1
|
Abstract
Neutralizing antibodies (nAbs) are being increasingly used as passive antiviral reagents in prophylactic and therapeutic modalities and to guide viral vaccine design. In vivo, nAbs can mediate antiviral functions through several mechanisms, including neutralization, which is defined by in vitro assays in which nAbs block viral entry to target cells, and antibody effector functions, which are defined by in vitro assays that evaluate nAbs against viruses and infected cells in the presence of effector systems. Interpreting in vivo results in terms of these in vitro assays is challenging but important in choosing optimal passive antibody and vaccine strategies. Here, I review findings from many different viruses and conclude that, although some generalizations are possible, deciphering the relative contributions of different antiviral mechanisms to the in vivo efficacy of antibodies currently requires consideration of individual antibody-virus interactions.
Collapse
Affiliation(s)
- Dennis R Burton
- Department of Immunology and Microbiology, Consortium for HIV/AIDS Vaccine Development, International AIDS Vaccine Initiative Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA.
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA.
| |
Collapse
|
2
|
Samsudin F, Yeo JY, Gan SKE, Bond PJ. Not all therapeutic antibody isotypes are equal: the case of IgM versus IgG in Pertuzumab and Trastuzumab. Chem Sci 2020; 11:2843-2854. [PMID: 32206268 PMCID: PMC7069520 DOI: 10.1039/c9sc04722k] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 02/12/2020] [Indexed: 01/06/2023] Open
Abstract
The therapeutic potential of immunoglobulin M (IgM) is of considerable interest in immunotherapy due to its complement-activating and cell-agglutinating abilities. Pertuzumab and Trastuzumab are monoclonal antibodies used to treat human epidermal growth factor receptor 2 (HER2)-positive breast cancer but exhibit significantly different binding affinities as IgM when compared to its IgG isotype. Using integrative multiscale modelling and simulations of complete antibody assemblies, we show that Pertuzumab IgM is able to utilize all of its V-regions to bind multiple HER2 receptors simultaneously, while similar binding in Trastuzumab IgM is prohibited by steric clashes caused by the large globular domain of HER2. This is subsequently validated by confirming that Pertuzumab IgM inhibits proliferation in HER2 over-expressing live cells more effectively than its IgG counterpart and Trastuzumab IgM. Our study highlights the importance of understanding the molecular details of antibody-antigen interactions for the design and isotype selection of therapeutic antibodies.
Collapse
Affiliation(s)
- Firdaus Samsudin
- Bioinformatics Institute (ASTAR) , 30 Biopolis Street, #07-01 Matrix , Singapore 138671 , Singapore . ;
| | - Joshua Yi Yeo
- Bioinformatics Institute (ASTAR) , 30 Biopolis Street, #07-01 Matrix , Singapore 138671 , Singapore . ;
| | - Samuel Ken-En Gan
- Bioinformatics Institute (ASTAR) , 30 Biopolis Street, #07-01 Matrix , Singapore 138671 , Singapore . ;
- p53 Laboratory (ASTAR) , 8A Biomedical Grove, #06-04/05 Neuros/Immunos , Singapore 138648
- Experimental Drug Development Center (ASTAR) , 10 Biopolis Road Chromos #05-01 , Singapore 138670
| | - Peter J Bond
- Bioinformatics Institute (ASTAR) , 30 Biopolis Street, #07-01 Matrix , Singapore 138671 , Singapore . ;
- Department of Biological Sciences , National University of Singapore , 14 Science Drive 4 , Singapore 117543 , Singapore
| |
Collapse
|
3
|
