1
|
Okba NMA, Raj VS, Widjaja I, GeurtsvanKessel CH, de Bruin E, Chandler FD, Park WB, Kim NJ, Farag EABA, Al-Hajri M, Bosch BJ, Oh MD, Koopmans MPG, Reusken CBEM, Haagmans BL. Sensitive and Specific Detection of Low-Level Antibody Responses in Mild Middle East Respiratory Syndrome Coronavirus Infections. Emerg Infect Dis 2019; 25:1868-1877. [PMID: 31423970 PMCID: PMC6759241 DOI: 10.3201/eid2510.190051] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV) infections in humans can cause asymptomatic to fatal lower respiratory lung disease. Despite posing a probable risk for virus transmission, asymptomatic to mild infections can go unnoticed; a lack of seroconversion among some PCR-confirmed cases has been reported. We found that a MERS-CoV spike S1 protein–based ELISA, routinely used in surveillance studies, showed low sensitivity in detecting infections among PCR-confirmed patients with mild clinical symptoms and cross-reactivity of human coronavirus OC43–positive serum samples. Using in-house S1 ELISA and protein microarray, we demonstrate that most PCR-confirmed MERS-CoV case-patients with mild infections seroconverted; nonetheless, some of these samples did not have detectable levels of virus-neutralizing antibodies. The use of a sensitive and specific serologic S1-based assay can be instrumental in the accurate estimation of MERS-CoV prevalence.
Collapse
|
2
|
Abstract
Human respiratory syncytial virus (RSV) is understood to be a significant human pathogen in infants, young children, and the elderly and the immunocompromised. Over the last decade many important mechanisms contributing to RSV infection, replication, and disease pathogenesis have been revealed; however, there is still insufficient knowledge which has in part hampered vaccine development. Considerable information is accumulating regarding how RSV proteins modulate molecular signaling and immune responses to infection. Understanding how RSV interacts with its host is crucial to facilitate the development of safe and effective vaccines and therapeutic treatments.In this chapter, we provide a brief introduction into RSV replication, pathogenesis, and host immune response, and summarize the state of RSV vaccine and antiviral compounds in clinical stages of development. This chapter frames features of this book and the molecular methods used for understanding RSV interaction with the host.
Collapse
Affiliation(s)
- Patricia A Jorquera
- Department of Infectious Diseases, College of Veterinary Medicine, Animal Health Research Center, University of Georgia, 111 Carlton Street, Athens, GA, 30602, USA
| | - Lydia Anderson
- Department of Infectious Diseases, College of Veterinary Medicine, Animal Health Research Center, University of Georgia, 111 Carlton Street, Athens, GA, 30602, USA
| | - Ralph A Tripp
- Department of Infectious Diseases, College of Veterinary Medicine, Animal Health Research Center, University of Georgia, 111 Carlton Street, Athens, GA, 30602, USA.
| |
Collapse
|
3
|
Enhanced Neutralizing Antibody Response Induced by Respiratory Syncytial Virus Prefusion F Protein Expressed by a Vaccine Candidate. J Virol 2015; 89:9499-510. [PMID: 26157122 DOI: 10.1128/jvi.01373-15] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 07/01/2015] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Respiratory syncytial virus (RSV) and human parainfluenza virus type 3 (HPIV3) are the first and second leading viral agents of severe respiratory tract disease in infants and young children worldwide. Vaccines are not available, and an RSV vaccine is particularly needed. A live attenuated chimeric recombinant bovine/human PIV3 (rB/HPIV3) vector expressing the RSV fusion (F) glycoprotein from an added gene has been under development as a bivalent vaccine against RSV and HPIV3. Previous clinical evaluation of this vaccine candidate suggested that increased genetic stability and immunogenicity of the RSV F insert were needed. This was investigated in the present study. RSV F expression was enhanced 5-fold by codon optimization and by modifying the amino acid sequence to be identical to that of an early passage of the original clinical isolate. This conferred a hypofusogenic phenotype that presumably reflects the original isolate. We then compared vectors expressing stabilized prefusion and postfusion versions of RSV F. In a hamster model, prefusion F induced increased quantity and quality of RSV-neutralizing serum antibodies and increased protection against wild-type (wt) RSV challenge. In contrast, a vector expressing the postfusion F was less immunogenic and protective. The genetic stability of the RSV F insert was high and was not affected by enhanced expression or the prefusion or postfusion conformation of RSV F. These studies provide an improved version of the previously well-tolerated rB/HPIV3-RSV F vaccine candidate that induces a superior RSV-neutralizing serum antibody response. IMPORTANCE Respiratory syncytial virus (RSV) and human parainfluenza virus type 3 (HPIV3) are two major causes of pediatric pneumonia and bronchiolitis. The rB/HPIV3 vector expressing RSV F protein is a candidate bivalent live vaccine against HPIV3 and RSV. Previous clinical evaluation indicated the need to increase the immunogenicity and genetic stability of the RSV F insert. Here, we increased RSV F expression by codon optimization and by modifying the RSV F amino acid sequence to conform to that of an early passage of the original isolate. This resulted in a hypofusogenic phenotype, which likely represents the original phenotype before adaptation to cell culture. We also included stabilized versions of prefusion and postfusion RSV F protein. Prefusion RSV F induced a larger quantity and higher quality of RSV-neutralizing serum antibodies and was highly protective. This provides an improved candidate for further clinical evaluation.
Collapse
|
4
|
Widjaja I, Rigter A, Jacobino S, van Kuppeveld FJM, Leenhouts K, Palomo C, Melero JA, Leusen JHW, Haijema BJ, Rottier PJM, de Haan CAM. Recombinant Soluble Respiratory Syncytial Virus F Protein That Lacks Heptad Repeat B, Contains a GCN4 Trimerization Motif and Is Not Cleaved Displays Prefusion-Like Characteristics. PLoS One 2015; 10:e0130829. [PMID: 26107504 PMCID: PMC4481108 DOI: 10.1371/journal.pone.0130829] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 05/25/2015] [Indexed: 11/30/2022] Open
Abstract
The respiratory syncytial virus (RSV) fusion protein F is considered an attractive vaccine candidate especially in its prefusion conformation. We studied whether recombinant soluble RSV F proteins could be stabilized in a prefusion-like conformation by mutation of heptad repeat B (HRB). The results show that soluble, trimeric, non-cleaved RSV F protein, produced by expression of the furin cleavage site-mutated F ectodomain extended with a GCN4 trimerization sequence, is efficiently recognized by pre- as well as postfusion-specific antibodies. In contrast, a similar F protein completely lacking HRB displayed high reactivity with prefusion-specific antibodies recognizing antigenic site Ø, but did not expose postfusion-specific antigenic site I, in agreement with this protein maintaining a prefusion-like conformation. These features were dependent on the presence of the GCN4 trimerization domain. Absence of cleavage also contributed to binding of prefusion-specific antibodies. Similar antibody reactivity profiles were observed when the prefusion form of F was stabilized by the introduction of cysteine pairs in HRB. To study whether the inability to form the 6HB was responsible for the prefusion-like antibody reactivity profile, alanine mutations were introduced in HRB. Although introduction of alanine residues in HRB inhibited the formation of the 6HB, the exposure of postfusion-specific antigenic site I was not prevented. In conclusion, proteins that are not able to form the 6HB, due to mutation of HRB, may still display postfusion-specific antigenic site I. Replacement of HRB by the GCN4 trimerization domain in a non-cleaved soluble F protein resulted, however, in a protein with prefusion-like characteristics, suggesting that this HRB-lacking protein may represent a potential prefusion F protein subunit vaccine candidate.
