Abstract
Little is known about the VP1unique region (VP1u), a part of one major capsid protein of human parvovirus B19 (B19), concerning its involvement in viral replication and infection cycle. Showing a phospholipase A2 (PLA2)-like activity, which is discussed to be necessary for viral release from host cell, its precise function remains unclear. The purpose of this study was to generate multifunctional monoclonal antibodies (mabs) for different applications that may be useful in investigating VP1u's relevance. To establish antiVP1u antibodies, spleen cells from Balb/c mice immunized with purified recombinant viral protein were used for generating antibody-producing hybridoma cell lines. Usability of the antibodies was tested in enzyme-linked immunosorbent assay (ELISA), Western-blot analysis, immunofluorescence and an inhibition assay of enzymatic activity of PLA2. Three hybridoma cell lines secreting mab's specifically directed against the VP1u protein of B19 could be generated and functioned in every screening method used in this study. These antibodies are helpful tools for investigations in B19 research and diagnosis. Furthermore, the antibodies could help in gaining a deeper understanding of VP1u's role in viral replication and infection especially in the importance of its constitutive PLA2-like activity.
Collapse
Affiliation(s)
- Maik D Drechsler
- Institute of Clinical Chemistry and Laboratory Medicine, Johannes Gutenberg-Universität Mainz Klinikum, Langenbeckstrasse 1, 55131 Mainz, Germany
| | | | | | | | | | | |
Collapse
|
23
|
Waldman M, Kopp JB. Parvovirus-B19-associated complications in renal transplant recipients. ACTA ACUST UNITED AC 2007; 3:540-50. [PMID: 17895931 DOI: 10.1038/ncpneph0609] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2007] [Accepted: 06/19/2007] [Indexed: 01/08/2023]
Abstract
Parvovirus B19 is a common human pathogen, causing erythema infectiosum in children, hydrops fetalis in pregnant women, and transient aplastic crisis in patients with chronic hemolytic anemia. Immunosuppressed patients can fail to mount an effective immune response to B19, resulting in prolonged or persistent viremia. Renal transplant recipients can develop symptomatic B19 infections as a result of primary infection acquired via the usual respiratory route or via the transplanted organ, or because of reactivation of latent or persistent viral infection. The most common manifestations of B19 infection in immunosuppressed patients are pure red cell aplasia and other cytopenias. Thus, this diagnosis should be considered in transplant recipients with unexplained anemia and reticulocytopenia or pancytopenia. Collapsing glomerulopathy and thrombotic microangiopathy have been reported in association with B19 infection in renal transplant recipients, but a causal relationship has not been definitively established. Prompt diagnosis of B19 infection in the renal transplant recipient requires a high index of suspicion and careful selection of diagnostic tests, which include serologies and polymerase chain reaction. Most patients benefit from intravenous immunoglobulin therapy and/or alteration or reduction of immunosuppressive therapy. Conservative therapy might be sufficient in some cases.
Collapse
Affiliation(s)
- Meryl Waldman
- Kidney Disease Section, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20814-9692, USA.
| | | |
Collapse
|
24
|
Abstract
Infection with parvovirus B19 causes several clinical syndromes (fifth disease, transient aplastic crisis, pure red cell aplasia, and hydrops fetalis) and may contribute to other illnesses. B19 has been linked to renal disease in three settings: As a cause of acute glomerulopathy and as a cause of anemia in ESRD and kidney transplantation. Case reports implicate parvovirus in the pathogenesis of proliferative glomerulonephritis and collapsing glomerulopathy, but a causal relationship has not been established. A proposed role for B19 infection is based on the temporal association of renal findings with viral infection, positive serology, and identification of the viral genome in the glomerulus. Mechanisms may include cytopathic effects on glomerular epithelial cells and/or endothelial cells and glomerular deposition of immune complexes. Patients who require dialysis may have increased susceptibility to acute and chronic anemia after parvoviral infection. Factors that predispose this population to complications of B19 infection include impaired immune response, deficient erythropoietin production, and possibly decreased erythrocyte survival. The clinical burden of parvovirus B19 infection in renal transplant recipients may be underestimated; these individuals may develop persistent viremia as a result of a dysfunctional immune response. Chronic anemia and pure red blood cell aplasia are the most common complications of parvovirus infection in this population; the diagnosis should be considered in transplant recipients with unexplained anemia or pancytopenia. Allograft rejection and dysfunction have been reported in association with infection, but a cause-effect relationship has not been proved. Further investigation of the relationship between B19 and kidney disease is warranted.
Collapse
Affiliation(s)
- Meryl Waldman
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland 20892-1268, USA. merylw@.niddk.nih.gov
| | | |
Collapse
|
25
|
Tzang BS, Lee YJ, Yang TP, Tsay GJ, Shi JY, Tsai CC, Hsu TC. Induction of antiphospholipid antibodies and antiphospholipid syndrome-like autoimmunity in naive mice with antibody against human parvovirus B19 VP1 unique region protein. Clin Chim Acta 2007; 382:31-6. [PMID: 17451664 DOI: 10.1016/j.cca.2007.03.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2007] [Revised: 03/09/2007] [Accepted: 03/12/2007] [Indexed: 11/27/2022]
Abstract
BACKGROUND Previous studies have postulated a connection between human parvovirus B19 (B19) infection and anti-phospholipid antibodies (APhL). B19 infection and anti-phospholipid syndrome (APS) exhibit congruent symptoms. Recently, phospholipase A2 (PLA2)-like activity has been linked to the VP1 unique region (VP1u) of B19. However, the precise role of B19-VP1u in pathogenesis of autoimmunity is still obscure. METHODS To elucidate the roles of VP1u in B19 infection and autoimmunity, the reactivity of B19-VP1u proteins with various autoantibodies were evaluated by ELISA and immunoblotting. Rabbits were immunized with purified recombinant B19-VP1u protein to generate anti-sera. Absorption experiments were conducted to determine the binding specificity of rabbit anti-sera against B19-VP1u, cardiolipin (CL) and beta-2-glycoprotein I (beta2GPI). Moreover, the effects of passive transfer of polyclonal rabbit anti-B19-VP1u IgG antibodies on platelets, activated partial thromboplastin time (aPTT), and autoantibodies were assessed. RESULTS Autoantibodies against CL, beta2GPI, and phospholipid (PhL) in sera from patients with B19 infection, were cross-reactive with B19-VP1u. Consistently, sera from rabbits immunized with recombinant B19-VP1u protein displayed raised detectable immunoglobulins against B19-VP1u, CL, beta2GPI and PhL. Additionally, the mice immunized with anti-B19-VP1u IgG developed thrombocytopenia, prolongation of aPTT, and autoantibody against beta2GPI and PhL. CONCLUSIONS These experimental results suggested the association between B19-VP1u and production of anti-beta2GPI antibodies, APhL, and APS-like autoimmunity. Altogether, it may provide a clue in understanding the role of B19-VP1u in inducing autoantibodies and B19-associated APS manifestations.