Glycoprotein-Specific Antibodies Produced by DNA Vaccination Protect Guinea Pigs from Lethal Argentine and Venezuelan Hemorrhagic Fever. J Virol 2016; 90:3515-29. [PMID: 26792737 DOI: 10.1128/jvi.02969-15] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 12/23/2015] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Several members of the Arenaviridae can cause acute febrile diseases in humans, often resulting in lethality. The use of convalescent-phase human plasma is an effective treatment in humans infected with arenaviruses, particularly species found in South America. Despite this, little work has focused on developing potent and defined immunotherapeutics against arenaviruses. In the present study, we produced arenavirus neutralizing antibodies by DNA vaccination of rabbits with plasmids encoding the full-length glycoprotein precursors of Junín virus (JUNV), Machupo virus (MACV), and Guanarito virus (GTOV). Geometric mean neutralizing antibody titers, as measured by the 50% plaque reduction neutralization test (PRNT(50)), exceeded 5,000 against homologous viruses. Antisera against each targeted virus exhibited limited cross-species binding and, to a lesser extent, cross-neutralization. Anti-JUNV glycoprotein rabbit antiserum protected Hartley guinea pigs from lethal intraperitoneal infection with JUNV strain Romero when the antiserum was administered 2 days after challenge and provided some protection (∼30%) when administered 4 days after challenge. Treatment starting on day 6 did not protect animals. We further formulated an IgG antibody cocktail by combining anti-JUNV, -MACV, and -GTOV antibodies produced in DNA-vaccinated rabbits. This cocktail protected 100% of guinea pigs against JUNV and GTOV lethal disease. We then expanded on this cocktail approach by simultaneously vaccinating rabbits with a combination of plasmids encoding glycoproteins from JUNV, MACV, GTOV, and Sabia virus (SABV). Sera collected from rabbits vaccinated with the combination vaccine neutralized all four targets. These findings support the concept of using a DNA vaccine approach to generate a potent pan-arenavirus immunotherapeutic. IMPORTANCE Arenaviruses are an important family of emerging viruses. In infected humans, convalescent-phase plasma containing neutralizing antibodies can mitigate the severity of disease caused by arenaviruses, particularly species found in South America. Because of variations in potency of the human-derived product, limited availability, and safety concerns, this treatment option has essentially been abandoned. Accordingly, despite this approach being an effective postinfection treatment option, research on novel approaches to produce potent polyclonal antibody-based therapies have been deficient. Here we show that DNA-based vaccine technology can be used to make potently neutralizing antibodies in rabbits that exclusively target the glycoproteins of several human-pathogenic arenaviruses found in South America, including JUNV, MACV, GTOV, and SABV. These antibodies protected guinea pigs from lethal disease when given post-virus challenge. We also generated a purified antibody cocktail with antibodies targeting three arenaviruses and demonstrated protective efficacy against all three targets. Our findings demonstrate that use of the DNA vaccine technology could be used to produce candidate antiarenavirus neutralizing antibody-based products.
Collapse
|
4
|
Botten JW, Kotturi MF. Adaptive immunity to Lymphocytic choriomeningitis virus: new insights into antigenic determinants. Future Virol 2007. [DOI: 10.2217/17460794.2.5.495] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Lymphocytic choriomeningitis virus (LCMV) is one of the most studied infectious disease models in mice. Human infection with LCMV can result in severe disease, ranging from aseptic meningitis in immunocompetent individuals, hydrocephalus, chorioretinitis or microcephaly in fetal infection, or to a highly lethal outcome in immunosuppressed individuals. This review examines recent advances in our understanding of the adaptive immune response to LCMV and how the cell-mediated and humoral immune responses contribute to protective immunity. New insights into the antigenicity of the LCMV proteome and the complexity of the cell-mediated immune response are addressed. We also discuss state-of-the-art approaches for T-cell epitope discovery in murine and human backgrounds and their recent application to LCMV. New findings regarding CD4+ T-cell dysregulation during chronic LCMV infection, and potential avenues for the treatment of chronic viral infection through modulation of the programmed cell death-1 receptor and/or IL-10 signaling pathways, are also evaluated.