Collapse
Affiliation(s)
- Ivy Widjaja
- Virology Division, Department of Infectious Diseases & Immunology, Faculty of Veterinary Medicine, Utrecht University, 3894 CL, Utrecht, The Netherlands
- Mucosis B.V., Meditech Center, L.J. Zielstraweg 1, 9713 GX Groningen, The Netherlands
| | - Alan Rigter
- Virology Division, Department of Infectious Diseases & Immunology, Faculty of Veterinary Medicine, Utrecht University, 3894 CL, Utrecht, The Netherlands
- Mucosis B.V., Meditech Center, L.J. Zielstraweg 1, 9713 GX Groningen, The Netherlands
| | - Shamir Jacobino
- Department of Immunology, University Medical Center Utrecht, 3508 AB, Utrecht, The Netherlands
| | - Frank J. M. van Kuppeveld
- Virology Division, Department of Infectious Diseases & Immunology, Faculty of Veterinary Medicine, Utrecht University, 3894 CL, Utrecht, The Netherlands
| | - Kees Leenhouts
- Mucosis B.V., Meditech Center, L.J. Zielstraweg 1, 9713 GX Groningen, The Netherlands
| | - Concepción Palomo
- Centro Nacional de Microbiología and CIBERES, Instituto de Salud Carlos III, Majadahonda, 28220, Madrid, Spain
| | - Jose A. Melero
- Centro Nacional de Microbiología and CIBERES, Instituto de Salud Carlos III, Majadahonda, 28220, Madrid, Spain
| | - Jeanette H. W. Leusen
- Department of Immunology, University Medical Center Utrecht, 3508 AB, Utrecht, The Netherlands
| | - Bert Jan Haijema
- Mucosis B.V., Meditech Center, L.J. Zielstraweg 1, 9713 GX Groningen, The Netherlands
| | - Peter J. M. Rottier
- Virology Division, Department of Infectious Diseases & Immunology, Faculty of Veterinary Medicine, Utrecht University, 3894 CL, Utrecht, The Netherlands
| | - Cornelis A. M. de Haan
- Virology Division, Department of Infectious Diseases & Immunology, Faculty of Veterinary Medicine, Utrecht University, 3894 CL, Utrecht, The Netherlands
- * E-mail:
| |
Collapse
|
5
|
Vaughan K, Ponomarenko J, Peters B, Sette A. Analysis of Human RSV Immunity at the Molecular Level: Learning from the Past and Present. PLoS One 2015; 10:e0127108. [PMID: 26001197 PMCID: PMC4441423 DOI: 10.1371/journal.pone.0127108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 04/10/2015] [Indexed: 11/18/2022] Open
Abstract
Human RSV is one of the most prevalent viral pathogens of early childhood for which no vaccine is available. Herein we provide an analysis of RSV epitope data to examine its application to vaccine design and development. Our objective was to provide an overview of antigenic coverage, identify critical antibody and T cell determinants, and then analyze the cumulative RSV epitope data from the standpoint of functional responses using a combinational approach to characterize antigenic structure and epitope location. A review of the cumulative data revealed, not surprisingly, that the vast majority of epitopes have been defined for the two major surface antigens, F and G. Antibody and T cell determinants have been reported from multiple hosts, including those from human subjects following natural infection, however human data represent a minority of the data. A structural analysis of the major surface antigen, F, showed that the majority of epitopes defined for functional antibodies (neutralizing and/or protective) were either shown to bind pre-F or to be accessible in both pre- and post-F forms. This finding may have has implications for on-going vaccine design and development. These interpretations are in agreement with previous work and can be applied in the larger context of functional epitopes on the F protein. It is our hope that this work will provide the basis for further RSV-specific epitope discovery and investigation into the nature of antigen conformation in immunogenicity.
Collapse
Affiliation(s)
- Kerrie Vaughan
- La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
- * E-mail:
| | - Julia Ponomarenko
- San Diego Supercomputer Center, University of California San Diego, La Jolla, California, United States of America
| | - Bjoern Peters
- La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
| | - Alessandro Sette
- La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
| |
Collapse
|
6
|
Human metapneumovirus virus-like particles induce protective B and T cell responses in a mouse model. J Virol 2014; 88:6368-79. [PMID: 24672031 DOI: 10.1128/jvi.00332-14] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
UNLABELLED Human metapneumovirus (HMPV) is a leading cause of respiratory disease in infants, children, and the elderly worldwide, yet no licensed vaccines exist. Live-attenuated vaccines present safety challenges, and protein subunit vaccines induce primarily antibody responses. Virus-like particles (VLPs) are an attractive alternative vaccine approach because of reduced safety concerns compared with live vaccines. We generated HMPV VLPs by expressing viral proteins in suspension-adapted human embryonic kidney epithelial (293-F) cells and found that the viral matrix (M) and fusion (F) proteins were sufficient to form VLPs. We previously reported that the VLPs resemble virus morphology and incorporate fusion-competent F protein (R. G. Cox, S. B. Livesay, M. Johnson, M. D. Ohi, and J. V. Williams, J. Virol. 86:12148-12160, 2012), which we hypothesized would elicit F-specific antibody and T cell responses. In this study, we tested whether VLP immunization could induce protective immunity to HMPV by using a mouse model. C57BL/6 mice were injected twice intraperitoneally with VLPs alone or with adjuvant and subsequently challenged with HMPV. Mice were euthanized 5 days postinfection, and virus titers, levels of neutralizing antibodies, and numbers of CD3(+) T cells were quantified. Mice immunized with VLPs mounted an F-specific antibody response and generated CD8(+) T cells recognizing an F protein-derived epitope. VLP immunization induced a neutralizing-antibody response that was enhanced by the addition of either TiterMax Gold or α-galactosylceramide adjuvant, though adjuvant reduced cellular immune responses. Two doses of VLPs conferred complete protection from HMPV replication in the lungs of mice and were not associated with a Th2-skewed cytokine response. These results suggest that nonreplicating VLPs are a promising vaccine candidate for HMPV. IMPORTANCE Human metapneumovirus (HMPV) is a leading cause of acute respiratory infection in infants, children, and the elderly worldwide, yet no licensed vaccines exist. Live-attenuated vaccines present safety challenges, and protein subunit vaccines induce primarily antibody responses. Virus-like particles (VLPs) are an attractive alternative vaccine approach. We generated HMPV VLPs by expressing the viral matrix (M) and fusion (F) proteins in mammalian cells. We found that mice immunized with VLPs mounted an F-specific antibody response and generated CD8(+) T cells recognizing an F protein-derived epitope. VLP immunization induced a neutralizing-antibody response that was enhanced by the addition of either TiterMax Gold or α-galactosylceramide adjuvant. Two doses of VLPs conferred complete protection against HMPV replication in the lungs of mice and were not associated with a Th2-skewed cytokine response. These results suggest that nonreplicating VLPs are a promising vaccine candidate for HMPV.
Collapse
|
7
|
Koellhoffer JF, Higgins CD, Lai JR. Protein engineering strategies for the development of viral vaccines and immunotherapeutics. FEBS Lett 2013; 588:298-307. [PMID: 24157357 DOI: 10.1016/j.febslet.2013.10.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 10/12/2013] [Accepted: 10/14/2013] [Indexed: 01/12/2023]
Abstract
Vaccines that elicit a protective broadly neutralizing antibody (bNAb) response and monoclonal antibody therapies are critical for the treatment and prevention of viral infections. However, isolation of protective neutralizing antibodies has been challenging for some viruses, notably those with high antigenic diversity or those that do not elicit a bNAb response in the course of natural infection. Here, we discuss recent work that employs protein engineering strategies to design immunogens that elicit bNAbs or engineer novel bNAbs. We highlight the use of rational, computational, and combinatorial strategies and assess the potential of these approaches for the development of new vaccines and immunotherapeutics.
Collapse
Affiliation(s)
- Jayne F Koellhoffer
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, United States
| | - Chelsea D Higgins
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, United States
| | - Jonathan R Lai
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, United States.
| |
Collapse
|
8
|
Hung TJ, Chang WT, Tomiya N, Lee YC, Chang HT, Chen CJ, Kuo PH, Fan TC, Chang MDT. Basic amino acid residues of human eosinophil derived neurotoxin essential for glycosaminoglycan binding. Int J Mol Sci 2013; 14:19067-85. [PMID: 24065103 PMCID: PMC3794821 DOI: 10.3390/ijms140919067] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Revised: 09/06/2013] [Accepted: 09/11/2013] [Indexed: 12/26/2022] Open
Abstract
Human eosinophil derived neurotoxin (EDN), a granule protein secreted by activated eosinophils, is a biomarker for asthma in children. EDN belongs to the human RNase A superfamily possessing both ribonucleolytic and antiviral activities. EDN interacts with heparin oligosaccharides and heparin sulfate proteoglycans on bronchial epithelial Beas-2B cells. In this study, we demonstrate that the binding of EDN to cells requires cell surface glycosaminoglycans (GAGs), and the binding strength between EDN and GAGs depends on the sulfation levels of GAGs. Furthermore, in silico computer modeling and in vitro binding assays suggest critical roles for the following basic amino acids located within heparin binding regions (HBRs) of EDN 34QRRCKN39 (HBR1), 65NKTRKN70 (HBR2), and 113NRDQRRD119 (HBR3) and in particular Arg35, Arg36, and Arg38 within HBR1, and Arg114 and Arg117 within HBR3. Our data suggest that sulfated GAGs play a major role in EDN binding, which in turn may be related to the cellular effects of EDN.