Collapse
Affiliation(s)
- Bor-Show Tzang
- Institute of Biochemistry and Biotechnology, Chung Shan Medical University, Taichung, Taiwan, ROC
| | | | | | | | | | | | | |
Collapse
|
26
|
Tzang BS, Tsay GJ, Lee YJ, Li C, Sun YS, Hsu TC. The association of VP1 unique region protein in acute parvovirus B19 infection and anti-phospholipid antibody production. Clin Chim Acta 2006; 378:59-65. [PMID: 17169353 DOI: 10.1016/j.cca.2006.10.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2006] [Revised: 10/20/2006] [Accepted: 10/23/2006] [Indexed: 11/22/2022]
Abstract
BACKGROUND Previous studies have postulated a connection between human parvovirus B19 (B19) infection and anti-phospholipid antibodies (aPL). Recently, the phospholipase domain of B19 has been linked to B19-VP1 unique region (VP1u). To elucidate the roles of VP1u in B19 infection and aPL production, the major reactivity of anti-B19-VP1u, anti-cardiolipin antibody (aCL), and anti-beta2-glycoprotein I (beta2GPI) antibody was evaluated. METHODS Sera from 102 clinically suspected cases of B19 infection were analyzed by nested PCR and ELISA. Humoral responses of anti-B19-VP1u and anti-B19-VP1uD175A IgM/IgG antibodies, aCL and the anti-beta2GPI antibody were assessed by Western blot and ELISA. Absorption experiments were also performed to determine the binding specificity of immunoglobulins to B19-VP1u, CL and beta2GPI. RESULTS Sera from patients with the diagnostic pattern DNA+/IgM+/IgG+ had a high frequency (57%) for recognition of CL and beta2GPI. Furthermore, adsorption experiments were performed by adding purified B19-VP1u, which partially suppressed the reactivity of anti-B19VP1u to CL and beta2GPI. CONCLUSIONS Serum from patients with acute B19 infection has a high frequency in recognition of CL and beta2GPI, and the phospholipase domain observed in the B19-VP1u may have contributed to the production of aPL. These findings may provide a clue for understanding the roles of B19-VP1u in B19 infection and aPL production.
Collapse
Affiliation(s)
- Bor-Show Tzang
- Institute of Biochemistry and Biotechnology, Chung Shan Medical University, and Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
| | | | | | | | | | | |
Collapse
|
27
|
Ros C, Gerber M, Kempf C. Conformational changes in the VP1-unique region of native human parvovirus B19 lead to exposure of internal sequences that play a role in virus neutralization and infectivity. J Virol 2006; 80:12017-24. [PMID: 17020940 PMCID: PMC1676260 DOI: 10.1128/jvi.01435-06] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The unique region of the capsid protein VP1 (VP1u) of human parvovirus B19 (B19) elicits a dominant immune response and has a phospholipase A(2) (PLA(2)) activity, which is necessary for the infection. In contrast to the rest of the parvoviruses, the VP1u of B19 is thought to occupy an external position in the virion, making this region a promising candidate for vaccine development. By using a monoclonal antibody against the most-N-terminal portion of VP1u, we revealed that this region rich in neutralizing epitopes is not accessible in native capsids. However, exposure of capsids to increasing temperatures or low pH led to its progressive accessibility without particle disassembly. Although unable to bind free virus or to block virus attachment to the cell, the anti-VP1u antibody was neutralizing, suggesting that the exposure of the epitope and the subsequent virus neutralization occur only after receptor attachment. The measurement of the VP1u-associated PLA(2) activity of B19 capsids revealed that this region is also internal but becomes exposed in heat- and in low-pH-treated particles. In sharp contrast to native virions, the VP1u of baculovirus-derived B19 capsids was readily accessible in the absence of any treatment. These results indicate that stretches of VP1u of native B19 capsids harboring neutralizing epitopes and essential functional motifs are not external to the capsid. However, a conformational change renders these regions accessible and triggers the PLA(2) potential of the virus. The results also emphasize major differences in the VP1u conformation between natural and recombinant particles.
Collapse
Affiliation(s)
- Carlos Ros
- Department of Chemistry and Biochemistry, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland.
| | | | | |
Collapse
|
28
|
Lupescu A, Bock CT, Lang PA, Aberle S, Kaiser H, Kandolf R, Lang F. Phospholipase A2 activity-dependent stimulation of Ca2+ entry by human parvovirus B19 capsid protein VP1. J Virol 2006; 80:11370-80. [PMID: 16956939 PMCID: PMC1642163 DOI: 10.1128/jvi.01041-06] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Recent reports demonstrated an association of human parvovirus B19 with inflammatory cardiomyopathy (iCMP), which is accompanied by endothelial dysfunction. As intracellular Ca(2+) activity is a key regulator of cell function and participates in mechanisms leading to endothelial dysfunction, the present experiments explored the effects of the B19 capsid proteins VP1 and VP2. A secreted phospholipase A2 (PLA2)-like activity has been located in the VP1 unique region of the B19 minor capsid protein. As PLA2 has recently been shown to activate the store-operated or capacitative Ca(2+) channel I(CRAC), we analyzed the impact of the viral PLA2 motif on Ca(2+) entry. We cloned the VP1 and VP2 genes isolated from a patient suffering from fatal B19 iCMP into eukaryotic expression vectors. We also generated a B19 replication-competent plasmid to demonstrate PLA2 activity under the control of the complete B19 genome. After the transfection of human endothelial cells (HMEC-1), cytosolic Ca(2+) activity was determined by utilizing Fura-2 fluorescence. VP1 and VP2 expression did not significantly modify basal cytosolic Ca(2+) activity or the decline of cytosolic Ca(2+) activity following the removal of extracellular Ca(2+). However, expression of VP1 and of the full-length B19 clone, but not of VP2, significantly accelerated the increase of cytosolic Ca(2+) activity following the readdition of extracellular Ca(2+) in the presence of thapsigargin, indicating an activation of I(CRAC.) The effect of VP1 was mimicked by the PLA2 product lysophosphatidylcholine and abolished by an inactivating mutation of the PLA2-encoding region of the VP1 gene. Our observations point to the activation of Ca(2+) entry by VP1 PLA2 activity, an effect likely participating in the pathophysiology of B19 infection.