Collapse
Affiliation(s)
- Jason W Botten
- The Scripps Research Institute, Molecular & Integrative Neurosciences Department, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Maya F Kotturi
- La Jolla Institute for Allergy & Immunology, Division of Vaccine Discovery, 9420 Athena Circle, La Jolla, CA 92037, USA
| |
Collapse
|
5
|
Botten J, Whitton JL, Barrowman P, Sidney J, Whitmire JK, Alexander J, Ting JPC, Bui HH, Sette A, Buchmeier MJ. HLA-A2-restricted protection against lethal lymphocytic choriomeningitis. J Virol 2007; 81:2307-17. [PMID: 17166907 PMCID: PMC1865925 DOI: 10.1128/jvi.02063-06] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2006] [Accepted: 12/01/2006] [Indexed: 12/12/2022] Open
Abstract
The consequences of human lymphocytic choriomeningitis virus (LCMV) infection can be severe, including aseptic meningitis in immunocompetent individuals, hydrocephalus or chorioretinitis in fetal infection, or a highly lethal outcome in immunosuppressed individuals. In murine models of LCMV infection, CD8(+) T cells play a primary role in providing protective immunity, and there is evidence that cellular immunity may also be important in related arenavirus infections in humans. For this reason, we sought to identify HLA-A2 supertype-restricted epitopes from the LCMV proteome and evaluate them as vaccine determinants in HLA transgenic mice. We identified four HLA-A*0201-restricted peptides-nucleoprotein NP(69-77), glycoprotein precursor GPC(10-18), GPC(447-455), and zinc-binding protein Z(49-58)-that displayed high-affinity binding (< or =275 nM) to HLA-A*0201, induced CD8(+) T-cell responses of high functional avidity in HLA-A*0201 transgenic mice, and were naturally processed from native LCMV antigens in HLA-restricted human antigen presenting cells. One of the epitopes (GPC(447-455)), after peptide immunization of HLA-A*0201 mice, induced CD8(+) T cells capable of killing peptide-pulsed HLA-A*0201-restricted target cells in vivo and protected mice against lethal intracranial challenge with LCMV.
Collapse
Affiliation(s)
- Jason Botten
- Molecular and Integrative Neurosciences Department, The Scripps Research Institute, SP30-2020, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
McCausland MM, Yusuf I, Tran H, Ono N, Yanagi Y, Crotty S. SAP regulation of follicular helper CD4 T cell development and humoral immunity is independent of SLAM and Fyn kinase. THE JOURNAL OF IMMUNOLOGY 2007; 178:817-28. [PMID: 17202343 DOI: 10.4049/jimmunol.178.2.817] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Mutations in SH2D1A resulting in lack of SLAM-associated protein (SAP) expression cause the human genetic immunodeficiency X-linked lymphoproliferative disease. A severe block in germinal center development and lack of long-term humoral immunity is one of the most prominent phenotypes of SAP(-) mice. We show, in this study, that the germinal center block is due to an essential requirement for SAP expression in Ag-specific CD4 T cells to develop appropriate follicular helper T cell functions. It is unknown what signaling molecules are involved in regulation of SAP-dependent CD4 T cell help functions. SAP binds to the cytoplasmic tail of SLAM, and we show that SLAM is expressed on resting and activated CD4 T cells, as well as germinal center B cells. In addition, SAP can recruit Fyn kinase to SLAM. We have now examined the role(s) of the SLAM-SAP-Fyn signaling axis in in vivo CD4 T cell function and germinal center development. We observed normal germinal center development, long-lived plasma cell development, and Ab responses in SLAM(-/-) mice after a viral infection (lymphocytic choriomeningitis virus). In a separate series of experiments, we show that SAP is absolutely required in CD4 T cells to drive germinal center development, and that requirement does not depend on SAP-Fyn interactions, because CD4 T cells expressing SAP R78A are capable of supporting normal germinal center development. Therefore, a distinct SAP signaling pathway regulates follicular helper CD4 T cell differentiation, separate from the SLAM-SAP-Fyn signaling pathway regulating Th1/Th2 differentiation.