Collapse
Affiliation(s)
- Ta-Jen Hung
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu 300, Taiwan; E-Mails: (T.-J.H.); (W.-T.C.); (Y.-C.L.); (C.-J.C.); (P.-H.K.)
| | - Wei-Tang Chang
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu 300, Taiwan; E-Mails: (T.-J.H.); (W.-T.C.); (Y.-C.L.); (C.-J.C.); (P.-H.K.)
| | - Noboru Tomiya
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA; E-Mail:
| | - Yuan-Chuan Lee
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu 300, Taiwan; E-Mails: (T.-J.H.); (W.-T.C.); (Y.-C.L.); (C.-J.C.); (P.-H.K.)
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA; E-Mail:
| | - Hao-Teng Chang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 404, Taiwan; E-Mail:
| | - Chien-Jung Chen
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu 300, Taiwan; E-Mails: (T.-J.H.); (W.-T.C.); (Y.-C.L.); (C.-J.C.); (P.-H.K.)
| | - Ping-Hsueh Kuo
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu 300, Taiwan; E-Mails: (T.-J.H.); (W.-T.C.); (Y.-C.L.); (C.-J.C.); (P.-H.K.)
| | - Tan-chi Fan
- Stem Cell and Translational Cancer Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan 333, Taiwan; E-Mail:
| | - Margaret Dah-Tsyr Chang
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu 300, Taiwan; E-Mails: (T.-J.H.); (W.-T.C.); (Y.-C.L.); (C.-J.C.); (P.-H.K.)
- Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +886-3-574-2463; Fax: +886-3-571-5934
| |
Collapse
|
9
|
Luo J, Yang J, Cheng Y, Li W, Yin TL, Xu WM, Zou YJ. Immunogenicity study of plasmid DNA encoding mouse cysteine-rich secretory protein-1 (mCRISP1) as a contraceptive vaccine. Am J Reprod Immunol 2012; 68:47-55. [PMID: 22429321 DOI: 10.1111/j.1600-0897.2012.01117.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Accepted: 02/03/2012] [Indexed: 11/28/2022] Open
Abstract
PROBLEM To examine the immunocontraceptive properties of the plasmid pcDNA-mCRISP1 and compare them to the corresponding recombinant mCRISP1 (r-mCRISP1). METHOD OF STUDY RT-PCR and indirect immunofluorescence were performed to observe the mCRISP1 protein expression in COS-7 cells. Three groups of mice received three injections of r-mCRISP1, pcDNA-mCRISP1 or pcDNA vector, respectively. ELISA and Western blot were used to examine the immune responses and immunoreactivity of antisera. Sperm-egg penetration assay was performed to examine the effect of anti-mCRISP1 antibodies in vitro fertilization of mouse oocytes. Fertility and mean litter size were analysed by natural mating. Histological analysis was carried out to look for potential immunopathologic effects of the antibodies. RESULTS COS-7 cells transfected with pcDNA-mCRISP1 present the expression of mCRISP1. Both r-mCRISP1 and pcDNA-mCRISP1 raised an immune response against r-mCRISP1 protein and native CRISP1 in mouse sperm. The titres of anti-mCRISP1 antibodies from DNA immunized mice were significantly lower than that of r-mCRISP1 immunized mice, but it lasted relatively longer. Male and female pcDNA-mCRISP1 injected animals presented a statistically significant reduction in their fertility with no signs of immunopathologic effects. CONCLUSION These studies demonstrated the feasibility of generating an immune response to mCRISP1 protein by DNA vaccine and pcDNA-mCRISP1 plasmid causing significant anti-fertility potential.
Collapse
Affiliation(s)
- Jin Luo
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China
| | | | | | | | | | | | | |
Collapse
|
10
|
Stegmann T, Kamphuis T, Meijerhof T, Goud E, de Haan A, Wilschut J. Lipopeptide-adjuvanted respiratory syncytial virus virosomes: A safe and immunogenic non-replicating vaccine formulation. Vaccine 2010; 28:5543-50. [DOI: 10.1016/j.vaccine.2010.06.041] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2010] [Revised: 05/19/2010] [Accepted: 06/10/2010] [Indexed: 11/29/2022]
|
11
|
Anderson R, Huang Y, Langley JM. Prospects for defined epitope vaccines for respiratory syncytial virus. Future Microbiol 2010; 5:585-602. [DOI: 10.2217/fmb.10.22] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The history of vaccines for respiratory syncytial virus (RSV) illustrates the complex immunity and immunopathology to this ubiquitous virus, starting from the failed formalin-inactivated vaccine trials performed in the 1960s. An attractive alternative to traditional live or killed virus vaccines is a defined vaccine composed of discrete antigenic epitopes for which immunological activities have been characterized as comprehensively as possible. Here we present cumulative data on murine and human CD4, CD8 and neutralization epitopes identified in RSV proteins along with information regarding their associated immune responses and host-dependent variability. Identification and characterization of RSV epitopes is a rapidly expanding topic of research with potential contributions to the tailored design of improved safe and effective vaccines.
Collapse
Affiliation(s)
- Robert Anderson
- Department of Microbiology & Immunology, Pediatrics and Canadian Center for Vaccinology, Dalhousie University, Halifax, Nova Scotia, B3H 1X5, Canada
| | - Yan Huang
- Department of Microbiology & Immunology and Canadian Center for Vaccinology, Dalhousie University, Halifax, Nova Scotia, B3H 1X5, Canada
| | - Joanne M Langley
- Department of Pediatrics, Community Health & Epidemiology and Canadian Center for Vaccinology, Dalhousie University, Halifax, Nova Scotia, B3H 1X5, Canada
| |
Collapse
|
12
|
Throsby M, de Kruif J. Towards a broadly active flavivirus therapeutic antibody. Future Virol 2009. [DOI: 10.2217/fvl.09.49] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Evaluation of: Sultana H, Foellmer HG, Neelakanta G et al.: Fusion loop peptide of the West Nile virus envelope protein is essential for pathogenesis and is recognized by a therapeutic cross-reactive human monoclonal antibody. J. Immunol. 183(1), 650–660 (2009). Flaviviruses, such as West Nile virus, the dengue serotypes, Japanese encephalitis virus and yellow fever virus are important human pathogens and can co-circulate in parts of the world. Humoral immunity is important in the mammalian protective response to flaviviruses and passive immunization has been shown to be effective in preventing infection and even treating disease. Monoclonal antibodies (mAbs) against flaviviruses can be divided basically into potent neutralizing mAbs with restricted flavivirus cross-reactivity and broadly cross-reactive mAbs with low neutralizing potency and protective activity. Here, Sultana and colleagues extend the characterization of the cross-reactive mAb11 by defining its epitope within the fusion loop of the West Nile virus envelope protein and link the reduced neuroinvasiveness of a mAb11 neutralization escape variant to a diminished ability to activate Toll-like receptor 3, induce inflammatory cytokine release and compromise blood–brain barrier integrity. The results highlight the importance of the fusion loop in viral pathogenesis; however, challenges remain to develop mAb11 into a viable therapeutic. Instead, mixtures of potent neutralizing mAbs with different flavivirus specificities may be a more feasible approach toward a broadly active flavivirus therapeutic antibody product.
Collapse
Affiliation(s)
- Mark Throsby
- Merus BV, Uppsalalaan 8, 3584CT Utrecht, The Netherlands
| | - John de Kruif
- Merus BV, Uppsalalaan 8, 3584CT Utrecht, The Netherlands
| |
Collapse
|
13
|
Oshansky CM, Zhang W, Moore E, Tripp RA. The host response and molecular pathogenesis associated with respiratory syncytial virus infection. Future Microbiol 2009; 4:279-97. [PMID: 19327115 DOI: 10.2217/fmb.09.1] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Since the isolation of respiratory syncytial virus (RSV) in 1956, its significance as an important human pathogen in infants, the elderly and the immunocompromised has been established. Many important mechanisms contributing to RSV infection, replication and disease pathogenesis have been uncovered; however, there is still insufficient knowledge in these and related areas, which must be addressed to facilitate the development of safe and effective vaccines and therapeutic treatments. A better understanding of the molecular pathogenesis of RSV infection, particularly the host-cell response and transcription profiles to RSV infection, is required to advance disease intervention strategies. Substantial information is accumulating regarding how RSV proteins modulate molecular signaling and regulation of cytokine and chemokine responses to infection, molecular signals regulating programmed cell death, and innate and adaptive immune responses to infection. This review discusses RSV manipulation of the host response to infection and related disease pathogenesis.