Collapse
Affiliation(s)
- Adrian Lupescu
- Physiologisches Institut, der Universität Tübingen, Gmelinstr. 5, D-72076 Tübingen, Germany
| | | | | | | | | | | | | |
Collapse
|
29
|
Sonntag F, Bleker S, Leuchs B, Fischer R, Kleinschmidt JA. Adeno-associated virus type 2 capsids with externalized VP1/VP2 trafficking domains are generated prior to passage through the cytoplasm and are maintained until uncoating occurs in the nucleus. J Virol 2006; 80:11040-54. [PMID: 16956943 PMCID: PMC1642181 DOI: 10.1128/jvi.01056-06] [Citation(s) in RCA: 198] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Common features of parvovirus capsids are open pores at the fivefold symmetry axes that traverse the virion shell. Upon limited heat treatment in vitro, the pores can function as portals to externalize VP1/VP2 protein N-terminal sequences which harbor infection-relevant functional domains, such as a phospholipase A(2) catalytic domain. Here we show that adeno-associated virus type 2 (AAV2) also exposes its VP1/VP2 N termini in vivo during infection, presumably in the endosomal compartment. This conformational change is influenced by treatment with lysosomotropic reagents. While incubation of cells with bafilomycin A1 reduced exposure of VP1/VP2 N termini, incubation with chloroquine stimulated externalization transiently. N-terminally located basic amino acid clusters with nuclear localization activity also become exposed in this process and are accessible on the virus capsid when it enters the cytoplasm. This is an obligatory step in AAV2 infection. However, a direct role of these sequences in nuclear translocation of viral capsids could not be determined by microinjection of wild-type or mutant viruses. This suggests that further modifications of the capsid have to take place in a precytoplasmic entry step that prepares the virus for nuclear entry. Microinjection of several capsid-specific antibodies into the cell nucleus blocked AAV2 infection completely, supporting the conclusion that AAV2 capsids bring the infectious genome into the nucleus.
Collapse
Affiliation(s)
- Florian Sonntag
- German Cancer Research Center, Infection and Cancer, Im Neuenheimer Feld 242, D-69120 Heidelberg, Germany
| | | | | | | | | |
Collapse
|
30
|
Zhi N, Mills IP, Lu J, Wong S, Filippone C, Brown KE. Molecular and functional analyses of a human parvovirus B19 infectious clone demonstrates essential roles for NS1, VP1, and the 11-kilodalton protein in virus replication and infectivity. J Virol 2006; 80:5941-50. [PMID: 16731932 PMCID: PMC1472615 DOI: 10.1128/jvi.02430-05] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In an attempt to experimentally define the roles of viral proteins encoded by the B19 genome in the viral life cycle, we utilized the B19 infectious clone constructed in our previous study to create two groups of B19 mutant genomes: (i) null mutants, in which either a translational initiation codon for each of these viral genes was substituted by a translational termination codon or a termination codon was inserted into the open reading frame by a frameshift; and (ii) a deletion mutant, in which half of the hairpin sequence was deleted at both the 5' and the 3' termini. The impact of these mutations on viral infectivity, DNA replication, capsid protein production, and distribution was systematically examined. Null mutants of the NS and VP1 proteins or deletion of the terminal hairpin sequence completely abolished the viral infectivity, whereas blocking expression of the 7.5-kDa protein or the putative protein X had no effect on infectivity in vitro. Blocking expression of the proline-rich 11-kDa protein significantly reduced B19 viral infectivity, and protein studies suggested that the expression of the 11-kDa protein was critical for VP2 capsid production and trafficking in infected cells. These findings suggest a previously unrecognized role for the 11-kDa protein, and together the results enhance our understanding of the key features of the B19 viral genome and proteins.
Collapse
Affiliation(s)
- Ning Zhi
- Hematology Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892-1202, USA.
| | | | | | | | | | | |
Collapse
|
31
|
Abstract
Human B19 erythrovirus is a ubiquitous viral pathogen, commonly infecting individuals before adulthood. As with all autonomous parvoviruses, its small single-stranded DNA genome is replicated with host cell machinery. While the mechanism of parvovirus genome replication has been studied in detail, the rate at which B19 virus evolves is unknown. By inferring the phylogenetic history and evolutionary dynamics of temporally sampled B19 sequences, we observed a surprisingly high rate of evolutionary change, at approximately 10(-4) nucleotide substitutions per site per year. This rate is more typical of RNA viruses and suggests that high mutation rates are characteristic of the Parvoviridae.