Collapse
Affiliation(s)
- Megan M McCausland
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92121, USA
| | | | | | | | | | | |
Collapse
|
7
|
Reading SA, Dimmock NJ. Neutralization of animal virus infectivity by antibody. Arch Virol 2007; 152:1047-59. [PMID: 17516034 DOI: 10.1007/s00705-006-0923-8] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2006] [Accepted: 12/11/2006] [Indexed: 11/24/2022]
Abstract
Neutralization is the ability of antibody to bind to and inactivate virus infectivity under defined conditions in vitro. Most neutralizing antibodies also protect animals in vivo, but protection is more complex as it also involves interaction of antibody with cells and molecules of the innate immune system. Neutralization by antibody can be mediated by a number of different mechanisms: by aggregation of virions, destabilization of the virion structure, inhibition of virion attachment to target cells, inhibition of the fusion of the virion lipid membrane with the membrane of the host cell, inhibition of the entry of the genome of non-enveloped viruses into the cell cytoplasm, inhibition of a function of the virion core through a signal transduced by an antibody, transcytosing IgA, and binding to nascent virions to block their budding or release from the cell surface. The mechanism of neutralization is determined by the properties of both a virion epitope and the antibody that reacts with it. Further, since a virus has at least several unique epitopes sited in different locations on the virion, and since the paratope and other properties of the reacting antibody can vary, this means that a virus can be neutralized by several different mechanisms. Understanding the processes of neutralization informs the creation of modern vaccines, and gives valuable insights into virus-cell interactions.
Collapse
Affiliation(s)
- S A Reading
- Department of Biological Sciences, University of Warwick, Coventry, UK.
| | | |
Collapse
|
8
|
Crotty S, McCausland MM, Aubert RD, Wherry EJ, Ahmed R. Hypogammaglobulinemia and exacerbated CD8 T-cell–mediated immunopathology in SAP-deficient mice with chronic LCMV infection mimics human XLP disease. Blood 2006; 108:3085-93. [PMID: 16788096 DOI: 10.1182/blood-2006-04-018929] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
AbstractThe human genetic disease X-linked lymphoproliferative disease (XLP), which is caused by mutations in SH2D1A/SAP that encode SLAM-associated protein (SAP), is characterized by an inability to control Epstein-Barr virus (EBV) and hypogammaglobulinemia. It is unclear which aspects of XLP disease are specific to herpesvirus infection and which reflect general immunologic functions performed by SAP. We examined SAP– mice during a chronic LCMV infection, specifically to address the following question: Which SAP deficiency immunologic problems are general, and which are EBV specific? Illness, weight loss, and prolonged viral replication were much more severe in SAP– mice. Aggressive immunopathology was observed. This inability to control chronic LCMV was associated with both CD8 T-cell and B-cell response defects. Importantly, we demonstrate that SAP– CD8 T cells are the primary cause of the immunopathology and clinical illness, because depletion of CD8 T cells blocked disease. This is the first direct demonstration of SAP– CD8 T-cell–mediated immunopathology, confirming 30 years of XLP clinical observations and indirect experimentation. In addition, germinal center formation was extremely defective in chronically infected SAP– animals, and hypogammaglobulinemia was observed. These findings in a chronic viral infection mouse model recapitulate key features of human XLP and clarify SAP's critical role regulating both cellular and humoral immunity.
Collapse
Affiliation(s)
- Shane Crotty
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA.
| | | | | | | | | |
Collapse
|
9
|
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
|
10
|
Drescher KM, Murray PD, David CS, Pease LR, Rodriguez M. CNS cell populations are protected from virus-induced pathology by distinct arms of the immune system. Brain Pathol 2006; 9:21-31. [PMID: 9989447 PMCID: PMC8098348 DOI: 10.1111/j.1750-3639.1999.tb00206.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: 11/29/2022] Open
Abstract
The basis for the distinct patterns of brain pathology in individuals experiencing virus-induced encephalitis may be related to either the tropism of the virus or the host's response to virus infection of the central nervous system (CNS). In these studies we used Theiler's murine encephalomyelitis virus (TMEV) and a series of mice deficient in various immune system components (alpha/beta T cells, antibody, Class I MHC, and Class II MHC) to examine the hypothesis that discrete populations of CNS cells are protected differentially from virus infection by distinct arms of the immune response. Here we demonstrate that the Class I-mediated immune response provided more protection from areas of the brain (brainstem, corpus callosum and cerebellum) with abundant white matter as there was significantly more disease in these areas in beta2m -/- (Class I-deficient) mice as compared to A beta(0) (Class II-deficient) mice. In contrast, the striatum, with an abundance of neurons, was protected from virus-induced pathology primarily by antibody. In addition, we determined that antibody and alpha/beta T cells provided protection from severe deficits and death during the acute phase of the disease. The data presented here support the hypothesis that distinct immune system components function to protect discrete areas of the CNS from virus-induced pathology.