Collapse
Affiliation(s)
- Christine M Oshansky
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA.
| | | | | | | |
Collapse
|
14
|
Zhao D, Peng D, Li L, Zhang Q, Zhang C. Inhibition of G1P3 expression found in the differential display study on respiratory syncytial virus infection. Virol J 2008; 5:114. [PMID: 18838000 PMCID: PMC2572611 DOI: 10.1186/1743-422x-5-114] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2008] [Accepted: 10/06/2008] [Indexed: 12/27/2022] Open
Abstract
Background Respiratory syncytial virus (RSV) is the leading viral pathogen associated with bronchiolitis and lower respiratory tract disease in infants and young children worldwide. The respiratory epithelium is the primary initiator of pulmonary inflammation in RSV infections, which cause significant perturbations of global gene expression controlling multiple cellular processes. In this study, differential display reverse transcription polymerase chain reaction amplification was performed to examine mRNA expression in a human alveolar cell line (SPC-A1) infected with RSV. Results Of the 2,500 interpretable bands on denaturing polyacrylamide gels, 40 (1.6%) cDNA bands were differentially regulated by RSV, in which 28 (70%) appeared to be upregulated and another 12 (30%) appeared to be downregulated. Forty of the expressed sequence tags (EST) were isolated, and 20 matched homologs in GenBank. RSV infection upregulated the mRNA expression of chemokines CC and CXC and interfered with type α/β interferon-inducible gene expression by upregulation of MG11 and downregulation of G1P3. Conclusion RSV replication could induce widespread changes in gene expression including both positive and negative regulation and play a different role in the down-regulation of IFN-α and up-regulation of IFN-γ inducible gene expression, which suggests that RSV interferes with the innate antiviral response of epithelial cells by multiple mechanisms.
Collapse
Affiliation(s)
- Dongchi Zhao
- Pediatrics Department, Zhongnan Hospital of Wuhan University Medical School, Donghu Road 169, Wuhan 430071, PR China.
| | | | | | | | | |
Collapse
|
15
|
Hamilton SK, Ikizler MR, Wallen C, Wright PF, Harth E. Effective delivery of IgG-antibodies into infected cells via dendritic molecular transporter conjugate IgGMT. MOLECULAR BIOSYSTEMS 2008; 4:1209-11. [DOI: 10.1039/b816645e] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
16
|
Herfst S, de Graaf M, Schrauwen EJA, Ulbrandt ND, Barnes AS, Senthil K, Osterhaus ADME, Fouchier RAM, van den Hoogen BG. Immunization of Syrian golden hamsters with F subunit vaccine of human metapneumovirus induces protection against challenge with homologous or heterologous strains. J Gen Virol 2007; 88:2702-2709. [PMID: 17872522 DOI: 10.1099/vir.0.83084-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Human metapneumovirus (hMPV), a newly discovered paramyxovirus, is associated with acute respiratory-tract illness, primarily in young children, individuals with underlying disease and the elderly. Two genetic lineages of hMPV circulate around the world, and viruses from these two lineages demonstrate antigenic differences. The clinical impact of hMPV warrants the development of vaccines. Recombinant soluble fusion (F) proteins of prototype viruses of the two main lineages of hMPV that can be produced in high yields have been constructed. In this study, the antigenicity, immunogenicity and protective efficacy of these soluble F subunit vaccines were evaluated in Syrian golden hamsters (Mesocricetus auratus). Immunization of hamsters with the soluble F proteins, adjuvanted with Specol or iscom matrix, induced high virus-neutralization titres, with higher titres against the homologous than the heterologous virus. The neutralizing antibodies protected from subsequent infection of the lungs with both homologous and heterologous virus. Upon challenge, viral titres in the nasal turbinates of immunized animals were reduced significantly compared with those of PBS-immunized animals. In conclusion, a soluble F subunit vaccine for hMPV that induces cross-protective immunity for infection of the lower respiratory tract in Syrian golden hamsters has been generated.
Collapse
Affiliation(s)
- Sander Herfst
- Department of Virology, Erasmus MC, Rotterdam, The Netherlands
| | | | | | | | - Arnita S Barnes
- MedImmune, Inc., 1 MedImmune Way, Gaithersburg, MD 20878, USA
| | - Kannaki Senthil
- MedImmune, Inc., 1 MedImmune Way, Gaithersburg, MD 20878, USA
| | | | | | | |
Collapse
|
17
|
Viral and host factors in human respiratory syncytial virus pathogenesis. J Virol 2007; 82:2040-55. [PMID: 17928346 DOI: 10.1128/jvi.01625-07] [Citation(s) in RCA: 362] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
18
|
Williams JV, Chen Z, Cseke G, Wright DW, Keefer CJ, Tollefson SJ, Hessell A, Podsiad A, Shepherd BE, Sanna PP, Burton DR, Crowe JE, Williamson RA. A recombinant human monoclonal antibody to human metapneumovirus fusion protein that neutralizes virus in vitro and is effective therapeutically in vivo. J Virol 2007; 81:8315-24. [PMID: 17522220 PMCID: PMC1951312 DOI: 10.1128/jvi.00106-07] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human metapneumovirus (hMPV) is a recently discovered paramyxovirus that is a major cause of lower-respiratory-tract disease. hMPV is associated with more severe disease in infants and persons with underlying medical conditions. Animal studies have shown that the hMPV fusion (F) protein alone is capable of inducing protective immunity. Here, we report the use of phage display technology to generate a fully human monoclonal antibody fragment (Fab) with biological activity against hMPV. Phage antibody libraries prepared from human donor tissues were selected against recombinant hMPV F protein with multiple rounds of panning. Recombinant Fabs then were expressed in bacteria, and supernatants were screened by enzyme-linked immunosorbent assay and immunofluorescent assays. A number of Fabs that bound to hMPV F were isolated, and several of these exhibited neutralizing activity in vitro. Fab DS7 neutralized the parent strain of hMPV with a 60% plaque reduction activity of 1.1 mug/ml and bound to hMPV F with an affinity of 9.8 x10(-10) M, as measured by surface plasmon resonance. To test the in vivo activity of Fab DS7, groups of cotton rats were infected with hMPV and given Fab intranasally 3 days after infection. Nasal turbinates and lungs were harvested on day 4 postinfection and virus titers determined. Animals treated with Fab DS7 exhibited a >1,500-fold reduction in viral titer in the lungs, with a modest 4-fold reduction in the nasal tissues. There was a dose-response relationship between the dose of DS7 and virus titer. Human Fab DS7 may have prophylactic or therapeutic potential against severe hMPV infection.
Collapse
Affiliation(s)
- John V Williams
- Pediatric Infectious Diseases, Vanderbilt University Medical Center, D-7235 Medical Center North, 1161 21st Avenue South, Nashville, TN 37232.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Stiasny K, Kiermayr S, Holzmann H, Heinz FX. Cryptic properties of a cluster of dominant flavivirus cross-reactive antigenic sites. J Virol 2006; 80:9557-68. [PMID: 16973559 PMCID: PMC1617264 DOI: 10.1128/jvi.00080-06] [Citation(s) in RCA: 184] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
A number of flaviviruses are important human pathogens, including yellow fever, dengue, West Nile, Japanese encephalitis, and tick-borne encephalitis (TBE) viruses. Infection with or immunization against any of these viruses induces a subset of antibodies that are broadly flavivirus cross-reactive but do not exhibit significant cross-neutralization. Nevertheless, these antibodies can efficiently bind to the major envelope protein (E), which is the main target of neutralizing and protective antibodies because of its receptor-binding and membrane fusion functions. The structural basis for this phenomenon is still unclear. In our studies with TBE virus, we have provided evidence that such cross-reactive antibodies are specific for a cluster of epitopes that are partially occluded in the cage-like assembly of E proteins at the surfaces of infectious virions and involve-but are not restricted to-amino acids of the highly conserved internal fusion peptide loop. Virus disintegration leads to increased accessibility of these epitopes, allowing the cross-reactive antibodies to bind with strongly increased avidity. The cryptic properties of these sites in the context of infectious virions can thus provide an explanation for the observed lack of efficient neutralizing activity of broadly cross-reactive antibodies, despite their specificity for a functionally important structural element in the E protein.