Collapse
|
32
|
Modrow S. Parvovirus B19: the causative agent of dilated cardiomyopathy or a harmless passenger of the human myocard? ERNST SCHERING RESEARCH FOUNDATION WORKSHOP 2006:63-82. [PMID: 16329658 DOI: 10.1007/3-540-30822-9_5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Parvovirus B19 infections may cause a widespread benign and self-limiting disease in children and adults known as erythema infectiosum (fifth disease). Several further manifestations are associated with B19 infections, such as arthralgias, arthritis, leucopenia and thrombocytopenia, anaemia and vasculitis and spontaneous abortion and hydrops fetalis in pregnant women. Persistent infections with continuous virus production may occur in immunocompetent as well as in immunosuppressed individuals. Parvovirus B19 infections have been frequently implicated as a cause or trigger of various forms of autoimmune diseases affecting joints, connective tissue and large and small vessels. Autoimmune neutropenia, thrombocytopenia and haemolytic anaemia are known as sequelae of B19 infections. The molecular basis of the autoimmune phenomena is unclear. Many patients with these long-lasting symptoms are not capable of eliminating the virus or controlling its propagation. Furthermore, latent viral genomes have been detected in cells of various organs and tissues by PCR. At present, it is not clear if these cells produce viral proteins and/or infectious B19 particles, if the virus genome can be reactivated to productive replication and if the presence of viral DNA indicates a causative role of parvovirus B19 with distinct diseases.
Collapse
Affiliation(s)
- S Modrow
- Institut für Medizinische Mikrobiologie und Hygiene, Universität Regensburg, Germany.
| |
Collapse
|
33
|
Mani B, Baltzer C, Valle N, Almendral JM, Kempf C, Ros C. Low pH-dependent endosomal processing of the incoming parvovirus minute virus of mice virion leads to externalization of the VP1 N-terminal sequence (N-VP1), N-VP2 cleavage, and uncoating of the full-length genome. J Virol 2006; 80:1015-24. [PMID: 16379002 PMCID: PMC1346861 DOI: 10.1128/jvi.80.2.1015-1024.2006] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Minute virus of mice (MVM) enters the host cell via receptor-mediated endocytosis. Although endosomal processing is required, its role remains uncertain. In particular, the effect of low endosomal pH on capsid configuration and nuclear delivery of the viral genome is unclear. We have followed the progression and structural transitions of DNA full-virus capsids (FC) and empty capsids (EC) containing the VP1 and VP2 structural proteins and of VP2-only virus-like particles (VLP) during the endosomal trafficking. Three capsid rearrangements were detected in FC: externalization of the VP1 N-terminal sequence (N-VP1), cleavage of the exposed VP2 N-terminal sequence (N-VP2), and uncoating of the full-length genome. All three capsid modifications occurred simultaneously, starting as early as 30 min after internalization, and all of them were blocked by raising the endosomal pH. In particles lacking viral single-stranded DNA (EC and VLP), the N-VP2 was not exposed and thus it was not cleaved. However, the EC did externalize N-VP1 with kinetics similar to those of FC. The bulk of all the incoming particles (FC, EC, and VLP) accumulated in lysosomes without signs of lysosomal membrane destabilization. Inside lysosomes, capsid degradation was not detected, although the uncoated DNA of FC was slowly degraded. Interestingly, at any time postinfection, the amount of structural proteins of the incoming virions accumulating in the nuclear fraction was negligible. These results indicate that during the early endosomal trafficking, the MVM particles are structurally modified by low-pH-dependent mechanisms. Regardless of the structural transitions and protein composition, the majority of the entering viral particles and genomes end in lysosomes, limiting the efficiency of MVM nuclear translocation.
Collapse
Affiliation(s)
- Bernhard Mani
- Department of Chemistry and Biochemistry, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland
| | | | | | | | | | | |
Collapse
|
34
|
Franssila R, Auramo J, Modrow S, Möbs M, Oker-Blom C, Käpylä P, Söderlund-Venermo M, Hedman K. T helper cell-mediated interferon-gamma expression after human parvovirus B19 infection: persisting VP2-specific and transient VP1u-specific activity. Clin Exp Immunol 2005; 142:53-61. [PMID: 16178856 PMCID: PMC1809488 DOI: 10.1111/j.1365-2249.2005.02886.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Human parvovirus B19 is a small non-enveloped DNA virus with an icosahedral capsid consisting of proteins of only two species, the major protein VP2 and the minor protein VP1. VP2 is contained within VP1, which has an additional unique portion (VP1u) of 227 amino acids. We determined the ability of eukaryotically expressed parvovirus B19 virus-like particles consisting of VP1 and VP2 in the ratio recommended for vaccine use, or of VP2 alone, to stimulate, in an HLA class II restricted manner, peripheral blood mononuclear cells (PBMC) to proliferate and to secrete interferon gamma (IFN-gamma) and interleukin (IL)-10 cytokines among recently and remotely B19 infected subjects. PBMC reactivity with VP1u was determined specifically with a prokaryotically expressed VP1u antigen. In general, B19-specific IFN-gamma responses were stronger than IL-10 responses in both recent and remote infection; however, IL-10 responses were readily detectable among both groups, with the exception of patients with relapsed or persisting symptoms who showed strikingly low IL-10 responses. Whereas VP1u-specific IFN-gamma responses were very strong among the recently infected subjects, the VP1u-specific IFN-gamma and IL-10 responses were virtually absent among the remotely infected subjects. The disappearance of VP1u-specific IFN-gamma expression is surprising, as B-cell immunity against VP1u is well maintained.
Collapse
Affiliation(s)
- R Franssila
- Department of Virology, Haartman Institute and HUCH Diagnostic, University of Helsinki, Finland.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Ros C, Baltzer C, Mani B, Kempf C. Parvovirus uncoating in vitro reveals a mechanism of DNA release without capsid disassembly and striking differences in encapsidated DNA stability. Virology 2005; 345:137-47. [PMID: 16242744 DOI: 10.1016/j.virol.2005.09.030] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2005] [Revised: 08/09/2005] [Accepted: 09/08/2005] [Indexed: 11/28/2022]
Abstract
The uncoating mechanism of parvoviruses is unknown. Their capsid robustness and increasing experimental data would suggest an uncoating mechanism without capsid disassembly. We have developed an in vitro system to detect and quantify viral DNA externalization and applied the assay on two parvoviruses with important differences in capsid structure, human B19 and minute virus of mice (MVM). Upon briefly treating the capsids to increasing temperatures, the viral genome became accessible in its full-length in a growing proportion of virions. Capsid disassembly started at temperatures above 60 degrees C for B19 and 70 degrees C for MVM. For both viruses, the externalization followed an all-or-nothing mechanism, without transitions exposing only a particular genomic region. However, the heat-induced DNA accessibility was remarkably more pronounced in B19 than in MVM. This difference was also evident under conditions mimicking endosomal acidification (pH 6.5 to 5), which triggered the externalization of B19-DNA but not of MVM-DNA. The externalized ssDNA was a suitable template for the full second-strand synthesis. Immunoprecipitation with antibodies against conformational epitopes and quantitative PCR revealed that the DNA externalized by heat was mostly dissociated from its capsid, however, the low pH-induced DNA externalization of B19 was predominantly capsid-associated. These results provide new insights into parvovirus uncoating suggesting a mechanism by which the full-length viral genome is released without capsid disassembly. The remarkable instability of the encapsidated B19 DNA, which is easily released from its capsid, would also explain the faster heat inactivation of B19 when compared to other parvoviruses.