Collapse
Affiliation(s)
- K M Drescher
- Department of Immunology, Mayo Medical School, Rochester, MN 55901, USA
| | | | | | | | | |
Collapse
|
11
|
Casadevall A, Pirofski LA. A Reappraisal of Humoral Immunity Based on Mechanisms of Antibody‐Mediated Protection Against Intracellular Pathogens. Adv Immunol 2006; 91:1-44. [PMID: 16938537 DOI: 10.1016/s0065-2776(06)91001-3] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Sometime in the mid to late twentieth century the study of antibody-mediated immunity (AMI) entered the doldrums, as many immunologists believed that the function of AMI was well understood, and was no longer deserving of intensive investigation. However, beginning in the 1990s studies using monoclonal antibodies (mAbs) revealed new functions for antibodies, including direct antimicrobial effects and their ability to modify host inflammatory and cellular responses. Furthermore, the demonstration that mAbs to several intracellular bacterial and fungal pathogens were protective issued a serious challenge to the paradigm that host defense against such microbes was strictly governed by cell-mediated immunity (CMI). Hence, a new view of AMI is emerging. This view is based on the concept that a major function of antibody (Ab) is to amplify or subdue the inflammatory response to a microbe. In this regard, the "damage-response framework" of microbial pathogenesis provides a new conceptual viewpoint for understanding mechanisms of AMI. According to this view, the ability of an Ab to affect the outcome of a host-microbe interaction is a function of its capacity to modify the damage ensuing from such an interaction. In fact, it is increasingly apparent that the efficacy of an Ab cannot be defined either by immunoglobulin or epitope characteristics alone, but rather by a complex function of Ab variables, such as specificity, isotype, and amount, host variables, such as genetic background and immune status, and microbial variables, such as inoculum, mechanisms of avoiding host immune surveillance and pathogenic strategy. Consequently, far from being understood, recent findings in AMI imply a system with unfathomable complexity and the field is poised for a long overdue renaissance.
Collapse
Affiliation(s)
- Arturo Casadevall
- Department of Medicine, Division of Infectious Diseases, Albert Einstein College of Medicine and Montefoire Medical Center, Bronx, New York, USA
| | | |
Collapse
|
12
|
Davies DH, McCausland MM, Valdez C, Huynh D, Hernandez JE, Mu Y, Hirst S, Villarreal L, Felgner PL, Crotty S. Vaccinia virus H3L envelope protein is a major target of neutralizing antibodies in humans and elicits protection against lethal challenge in mice. J Virol 2005; 79:11724-33. [PMID: 16140750 PMCID: PMC1212608 DOI: 10.1128/jvi.79.18.11724-11733.2005] [Citation(s) in RCA: 176] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The smallpox vaccine is the prototypic vaccine, yet the viral targets critical for vaccine-mediated protection remain unclear in humans. We have produced protein microarrays of a near-complete vaccinia proteome and used them to determine the major antigen specificities of the human humoral immune response to the smallpox vaccine (Dryvax). H3L, an intracellular mature virion envelope protein, was consistently recognized by high-titer antibodies in the majority of human donors, particularly after secondary immunization. We then focused on examining H3L as a valuable human antibody target. Purified human anti-H3L antibodies exhibited substantial vaccinia virus-neutralizing activity in vitro (50% plaque reduction neutralization test [PRNT50] = 44 microg/ml). Mice also make an immunodominant antibody response to H3L after vaccination with vaccinia virus, as determined by vaccinia virus protein microarray. Mice were immunized with recombinant H3L protein to examine H3L-specific antibody responses in greater detail. H3L-immunized mice developed high-titer vaccinia virus-neutralizing antibodies (mean PRNT50 = 1:3,760). Importantly, H3L-immunized mice were subsequently protected against lethal intranasal challenges with 1 or 5 50% lethal doses (LD50) of pathogenic vaccinia virus strain WR, demonstrating the in vivo value of an anti-H3L response. To formally demonstrate that neutralizing anti-H3L antibodies are protective in vivo, we performed anti-H3L serum passive-transfer experiments. Mice receiving H3L-neutralizing antiserum were protected from a lethal challenge with 3 LD50 of vaccinia virus strain WR (5/10 versus 0/10; P < 0.02). Together, these data show that H3L is a major target of the human anti-poxvirus antibody response and is likely to be a key contributor to protection against poxvirus infection and disease.