Collapse
Affiliation(s)
- Karin Stiasny
- Institute of Virology, Medical University of Vienna, Kinderspitalgasse 15, A-1095 Vienna, Austria
| | | | | | | |
Collapse
|
20
|
Cseke G, Wright DW, Tollefson SJ, Johnson JE, Crowe JE, Williams JV. Human metapneumovirus fusion protein vaccines that are immunogenic and protective in cotton rats. J Virol 2006; 81:698-707. [PMID: 17050599 PMCID: PMC1797435 DOI: 10.1128/jvi.00844-06] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human metapneumovirus (hMPV) is a recently described paramyxovirus that is a major cause of upper and lower respiratory infection in children and adults worldwide. A safe and effective vaccine could decrease the burden of disease associated with this novel pathogen. We previously reported the development of the cotton rat model of hMPV infection and pathogenesis (J. V. Williams et al., J. Virol. 79:10944-10951, 2005). We report here the immunogenicity of an hMPV fusion (F) protein in this model. We constructed DNA plasmids that exhibited high levels of expression of hMPV F in mammalian cells (DNA-F). These constructs were used to develop a novel strategy to produce highly pure, soluble hMPV F protein lacking the transmembrane domain (FDeltaTM). We then immunized cotton rats at 0 and 14 days with either control vector, DNA-F alone, DNA-F followed by FDeltaTM protein, or FDeltaTM alone. All groups were challenged intranasally at 28 days with live hMPV. All three groups that received some form of hMPV F immunization mounted neutralizing antibody responses and exhibited partial protection against virus shedding in the lungs compared to controls. The FDeltaTM-immunized animals showed the greatest degree of protection (>1,500-fold reduction in lung virus titer). All three immunized groups showed a modest reduction of nasal virus shedding. Neither evidence of a Th2-type response nor increased lung pathology were present in the immunized animals. We conclude that sequence-optimized hMPV F protein protects against hMPV infection when delivered as either a DNA or a protein vaccine in cotton rats.
Collapse
Affiliation(s)
- Gabriella Cseke
- Department of Chemistry, Vanderbilt University Medical Center, D-7235 Medical Center North, 1161 21st Ave. South, Nashville, TN 37232-2581, USA
| | | | | | | | | | | |
Collapse
|
21
|
Margolin DH, Saunders EH, Bronfin B, de Rosa N, Axthelm MK, Goloubeva OG, Eapen S, Gelman RS, Letvin NL. Germinal center function in the spleen during simian HIV infection in rhesus monkeys. THE JOURNAL OF IMMUNOLOGY 2006; 177:1108-19. [PMID: 16818768 DOI: 10.4049/jimmunol.177.2.1108] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Infection with HIV-1, SIV, or simian HIV is associated with abnormalities in the number, size, and structure of germinal centers (GCs). To determine whether these histopathologic abnormalities are associated with abnormalities in Ab development, we analyzed nucleotide sequences of Igs from splenic GCs of simian HIV-infected macaques. Virus-specific GCs were identified in frozen splenic tissue sections by inverse immunohistochemistry using rHIV-1 gp120 as a probe. B cells from envelope-specific GCs were isolated from these sections using laser capture microdissection. Their Igs were amplified from cDNA using nested PCR, then cloned and sequenced. Nucleotide sequences were recovered from nine multimember clonal lineages. Within each lineage, sequences had similar V-D-J or V-J junctions but differed by somatic mutations distributed throughout the variable domain. The clones were highly mutated, similar to that previously reported for HIV-1-specific human IgG Abs. The average clone had 37 mutations in the V region, for a frequency of 0.11 mutations/base. The mutational pattern was strikingly nonrandom, with somatic mutations occurring preferentially at RGYW/WRCY hotspots. Transition mutations were favored over transversions, with C-->T and G-->A replacements together accounting for almost one-third of all mutations. Analysis of replacement and silent mutations in the framework and CDRs suggests that the Igs were subjected to affinity selection. These data demonstrate that the process of Ab maturation is not seriously disrupted in GCs during the early stages of immunodeficiency virus infection, and that Env-specific Igs developing in GCs are subject to extensive somatic mutation and profound selection pressures.
Collapse
Affiliation(s)
- David H Margolin
- Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Hangartner L, Zellweger RM, Giobbi M, Weber J, Eschli B, McCoy KD, Harris N, Recher M, Zinkernagel RM, Hengartner H. Nonneutralizing antibodies binding to the surface glycoprotein of lymphocytic choriomeningitis virus reduce early virus spread. ACTA ACUST UNITED AC 2006; 203:2033-42. [PMID: 16880253 PMCID: PMC2118372 DOI: 10.1084/jem.20051557] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
The biological relevance of nonneutralizing antibodies elicited early after infection with noncytopathic persistence-prone viruses is unclear. We demonstrate that cytotoxic T lymphocyte-deficient TgH(KL25) mice, which are transgenic for the heavy chain of the lymphocytic choriomeningitis virus (LCMV)-neutralizing monoclonal antibody KL25, mount a focused neutralizing antibody response following LCMV infection, and that this results in the emergence of neutralization escape virus variants. Further investigation revealed that some of the escape variants that arose early after infection could still bind to the selecting antibody. In contrast, no antibody binding could be detected for late isolates, indicating that binding, but nonneutralizing, antibodies exerted a selective pressure on the virus. Infection of naive TgH(KL25) mice with distinct escape viruses differing in their antibody-binding properties revealed that nonneutralizing antibodies accelerated clearance of antibody-binding virus variants in a partly complement-dependent manner. Virus variants that did not bind antibodies were not affected. We therefore conclude that nonneutralizing antibodies binding to the same antigenic site as neutralizing antibodies are biologically relevant by limiting early viral spread.
Collapse
Affiliation(s)
- Lars Hangartner
- Institute of Experimental Immunology, University Hospital Zürich, 8091 Zürich, Switzerland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Hangartner L, Zinkernagel RM, Hengartner H. Antiviral antibody responses: the two extremes of a wide spectrum. Nat Rev Immunol 2006; 6:231-43. [PMID: 16498452 DOI: 10.1038/nri1783] [Citation(s) in RCA: 254] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Viruses elicit a diverse spectrum of antiviral antibody responses. In this review, we discuss two widely used experimental model systems for viral infections - non-cytopathic lymphocytic choriomeningitis virus (LCMV) and acutely cytopathic vesicular stomatitis virus (VSV) - to analyse two fundamentally different types of antiviral antibody response. The basic principles found in these model infections are discussed in the context of other viral infections, and with regard to protective neutralizing versus non-protective enzyme-linked immunosorbent assay (ELISA)-detected antibody responses. Issues of antibody specificity, affinity and avidity, maturation and escape are discussed in the context of co-evolution of the host and viruses.
Collapse
Affiliation(s)
- Lars Hangartner
- Institute of Experimental Immunology, University Hospital Zurich, Schmelzbergstrasse 12, 8091 Zürich, Switzerland
| | | | | |
Collapse
|
24
|
McGill A, Marsh R, Craft AW, Toms GL. Analysis of the binding of monoclonal and polyclonal antibodies to the glycoproteins of antigenic variants of human respiratory syncytial virus by surface plasmon resonance. J Immunol Methods 2005; 297:143-52. [PMID: 15777938 DOI: 10.1016/j.jim.2004.12.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2004] [Revised: 11/18/2004] [Accepted: 12/07/2004] [Indexed: 11/20/2022]
Abstract
The surface glycoproteins of human respiratory syncytial virus (hRSV), F and G, are the major protective antigens of the virus. Both are antigenically variable, although to different degrees, but the role of antigenic variation in the pathogenesis of hRSV disease has not been fully evaluated. Assessment of immunity to different virus strains is difficult with conventional antibody assays where differing properties of the virus antigens, other than antigenicity, may influence the outcome of the assay. Here, we have developed BIAcore surface plasmon resonance based assays for antibodies to the glycoproteins of hRSV which allow valid comparison of antibody titres against multiple hRSV strains. Glycoproteins from a number of lineages of hRSV sub-group A were captured from lysates of infected cells onto the dextran coated surface of a BIAcore sensor chip via primary monoclonal antibodies (MAbs) to conserved epitopes. For the G glycoprotein, primary MAbs were conjugated directly to the dextran of the sensor chip via free amide groups. For the F glycoprotein, direct conjugation was found to inactivate the MAb and primary MAb was immobilised on the chip via rabbit anti-mouse Fc antibody fragments in an indirect system. Using monoclonal antibodies as secondary MAbs, the glycoproteins in both systems were shown to exhibit a sub-set of conserved and variable epitopes, with some epitopes of both sorts being unavailable, presumably blocked by the primary antibody. Polyclonal anti-hRSV sera raised against viruses of different genotype bound equally to both F and G glycoproteins from homologous and heterologous viruses suggesting that mice immunised systemically with lysates of cells infected with recent isolates of virus do not respond well to genotype specific epitopes.