Collapse
Affiliation(s)
- Carlos Ros
- Department of Chemistry and Biochemistry, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland.
| | | | | | | |
Collapse
|
36
|
Lindner J, Barabas S, Saar K, Altmann D, Pfister A, Fleck M, Deml L, Modrow S. CD4+ T-Cell Responses Against the VP1-Unique Region in Individuals with Recent and Persistent Parvovirus B19 Infection. ACTA ACUST UNITED AC 2005; 52:356-61. [PMID: 16316401 DOI: 10.1111/j.1439-0450.2005.00860.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
To date cellular immune responses against parvovirus B19 (B19) have not been studied extensively. The aim of this study was to examine the T-cell response against the VP1-unique region as the immunodominant part of the viral structural protein VP1 in individuals with different courses of B19 infection. Therefore, a group of 13 parvovirus-positive probands was separated into subgroups characterized for recent or acute, past or persistent infection by means of the presence of specific immunoglobulin (Ig)M and IgG isotypes and of viral DNA in blood and tissue. Transiently transfected B-cells expressing VP1-unique region were used in ELISpot assays to investigate T-cell responses directed against the VP1-unique region in peripheral blood mononuclear cells (PBMC) of individual donors. Significant numbers of interferon-gamma (IFN-gamma) secreting lymphocytes were detectable in PBMC of all individuals with recent, acute or persistent B19 infection, but not in PBMC of donors with past B19 infection and seronegative individuals. A more detailed analysis of IFN-gamma producing cells by intracellular cytokine staining by flow cytometry revealed, that CD4(+) T cells but not CD8(+) cytotoxic lymphocytes (CTL) were the major subpopulation of IFN-gamma producing cells. These data strongly suggest the need of virus protein production for the maintenance of VP1-unique region-specific CD4(+) T-helper cell responses in B19-infected individuals.
Collapse
Affiliation(s)
- J Lindner
- Institute of Medical Microbiology and Hygiene, University of Regensburg, 93053 Regensburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Grieger JC, Samulski RJ. Packaging capacity of adeno-associated virus serotypes: impact of larger genomes on infectivity and postentry steps. J Virol 2005; 79:9933-44. [PMID: 16014954 PMCID: PMC1181570 DOI: 10.1128/jvi.79.15.9933-9944.2005] [Citation(s) in RCA: 264] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2005] [Accepted: 04/08/2005] [Indexed: 11/20/2022] Open
Abstract
The limited packaging capacity of adeno-associated virus (AAV) precludes the design of vectors for the treatment of diseases associated with larger genes. Autonomous parvoviruses, such as minute virus of mice and B19, while identical in size (25 nm), are known to package larger genomes of 5.1 and 5.6 kb, respectively, compared to AAV genomes of 4.7 kb. One primary difference is the fact that wild-type (wt) AAV utilizes three capsid subunits instead of two to form the virion shell. In this study, we have characterized the packaging capacity of AAV serotypes 1 through 5 with and without the Vp2 subunit. Using reporter transgene cassettes that range in size from 4.4 to 6.0 kb, we determined that serotypes 1 through 5 with and without Vp2 could successfully package, replicate in, and transduce cells. Dot blot analysis established that packaging efficiency was similar for all vector cassettes and that the integrity of encapsidated genomes was intact regardless of size. Although physical characterization determined that virion structures were indistinguishable from wt, transduction experiments determined that all serotype vectors carrying larger genomes (5.3 kb and higher) transduced cells less efficiently (within a log) than AAV encapsidating wt size genomes. This result was not unique to reporter genes and was observed for CFTR vector cassettes ranging in size from 5.1 to 5.9 kb. No apparent advantage in packaging efficiency was observed when Vp2 was present or absent from the virion. Further analysis determined that a postentry step was responsible for the block in infection and specific treatment of cells upon infection with proteasome inhibitors increased transduction of AAV encapsidating larger DNA templates to wt levels, suggesting a preferential degradation of virions encapsidating larger-than-wt genomes. This study illustrates that AAV is capable of packaging and protecting recombinant genomes as large as 6.0 kb but the larger genome-containing virions are preferentially degraded by the proteasome and that this block can be overcome by the addition of proteasome inhibitors.
Collapse
Affiliation(s)
- Joshua C Grieger
- Curriculm in Genetics and Molecular Biology and Gene Therapy Center, University of North Carolina at Chapel Hill, 27599-7352, USA
| | | |
Collapse
|
38
|
Franssila R, Hedman K. T-helper cell-mediated interferon-γ, interleukin-10 and proliferation responses to a candidate recombinant vaccine for human parvovirus B19. Vaccine 2004; 22:3809-15. [PMID: 15315862 DOI: 10.1016/j.vaccine.2003.06.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2003] [Revised: 06/05/2003] [Accepted: 06/06/2003] [Indexed: 11/19/2022]
Abstract
Recombinantly expressed virus-like particles of human parvovirus B19 containing the two structural proteins VP1 and VP2 (VP1/2 capsids) or VP2 alone (VP2 capsids) elicit vigorous antibody responses in animal models, whereas only VP1/2 capsids elicit neutralizing antibodies. VP1 is, therefore, essential for protective B-cell immunity. In this study, we determined the ability of VP1/2 capsids containing VP1 and VP2 in the ratio recommended for vaccine use, and of sole VP2 capsids to stimulate T-helper (Th) cells to proliferate and to secrete interferon gamma (IF-gamma) and interleukin-10 (IL-10) in humans long after natural infection. Similar proliferation, IF-gamma and IL-10 responses were found with the VP1/2 and VP2 capsids. We conclude that, whereas VP1 contains important B-cell epitopes, VP2, the major structural protein of human parvovirus B19, appears to provide the major target for B19-specific Th-cells years or decades after natural infection.
Collapse
Affiliation(s)
- Rauli Franssila
- Department of Virology, Haartman Institute and HUCH Diagnostic, University of Helsinki, P.O. Box 21 (Haarmaninkatu 3), Helsinki 00014, Finland.
| | | |
Collapse
|
39
|
Chisaka H, Morita E, Yaegashi N, Sugamura K. Parvovirus B19 and the pathogenesis of anaemia. Rev Med Virol 2004; 13:347-59. [PMID: 14625883 DOI: 10.1002/rmv.395] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Human parvovirus B19 (B19) infection causes human bone marrow failure, by affecting erythroid-lineage cells which are well-known target cells for B19. The anaemia induced by B19 infection is of minor clinical significance in healthy children and adults, however, it becomes critical in those afflicted with haemolytic diseases. This condition is called transient aplastic crisis, and the pathogenesis is explained by the short life-span of red blood cells. Similarly, fetuses are thought to be severely affected by B19-intrauterine infection in the first and second trimester, as the half-life of red blood cells is apparently shorter than RBC at the bone marrow haematopoietic stage. On the other hand, B19 is also the causative agent of persistent anaemia in immunocompromised patients, transplant recipients and infants. The deficiencies of appropriate immune responses to B19 impair viral elimination in vivo, which results in enlargement of B19-infected erythroid-lineage cells. The B19-associated damage of erythroid lineage cells is due to cytotoxicity mediated by viral proteins. B19-infected erythroid-lineage cells show apoptotic features, which are thought to be induced by the non-structural protein, NS1, of B19. In addition, B19 infection induces cell cycle arrests at the G(1) and G(2) phases. The G(1) arrest is induced by NS1 expression prior to apoptosis induction in B19-infected cells, while the G(2) arrest is induced not only by infectious B19 but also by UV-inactivated B19, which lacks the ability to express NS1. In this review, we address the clinical manifestations and molecular mechanisms for B19-induced anaemia in humans and a mouse model, and of B19-induced cell cycle arrests in erythroid cells.
Collapse
Affiliation(s)
- Hiroshi Chisaka
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | | | | | | |
Collapse
|
40
|
Barah F, Vallely PJ, Cleator GM, Kerr JR. Neurological manifestations of human parvovirus B19 infection. Rev Med Virol 2003; 13:185-99. [PMID: 12740833 DOI: 10.1002/rmv.388] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Since its discovery, human parvovirus B19 has been linked with a broad spectrum of clinical syndromes. An aetiological role for the virus has been confirmed in erythema infectiosum, transient aplastic crisis, persistent infection manifesting as pure red cell aplasia in immunocompromised persons, non-immune hydrops fetalis and arthritis. Less commonly recognised, but receiving increasing attention recently, are the neurological manifestations, a variety of which have been described in patients with either clinically diagnosed or laboratory confirmed B19 infection. The purpose of this review is to summarise present knowledge of B19, its known and potential pathogenic mechanisms and its association with human diseases, particularly those with neurological manifestations. The outcome of the review supports an aetiological role of the virus in neurological disease. However, the pathogenesis remains unknown and elucidating this is a priority.
Collapse
Affiliation(s)
- Faraj Barah
- Laboratory Medicine Academic Group, University of Manchester, Clinical Sciences Building 3rd Floor, Manchester Royal Infirmary, Oxford Road, Manchester M13 9WL, UK.
| | | | | | | |
Collapse
|
41
|
Abstract
Parvovirus B19 (B19) was discovered in 1974 and is the only member of the family Parvoviridae known to be pathogenic in humans. Despite the inability to propagate the virus in cell cultures, much has been learned about the pathophysiology of this virus, including the identification of the cellular receptor (P antigen), and the control of the virus by the immune system. B19 is widespread, and manifestations of infection vary with the immunologic and hematologic status of the host. In healthy immunocompetent individuals B19 is the cause of erythema infectiosum and, particularly in adults, acute symmetric polyarthropathy. Due to the tropism of B19 to erythroid progenitor cells, infection in individuals with an underlying hemolytic disorder causes transient aplastic crisis. In the immunocompromised host persistent B19 infection is manifested as pure red cell aplasia and chronic anemia. Likewise, the immature immune response of the fetus may render it susceptible to infection, leading to fetal death in utero, hydrops fetalis, or development of congenital anemia. B19 has also been suggested as the causative agent in a variety of clinical syndromes, but given the common nature, causality is often difficult to infer. Diagnosis is primarily based on detection of specific antibodies by enzyme-linked immunosorbent assay or detection of viral DNA by dot blot hybridization or PCR. Treatment of persistent infection with immunoglobulin reduces the viral load and results in a marked resolution of anemia. Vaccine phase I trials show promising results.
Collapse
Affiliation(s)
- Erik D Heegaard
- Department of Clinical Microbiology, University State Hospital, Rigshospitalet, Copenhagen, Denmark
| | | |
Collapse
|
42
|
Abstract
Parvovirus B19 is the causative agent of erythema infectiosum. In addition, the infection may be associated with other disease manifestations: anemia and aplastic crisis, thrombo- or granulocytopenies; spontaneous abortion or hydrops fetalis in pregnant women; acute and chronic arthritis in adults and children, myocarditis and hepatitis. Both acute and persistent courses of B19-infections have been reported. All patients develop IgG against the capsid proteins VP1 and VP2, the majority of virus neutralizing antibodies that offer life-long protection against reinfections are directed against the VP1-unique region. IgM is mainly directed against VP2-specific epitopes. These antibodies may be present for only a rather short period of two to ten weeks after acute infection. IgG-antibodies against the nonstructural protein NS1 are preferentially found in patients which are unable to eliminate the virus and develop persisting viremia or virus persistence in distinct organs, e.g. synovial fluid, liver, bone marrow.