Collapse
Affiliation(s)
- A McGill
- The School of Clinical Medical Sciences, The University of Newcastle upon Tyne, The Medical School, Framlington Place, Newcastle upon Tyne, NE2 4HH, United Kingdom
| | | | | | | |
Collapse
|
25
|
|
26
|
Easton AJ, Domachowske JB, Rosenberg HF. Animal pneumoviruses: molecular genetics and pathogenesis. Clin Microbiol Rev 2004; 17:390-412. [PMID: 15084507 PMCID: PMC387412 DOI: 10.1128/cmr.17.2.390-412.2004] [Citation(s) in RCA: 126] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Pneumoviruses are single-stranded, negative-sense, nonsegmented RNA viruses of the family Paramyxoviridae, subfamily Pneumovirinae, and include pathogens that infect humans (respiratory syncytial virus and human metapneumovirus), domestic mammals (bovine, ovine, and caprine respiratory syncytial viruses), rodents (pneumonia virus of mice), and birds (avian metapneumovirus). Among the topics considered in this review are recent studies focused on the roles of the individual virus-encoded components in promoting virus replication as well as in altering and evading innate antiviral host defenses. Advances in the molecular technology of pneumoviruses and the emergence of recombinant pneumoviruses that are leading to improved virus-based vaccine formulations are also discussed. Since pneumovirus infection in natural hosts is associated with a profound inflammatory response that persists despite adequate antiviral therapy, we also review the recent experimental treatment strategies that have focused on combined antiviral, anti-inflammatory, and immunomodulatory approaches.
Collapse
|
27
|
Tripp RA. The Brume Surrounding Respiratory Syncytial Virus Persistence. Am J Respir Crit Care Med 2004; 169:778-9. [PMID: 15044217 DOI: 10.1164/rccm.2401013] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
28
|
Sanders RW, Dankers MM, Busser E, Caffrey M, Moore JP, Berkhout B. Evolution of the HIV-1 envelope glycoproteins with a disulfide bond between gp120 and gp41. Retrovirology 2004; 1:3. [PMID: 15169554 PMCID: PMC416572 DOI: 10.1186/1742-4690-1-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2004] [Accepted: 03/09/2004] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND We previously described the construction of an HIV-1 envelope glycoprotein complex (Env) that is stabilized by an engineered intermolecular disulfide bond (SOS) between gp120 and gp41. The modified Env protein antigenically mimics the functional wild-type Env complex. Here, we explore the effects of the covalent gp120 - gp41 interaction on virus replication and evolution. RESULTS An HIV-1 molecular clone containing the SOS Env gene was only minimally replication competent, suggesting that the engineered disulfide bond substantially impaired Env function. However, virus evolution occurred in cell culture infections, and it eventually always led to elimination of the intermolecular disulfide bond. In the course of these evolution studies, we identified additional and unusual second-site reversions within gp41. CONCLUSIONS These evolution paths highlight residues that play an important role in the interaction between gp120 and gp41. Furthermore, our results suggest that a covalent gp120 - gp41 interaction is incompatible with HIV-1 Env function, probably because this impedes conformational changes that are necessary for fusion to occur, which may involve the complete dissociation of gp120 from gp41.
Collapse
Affiliation(s)
- Rogier W Sanders
- Dept. of Human Retrovirology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Dept. of Microbiology and Immunology, Weill Medical College of Cornell University, 1300 York Ave., New York, NY 1002, USA
| | - Martijn M Dankers
- Dept. of Human Retrovirology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Els Busser
- Dept. of Human Retrovirology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Michael Caffrey
- Dept. of Biochemistry and Molecular Biology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - John P Moore
- Dept. of Microbiology and Immunology, Weill Medical College of Cornell University, 1300 York Ave., New York, NY 1002, USA
| | - Ben Berkhout
- Dept. of Human Retrovirology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
29
|
Weitkamp JH, Kallewaard N, Kusuhara K, Feigelstock D, Feng N, Greenberg HB, Crowe JE. Generation of recombinant human monoclonal antibodies to rotavirus from single antigen-specific B cells selected with fluorescent virus-like particles. J Immunol Methods 2003; 275:223-37. [PMID: 12667686 DOI: 10.1016/s0022-1759(03)00013-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Technical difficulties have severely limited the yield of methods for the generation of human antiviral monoclonal antibodies (Mabs) in the past. We describe here a novel method for the efficient development of human Mabs against viruses. Rotavirus (RV) is a major cause of gastroenteritis in infants and adults worldwide. We generated fluorescent virus-like particles (VLPs) to identify and physically sort single RV-specific B cells from healthy adult blood donors, or RV-infected infants or adults. We expanded the sorted single B cells in culture, tested for RV-specific antibody secretion, and cloned and sequenced the antibody heavy and light chain variable region (VH and VL) genes. The percentage of wells that produced antibodies after sorting and expanding RV-specific adult B cell clones was high at 23%. The overall efficiency of RV-specific antibody gene recovery after the isolation, confirmation, and cloning of RV-specific VH segments was 1.3% of sorted cells in adults. RV-specific variable gene segments also were obtained from acutely infected infants, although infant B cells did not proliferate and differentiate in culture as well as adult B cells. We expressed recombinant Fabs incorporating the VH and VL genes from RV-specific B cell clones using a new modified bacterial Fab expression vector that we describe. Finally, we demonstrated binding of purified Fabs to RV proteins by immunofluorescence and ELISA. This method for the generation of recombinant human Mabs to RV from single antigen-specific B cell clones selected with fluorescent VLPs could be used to generate human Mabs to many other viruses whose proteins can self-assemble into VLPs.
Collapse
Affiliation(s)
- Jörn Hendrik Weitkamp
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232-2581, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
Neutralizing antibodies are crucial for vaccine-mediated protection against viral diseases. They probably act, in most cases, by blunting the infection, which is then resolved by cellular immunity. The protective effects of neutralizing antibodies can be achieved not only by neutralization of free virus particles, but also by several activities directed against infected cells. In certain instances, non-neutralizing antibodies contribute to protection. Several viruses, such as HIV, have evolved mechanisms to evade neutralizing-antibody responses, and these viruses present special challenges for vaccine design that are now being tackled.
Collapse
Affiliation(s)
- Dennis R Burton
- Department of Immunology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA.
| |
Collapse
|
31
|
Tsui P, Tornetta MA, Ames RS, Silverman C, Porter T, Weston C, Griego S, Sweet RW. Progressive epitope-blocked panning of a phage library for isolation of human RSV antibodies. J Immunol Methods 2002; 263:123-32. [PMID: 12009209 DOI: 10.1016/s0022-1759(02)00032-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Epitope-blocked panning is an approach to mining antigen-specific diversity from phage display antibody libraries. Previously, we developed and used this method to recover a neutralizing antibody to respiratory syncytial virus (RSV) by blocking a dominant response to a nonneutralizing epitope on a recombinant derivative of the viral F antigen. We have extended this approach to the blocking of multiple epitopes simultaneously, which led to the recovery of new antibodies of different specificity, including one new neutralizing activity. A phage display Fab library was selected on recombinant F antigen in the presence of three representative antibodies recovered in the unblocked and subsequent single-blocked panning procedures. Restriction endonuclease fingerprinting of 13 F+ clones revealed seven unique Fabs. DNA sequence analysis of five of these clones revealed five new light chains in combination with different heavy chains, three of which were very similar or identical to Fabs previously isolated from this library. The blocking antibodies did not compete with the new Fabs, demonstrating effective masking of their binding sites in the panning procedure. Conversely, these Fabs did show variable inhibition of two of the blocking antibodies suggesting a close proximity or interdependence of their epitopes. One of the antibodies did inhibit virus infection, albeit with modest potency. These results demonstrate that epitope-blocked panning is a self-progressing approach to retrieving diverse antibodies from phage libraries.
Collapse
Affiliation(s)
- Ping Tsui
- Department of Molecular Biology, GlaxoSmithKline Pharmaceuticals, 709 Swedeland Road, P.O. Box 1539, King of Prussia, PA 19406, USA.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Binley JM, Sanders RW, Master A, Cayanan CS, Wiley CL, Schiffner L, Travis B, Kuhmann S, Burton DR, Hu SL, Olson WC, Moore JP. Enhancing the proteolytic maturation of human immunodeficiency virus type 1 envelope glycoproteins. J Virol 2002; 76:2606-16. [PMID: 11861826 PMCID: PMC135977 DOI: 10.1128/jvi.76.6.2606-2616.2002] [Citation(s) in RCA: 125] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2001] [Accepted: 12/06/2001] [Indexed: 12/31/2022] Open
Abstract
In virus-infected cells, the envelope glycoprotein (Env) precursor, gp160, of human immunodeficiency virus type 1 is cleaved by cellular proteases into a fusion-competent gp120-gp41 heterodimer in which the two subunits are noncovalently associated. However, cleavage can be inefficient when recombinant Env is expressed at high levels, either as a full-length gp160 or as a soluble gp140 truncated immediately N-terminal to the transmembrane domain. We have explored several methods for obtaining fully cleaved Env for use as a vaccine antigen. We tested whether purified Env could be enzymatically digested with purified protease in vitro. Plasmin efficiently cleaved the Env precursor but also cut at a second site in gp120, most probably the V3 loop. In contrast, a soluble form of furin was specific for the gp120-gp41 cleavage site but cleaved inefficiently. Coexpression of Env with the full-length or soluble form of furin enhanced Env cleavage but also reduced Env expression. When the Env cleavage site (REKR) was mutated in order to see if its use by cellular proteases could be enhanced, several mutants were found to be processed more efficiently than the wild-type protein. The optimal cleavage site sequences were RRRRRR, RRRRKR, and RRRKKR. These mutations did not significantly alter the capacity of the Env protein to mediate fusion, so they have not radically perturbed Env structure. Furthermore, unlike that of wild-type Env, expression of the cleavage site mutants was not significantly reduced by furin coexpression. Coexpression of Env cleavage site mutants and furin is therefore a useful method for obtaining high-level expression of processed Env.
Collapse
Affiliation(s)
- James M Binley
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, New York 10021, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Crowe JE, Suara RO, Brock S, Kallewaard N, House F, Weitkamp JH. Genetic and structural determinants of virus neutralizing antibodies. Immunol Res 2002; 23:135-45. [PMID: 11444379 DOI: 10.1385/ir:23:2-3:135] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Neutralizing antibodies (Abs) are the principal protective mechanism against disease caused by reinfection with viruses. Ab-mediated neutralization of viruses is a complex process comprising multiple mechanisms. Every structural aspect of Abs is potentially capable of modulating the level of neutralizing activity or the mechanisms of neutralization. The focus of our laboratory is to understand the genetic and structural basis of Ab-mediated neutralization of human viral pathogens. We demonstrated the unexpected finding that virus antigen-binding fragments of Abs (Fabs) mediate potent virus neutralizing effects in vivo. This work has led to a broad investigation of the importance of the genetics, chemistry, and structure of the combining site to the neutralizing activity of antiviral Abs. Ongoing work in our laboratory reveals that effect or functions specified by the Ab isotype such as polymer formation, interactions with complement, interactions with Fc receptors, and the ability to transcytose mucosal epithelia, also modulate the mechanism and level of neutralizing effects mediated by antiviral Abs.
Collapse
Affiliation(s)
- J E Crowe
- Department of Pediatrics and Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232-2581, USA.
| | | | | | | | | | | |
Collapse
|
34
|
Burton DR, Saphire EO, Parren PW. A model for neutralization of viruses based on antibody coating of the virion surface. Curr Top Microbiol Immunol 2001; 260:109-43. [PMID: 11443871 DOI: 10.1007/978-3-662-05783-4_7] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- D R Burton
- Departments of Immunology and Molecular Biology, Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
35
|
Sugrue RJ, Brown C, Brown G, Aitken J, McL Rixon HW. Furin cleavage of the respiratory syncytial virus fusion protein is not a requirement for its transport to the surface of virus-infected cells. J Gen Virol 2001; 82:1375-1386. [PMID: 11369882 DOI: 10.1099/0022-1317-82-6-1375] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The intracellular cleavage of respiratory syncytial virus (RSV) fusion (F) protein by furin was examined. In RSV-infected LoVo cells, which express an inactive form of furin, and in RSV-infected Vero cells treated with the furin inhibitor decanoyl-Arg-Val-Lys-Arg-chloromethyl ketone (dec-RVKR-cmk), the F protein was expressed as a non-cleaved 73 kDa species. In both cases the F protein was initially expressed as an endoglycosidase H (Endo H)-sensitive precursor (F0(EHs)) which was modified approximately 40 min post-synthesis by the addition of complex carbohydrates to produce the Endo H-resistant form (F0(EHr)). The size and glycosylation state of F0(EHr) were identical to a transient intermediate form of non-cleaved F protein which was detected in RSV-infected Vero cells in the absence of inhibitor. Cell surface biotinylation and surface immunofluorescence staining showed that F0(EHr) was present on the surface of RSV-infected cells. RSV filaments have been shown to be the predominant form of the budding virus that is detected during virus replication. Analysis of the RSV-infected cells using scanning electron microscopy (SEM) showed that, in the presence of dec-RVKR-cmk, virus budding was impaired, producing fewer and much smaller viral filaments than in untreated cells. A comparison of immunofluorescence and SEM data showed that F0(EHr) was routed to the surface of virus-infected cells but not located in these smaller structures. Our findings suggest that activation of the F protein is required for the efficient formation of RSV filaments.
Collapse
Affiliation(s)
- Richard J Sugrue
- MRC Virology Unit, Institute of Virology, Church Street, G11 5JR, Glasgow, UK1
| | - Craig Brown
- MRC Virology Unit, Institute of Virology, Church Street, G11 5JR, Glasgow, UK1
| | - Gaie Brown
- MRC Virology Unit, Institute of Virology, Church Street, G11 5JR, Glasgow, UK1
| | - James Aitken
- Division of Virology, University of Glasgow, Institute of Virology, Church Street, G11 5JR, Glasgow, UK2
| | - Helen W McL Rixon
- MRC Virology Unit, Institute of Virology, Church Street, G11 5JR, Glasgow, UK1
| |
Collapse
|
36
|
Schmidt AC, Couch RB, Galasso GJ, Hayden FG, Mills J, Murphy BR, Chanock RM. Current research on respiratory viral infections: Third International Symposium. Antiviral Res 2001; 50:157-96. [PMID: 11397506 PMCID: PMC7133842 DOI: 10.1016/s0166-3542(01)00136-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2001] [Accepted: 02/28/2001] [Indexed: 12/27/2022]
Affiliation(s)
- A C Schmidt
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 7 Center Drive, Bethesda, MD 20892-0720, USA.
| | | | | | | | | | | | | |
Collapse
|
37
|
Dollenmaier G, Mosier SM, Scholle F, Sharma N, McKnight KL, Lemon SM. Membrane-associated respiratory syncytial virus F protein expressed from a human rhinovirus type 14 vector is immunogenic. Virology 2001; 281:216-30. [PMID: 11277694 DOI: 10.1006/viro.2000.0796] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Human rhinovirus (HRV) replicons have the potential to serve as respiratory vaccine vectors for mucosal immunization in humans. However, since many vaccine immunogens of interest are glycosylated, an important concern is whether HRV replicons are capable of expressing glycosylated proteins. The human respiratory syncytial virus (RSV) fusion (F) protein was chosen as a model glycoprotein and the HRV replicon DeltaP1FVP3 was generated by inserting the F protein-coding sequence in frame and in lieu of the 5' proximal 1489 nucleotides of the capsid-coding segment in the HRV-14 genome. When transfected into H1-HeLa cells, DeltaP1FVP3 replicated and led to the expression of the F protein. Inhibition with guanidine demonstrated that F-protein expression was dependent on DeltaP1FVP3 replication and did not result from translation of input RNA. Although most of the F protein remained as an immature, glycosylated precursor (F0), a readily detectable fraction of the protein was processed into the mature glycosylated subunit F1, an event known to occur within the Golgi apparatus. Packaged DeltaP1FVP3 replicons were generated in transfected HeLa cells by coexpression of homologous HRV capsid proteins using the vaccinia virus/T7 RNA polymerase hybrid system. Packaged replicon RNAs were capable of infecting fresh cells, leading to accumulation of the F protein as in RNA-transfected cells. Mice immunized with HeLa cell lysates containing F protein expressed from DeltaP1FVP3 produced neutralizing antibodies against RSV. These results indicate that an HRV-14 replicon can express a foreign glycosylated protein, providing further support for the potential of HRV replicons as a vaccine delivery system.