Collapse
Affiliation(s)
- Susanne Modrow
- Institut für Medizinische Mikrobiologie, Unversität Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany.
| | | |
Collapse
|
43
|
Vihinen-Ranta M, Wang D, Weichert WS, Parrish CR. The VP1 N-terminal sequence of canine parvovirus affects nuclear transport of capsids and efficient cell infection. J Virol 2002; 76:1884-91. [PMID: 11799183 PMCID: PMC135906 DOI: 10.1128/jvi.76.4.1884-1891.2002] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The unique N-terminal region of the parvovirus VP1 capsid protein is required for infectivity by the capsids but is not required for capsid assembly. The VP1 N terminus contains a number of groups of basic amino acids which resemble classical nuclear localization sequences, including a conserved sequence near the N terminus comprised of four basic amino acids, which in a peptide can act to transport other proteins into the cell nucleus. Testing with a monoclonal antibody recognizing residues 2 to 13 of VP1 (anti-VP1-2-13) and with a rabbit polyclonal serum against the entire VP1 unique region showed that the VP1 unique region was not exposed on purified capsids but that it became exposed after treatment of the capsids with heat (55 to 75 degrees C), or urea (3 to 5 M). A high concentration of anti-VP1-2-13 neutralized canine parvovirus (CPV) when it was incubated with the virus prior to inoculation of cells. Both antibodies blocked infection when injected into cells prior to virus inoculation, but neither prevented infection by coinjected infectious plasmid DNA. The VP1 unique region could be detected 4 and 8 h after the virus capsids were injected into cells, and that sequence exposure appeared to be correlated with nuclear transport of the capsids. To examine the role of the VP1 N terminus in infection, we altered that sequence in CPV, and some of those changes made the capsids inefficient at cell infection.
Collapse
Affiliation(s)
- Maija Vihinen-Ranta
- James A. Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, New York 14853, USA
| | | | | | | |
Collapse
|
44
|
Zádori Z, Szelei J, Lacoste MC, Li Y, Gariépy S, Raymond P, Allaire M, Nabi IR, Tijssen P. A viral phospholipase A2 is required for parvovirus infectivity. Dev Cell 2001; 1:291-302. [PMID: 11702787 DOI: 10.1016/s1534-5807(01)00031-4] [Citation(s) in RCA: 381] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Sequence analysis revealed phospholipase A2 (PLA2) motifs in capsid proteins of parvoviruses. Although PLA2 activity is not known to exist in viruses, putative PLA2s from divergent parvoviruses, human B19, porcine parvovirus, and insect GmDNV (densovirus from Galleria mellonella), can emulate catalytic properties of secreted PLA2. Mutations of critical amino acids strongly reduce both PLA2 activity and, proportionally, viral infectivity, but cell surface attachment, entry, and endocytosis by PLA2-deficient virions are not affected. PLA2 activity is critical for efficient transfer of the viral genome from late endosomes/lysosomes to the nucleus to initiate replication. These findings offer the prospect of developing PLA2 inhibitors as a new class of antiviral drugs against parvovirus infections and associated diseases.
Collapse
Affiliation(s)
- Z Zádori
- Centre de microbiologie et biotechnologie, INRS-Institut Armand-Frappier Université du Quebec, Laval, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Fifth (erythema infectiosum) and sixth (roseola infantum) diseases are common rash illnesses of childhood that have long been recognized in clinical medicine. The discovery of the viruses that cause these illnesses has revealed relationships with other syndromes. Primary infection with the agent of erythema infectiosum, human parvovirus B19, is associated with transient aplastic crisis in hemolytic anemia, arthropathy in adults, chronic anemia in immunocompromised patients, and nonimmune fetal hydrops in pregnant women. The only documented illness associated with primary infection with human herpesvirus 6 is roseola or exanthema subitum in young children. However, reactivated infections in adults and immunocompromised patients may be associated with serious illness such as encephalitis/encephalopathy, and bone marrow suppression leading to transplant failure or graft-versus-host disease. Diagnostic studies for both viruses have been limited, although reliable serologic tests for human parvovirus B19 have recently become available. Diagnosis of human herpesvirus 6 remains problematic, because current tests cannot differentiate primary from reactivated disease. This is more of an issue for the putative relationship of these viruses to more chronic conditions, such as rheumatologic disease for human parvovirus B19 and multiple sclerosis for human herpesvirus 6. The relationship between the viruses and these conditions remains controversial, and better diagnostic tests and further information on viral pathogenesis for both viruses are required in order to make a reliable judgment in this regard.
Collapse
Affiliation(s)
- W C Koch
- Department of Pediatrics, Division of Infectious Diseases, Medical College of Virginia of Virginia Commonwealth University, Richmond, Virginia 23298, USA.
| |
Collapse
|
46
|
Pereira RF, Paula WN, Cubel RDC, Nascimento JP. Anti-VP1 and anti-VP2 antibodies detected by immunofluorescence assays in patients with acute human parvovirus B19 infection. Mem Inst Oswaldo Cruz 2001; 96:507-13. [PMID: 11391423 DOI: 10.1590/s0074-02762001000400011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Acute human parvovirus B19 infection is followed by an antibody response to the structural proteins of the viral capsid (VP1 and VP2). We used 80 sera collected from 58 erythema infectiosum and 6 transient aplastic crisis patients to test IgM and IgG antibodies against these two proteins in an immunofluorescence assay (IFA) using Sf9 cells infected with recombinant baculovirus expressing either VP1 or VP2 antigen. Although less sensitive than IgM capture enzyme immunoassay using native antigen (MACEIA), we could detect anti-VP1 or anti-VP2 IgM antibodies by IFA in 49 patients with acute infection (76.6%). Detection of IgG anti-VP1 and anti-VP2 by IFA, however, was as sensitive as IgG detection by indirect enzyme immunoassay. By applying IgG avidity IFA to sera of the 15 IgM IFA negative patients we were able to confirm acute infection in further 12 cases by IFA. Overall, acute infection was confirmed by IFA in 61 (95.3%) of the 64 patients.