Collapse
Affiliation(s)
- G Dollenmaier
- Department of Microbiology and Immunology, The University of Texas Medical Branch at Galveston, 301 University Boulevard, 4.112 MRB, Galveston, Texas 77555-1019, USA
| | | | | | | | | | | |
Collapse
|
38
|
Abstract
This chapter discusses in vitro and in vivo antiviral activities of antibody. Since experimentation is far easier in vitro , researchers have been sought to develop in vitro assays that are expected to predict activity in vivo . This could be important in both vaccine design and in passive antibody administration. The proposed mechanisms of in vitro neutralization range from those requiring binding of a single antibody molecule to virus to those requiring substantially complete antibody coating of virus. In vitro, antiviral activity can be separated into activity against virions and activity against infected cells. The activity against virions most often considered is neutralization that can be defined as the loss of infectivity, which ensues when antibody molecule(s) bind to a virus particle, and occurs without the involvement of any other agency. In vivo, it is conventional to distinguish phenomenologically between two types of antibody antiviral activity. One of them is the ability of antibody to protect against infection when it is present before or immediately following infection. Evidence for a number of viruses in vitro indicates that lower antibody concentrations are required to inhibit infection propagated by free virus than are required to inhibit infection propagated by cell-to-cell spread.
Collapse
Affiliation(s)
- P W Parren
- Departments of Immunology and Molecular Biology, Scripps Research Institute, La Jolla, California 92037, USA
| | | |
Collapse
|
39
|
Abstract
The application of surface plasmon resonance biosensors in life sciences and pharmaceutical research continues to increase. This review provides a comprehensive list of the commercial 1999 SPR biosensor literature and highlights emerging applications that are of general interest to users of the technology. Given the variability in the quality of published biosensor data, we present some general guidelines to help increase confidence in the results reported from biosensor analyses.
Collapse
Affiliation(s)
- R L Rich
- Center for Biomolecular Interaction Analysis, University of Utah School of Medicine, Salt Lake City 84132, USA
| | | |
Collapse
|
40
|
Parren PW, Poignard P, Ditzel HJ, Williamson RA, Burton DR. Antibodies in human infectious disease. Immunol Res 2000; 21:265-78. [PMID: 10852127 DOI: 10.1385/ir:21:2-3:265] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Investigation of human antibody responses to viral pathogens at the molecular level is revealing novel aspects of the interplay of viruses with the humoral immune system. In viral infection, at least two types of human antibody responses exist: a response to mature envelope on virions that is neutralizing and a response to immature forms of envelope (viral debris) that is not. Many pathogens have, to varying degrees, evolved envelopes to minimize antibody responses against epitopes exposed on the virion. In this article, we review recent studies on human immunodeficiency virus type 1, Ebola virus, and respiratory syncytial virus. Prion diseases are diseases of protein conformation. We have generated a large panel of antibodies recognizing the cellular prion protein (PrP(c)), some of which also react with the abnormally folded infectious prion protein (PrP(Sc)). These antibodies are being used to gain insight into both the molecular events leading to the formation of infectious PrP and the physiologic role played by PrP in normal and prion-infected cells.
Collapse
Affiliation(s)
- P W Parren
- Department of Immunology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | | | | |
Collapse
|
41
|
Nguyen H, Hay J, Mazzulli T, Gallinger S, Sandhu J, Teng Y, Hozumi N. Efficient generation of respiratory syncytial virus (RSV)-neutralizing human MoAbs via human peripheral blood lymphocyte (hu-PBL)-SCID mice and scFv phage display libraries. Clin Exp Immunol 2000; 122:85-93. [PMID: 11012623 PMCID: PMC1905756 DOI: 10.1046/j.1365-2249.2000.01345.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
RSV is one of the major causes of pneumonia and bronchiolitis in infants and young children and is associated with high mortality. RSV neutralizing human antibody (hu-Ab) is known to mediate resistance to viral infection as well as to be an effective treatment for severe lower respiratory tract RSV infection. We have previously demonstrated that human primary and secondary immune responses can be established in severe combined immunodeficient mice engrafted with human peripheral blood lymphocytes (hu-PBL-SCID). By combining this animal model with the single-chain Fv antibody (scFv) phage display library technique, we were able to investigate further its clinical potential by generating a panel of human scFvs that exhibit both high F glycoprotein (RSV-F) binding affinities ( approximately 108 M(-1)) and strong neutralizing activities against RSV infection in vitro. Sequencing analysis of the randomly isolated anti-RSV-F scFv clones revealed that they were derived from different VH families with mutations in the complementarity-determining region 1 (CDR1). The results suggest that: (i) RSV-F-specific human immune responses and affinity maturation can be induced in hu-PBL-SCID mice; and (ii) this approach can be applied to generate large numbers of human scFvs with therapeutic potential. Despite the fact that hu-PBL-SCID mouse and human scFv phage display library have individually been established, our approach contributes a simple and significant step toward the generalization of antigen-specific human monoclonal antibody (hu-MoAb) production and their clinical applications.
Collapse
Affiliation(s)
- H Nguyen
- Department of Laboratory Medicine and Pathobiology, Samuel Lunenfeld Research Institute, Toronto, Ontario, Canada.
| | | | | | | | | | | | | |
Collapse
|
42
|
Sanders RW, Schiffner L, Master A, Kajumo F, Guo Y, Dragic T, Moore JP, Binley JM. Variable-loop-deleted variants of the human immunodeficiency virus type 1 envelope glycoprotein can be stabilized by an intermolecular disulfide bond between the gp120 and gp41 subunits. J Virol 2000; 74:5091-100. [PMID: 10799583 PMCID: PMC110861 DOI: 10.1128/jvi.74.11.5091-5100.2000] [Citation(s) in RCA: 91] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/1999] [Accepted: 02/17/2000] [Indexed: 11/20/2022] Open
Abstract
We have described an oligomeric gp140 envelope glycoprotein from human immunodeficiency virus type 1 that is stabilized by an intermolecular disulfide bond between gp120 and the gp41 ectodomain, termed SOS gp140 (J. M. Binley, R. W. Sanders, B. Clas, N. Schuelke, A. Master, Y. Guo, F. Kajumo, D. J. Anselma, P. J. Maddon, W. C. Olson, and J. P. Moore, J. Virol. 74:627-643, 2000). In this protein, the protease cleavage site between gp120 and gp41 is fully utilized. Here we report the characterization of gp140 variants that have deletions in the first, second, and/or third variable loop (V1, V2, and V3 loops). The SOS disulfide bond formed efficiently in gp140s containing a single loop deletion or a combination deletion of the V1 and V2 loops. However, deletion of all three variable loops prevented formation of the SOS disulfide bond. Some variable-loop-deleted gp140s were not fully processed to their gp120 and gp41 constituents even when the furin protease was cotransfected. The exposure of the gp120-gp41 cleavage site is probably affected in these proteins, even though the disabling change is in a region of gp120 distal from the cleavage site. Antigenic characterization of the variable-loop-deleted SOS gp140 proteins revealed that deletion of the variable loops uncovers cryptic, conserved neutralization epitopes near the coreceptor-binding site on gp120. These modified, disulfide-stabilized glycoproteins might be useful as immunogens.
Collapse
Affiliation(s)
- R W Sanders
- Department of Human Retrovirology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Affiliation(s)
- D R Burton
- Department of Immunology, Scripps Research Institute, La Jolla, California 92037, USA
| | | | | |
Collapse
|