Collapse
Affiliation(s)
- R F Pereira
- Departamento de Microbiologia e Parasitologia, Instituto Biomédico, UFF, Niterói, RJ, Brasil
| | | | | | | |
Collapse
|
47
|
Hsu TC, Tsay GJ. Human parvovirus B19 infection in patients with systemic lupus erythematosus. Rheumatology (Oxford) 2001; 40:152-7. [PMID: 11257151 DOI: 10.1093/rheumatology/40.2.152] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE The clinical significance of the presence of B19 DNA in patients with SLE was studied. METHODS Sera from 72 patients with systemic lupus erythematosus (SLE), 23 patients with rheumatoid arthritis (RA), 18 patients with Sjögren's syndrome (SS), eight patients with Raynaud's phenomenon (RP), five patients with primary biliary cirrhosis (PBC), five patients with polymyositis (PM), four patients with erythema infectiosum (EI) and 22 normal controls were examined for parvovirus B19 (B19) infection by serological assays, nested PCR and Southern blotting. RESULTS Parvovirus B19 DNA was detected in 17 of 72 patients with SLE and in three of four patients with EI, but not in patients with other systemic rheumatic diseases. Of the 17 patients with B19 DNA, only one had IgG anti-B19 antibody and two had IgM anti-B19 antibodies, whereas IgG and IgM anti-B19 antibodies were detected in 27 (49.1%) and 21 (38.2%) of 55 SLE patients without B19 DNA respectively. All sera from the patients with EI contained both IgG and IgM anti-B19 antibodies. B19 DNA was found more commonly in sera from SLE patients without anti-B19 antibodies than in those with anti-B19 antibodies (P<0.05). CONCLUSIONS B19 infection in patients with SLE may be due to lack of anti-B19 antibodies because of either the immunocompromised nature of the host or the use of immunosuppressive drugs. There was a higher prevalence of hypocomplementaemia and RP in patients with parvovirus B19 viraemia than in those without parvovirus B19 viraemia.
Collapse
Affiliation(s)
- T C Hsu
- Department of Medicine and Institutes of Immunology and. Medicine, Chung Shan Medical and Dental College, Taichung, Taiwan
| | | |
Collapse
|
48
|
Dorsch S, Kaufmann B, Schaible U, Prohaska E, Wolf H, Modrow S. The VP1-unique region of parvovirus B19: amino acid variability and antigenic stability. J Gen Virol 2001; 82:191-199. [PMID: 11125172 DOI: 10.1099/0022-1317-82-1-191] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The unique region of structural protein VP1 of parvovirus B19 (erythrovirus B19) is important for eliciting neutralizing antibodies that are responsible for eliminating the virus from the peripheral blood and for inducing lifelong immunity. Neutralizing human MAbs bind a conformationally defined epitope spanning VP1 residues 30-42. The DNA sequence encoding the VP1-unique region was determined in parvovirus B19 isolated from peripheral blood and amniotic fluid of nine acutely infected pregnant women, five arthritis patients and two chronically infected children. The amino acid sequences of the VP1-unique region exhibited higher variability in comparison with other B19-specific proteins. To analyse the influence of amino acid variations on antibody binding and protein conformation, two variants of the VP1-unique region were selected and expressed in E. coli as intein-fusion proteins. The selected variants displayed a number of amino acid exchanges in the VP1-unique region and had mutations in the determined epitope and adjacent regions. After purification via affinity chromatography, the dissociation constants K(D) of VP1-specific human MAbs interacting with the variant antigens and a viral prototype of the VP1-unique region were determined with a quartz crystal microbalance biosensor. A value of 5.4 x 10(-8) M was determined for the prototype isolate pJB; the affinity constants for the variant VP1-unique regions were similar. Comparable values were obtained for interaction of antibodies with non-infectious VP1/VP2 capsids produced by recombinant baculovirus and with B19 virions from amniotic fluid. It is concluded that the conformation of the epitope is unaffected by mutations or the environment of the VP1-unique region in virus capsids.
Collapse
Affiliation(s)
- Simone Dorsch
- Institut für Medizinische Mikrobiologie und Hygiene, Universität Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany1
| | - Bärbel Kaufmann
- Institut für Physikalische Biochemie, Universität Potsdam, Im Biotechnologiepark, 14943 Luckenwalde, Germany2
| | - Uwe Schaible
- Institut für Medizinische Mikrobiologie und Hygiene, Universität Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany1
| | - Elke Prohaska
- Institut für Medizinische Mikrobiologie und Hygiene, Universität Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany1
| | - Hans Wolf
- Institut für Medizinische Mikrobiologie und Hygiene, Universität Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany1
| | - Susanne Modrow
- Institut für Medizinische Mikrobiologie und Hygiene, Universität Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany1
| |
Collapse
|
49
|
Fukada K, Matumoto K, Takakura F, Yamaki M, Sato H, Okochi K, Maeda Y. Four putative subtypes of human parvovirus B19 based on amino acid polymorphism in the C-terminal region of non-structural protein. J Med Virol 2000; 62:60-9. [PMID: 10935990 DOI: 10.1002/1096-9071(200009)62:1<60::aid-jmv10>3.0.co;2-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The nucleotide sequence of 10 isolates of human parvovirus B19 (B19) were determined and compared throughout 96.3% of the open reading frames (4145 nucleotides from nt. 509-4653). In the 4145 nucleotides, 122 mutation sites were found, of which 24 were accompanied by amino acid displacement. Furthermore, the polymorphism of the amino acids was seen in about 110 bases near the carboxy terminal of the non-structural protein, ranging from nt. 2011 to 2123, where four amino acid mutation points were found to exist. Based on the amino acid polymorphism of these four mutation sites in this area, 10 isolates of the B19 parvovirus could be divided into 4 subtypes (subtypes A, B, C, and D). The frequency of isolation of the subtypes depended on the time and location of collection of the B19 viremic blood specimens.
Collapse
Affiliation(s)
- K Fukada
- Fukuoka Red Cross Blood Center, Japan.
| | | | | | | | | | | | | |
Collapse
|
50
|
Affiliation(s)
- J R Kerr
- Department of Medical Microbiology and Virology, Manchester Royal Infirmary, Oxford Road, Manchester M13 9WL, UK.
| |
Collapse
|