Collapse
Affiliation(s)
- D Huw Davies
- Center for Virus Research, Department of Molecular Biology and Biochemistry, McGaugh Hall, University of California, Irvine 92697, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Whitton JL, Slifka MK, Liu F, Nussbaum AK, Whitmire JK. The regulation and maturation of antiviral immune responses. Adv Virus Res 2005; 63:181-238. [PMID: 15530562 PMCID: PMC7125551 DOI: 10.1016/s0065-3527(04)63003-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- J Lindsay Whitton
- Department of Neuropharmacology, CVN-9, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | | | | |
Collapse
|
14
|
Abstract
Little has been known about the components of the immune system that are effective in the protection of a pig against PRRSV infection. Although antibodies were initially perceived as a deleterious, ineffective component of the PRRSV-specific immune response, neutralizing antibodies (NA) are now considered to be an important correlate of protective immunity against PRRSV. This paper reviews the current knowledge on arterivirus-specific NA, the role that NA have in protection against infection with PRRSV, as well as the viral molecular structures that are responsible for the production of this type of antibodies by the pig. This information should prove central to the design of new generation vaccines against PRRSV.
Collapse
Affiliation(s)
- O J Lopez
- Department of Veterinary and Biomedical Sciences, University of Nebraska-Lincoln, 141 VBS, East Campus, UNL, Lincoln, NE 68583-0905, USA
| | | |
Collapse
|
15
|
Affiliation(s)
- Arturo Casadevall
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY 10461, USA.
| | | |
Collapse
|
16
|
Casadevall A, Pirofski LA. Antibody-mediated regulation of cellular immunity and the inflammatory response. Trends Immunol 2003; 24:474-8. [PMID: 12967670 DOI: 10.1016/s1471-4906(03)00228-x] [Citation(s) in RCA: 101] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- Arturo Casadevall
- Department of Medicine, Division of Infectious Diseases, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, New York, NY 10461, USA.
| | | |
Collapse
|
17
|
Johansson M, Berg M, Berg AL. Humoral immune response against Borna disease virus (BDV) in experimentally and naturally infected cats. Vet Immunol Immunopathol 2002; 90:23-33. [PMID: 12406652 DOI: 10.1016/s0165-2427(02)00226-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
In order to investigate the peripheral and intracerebral humoral immune response against Borna disease virus (BDV) in cats, serum and cerebrospinal fluid (CSF) samples from experimentally and naturally BDV-infected cats were analysed in two different test systems (indirect enzyme-linked immunosorbent assay and indirect immunofluorescent test). The experimentally infected cats developed high antibody titres against the major immunogenic BDV-proteins, p24 and p40. In contrast, the naturally infected cats showed a comparatively weak humoral immune response. The experimentally infected cats were inoculated with either BDV laboratory strain V or a feline BDV-isolate. Some differences existed between the two groups of cats. The former group developed a higher response against p40, whereas the latter group showed, beside the p40-response, a more pronounced p24-response, similar to the situation in the naturally infected cats.
Collapse
Affiliation(s)
- M Johansson
- Department of Pathology, Swedish University of Agricultural Sciences (SUAS), 750 07, Uppsala, Sweden
| | | | | |
Collapse
|
18
|
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
|
19
|
Slifka MK. Mechanisms of humoral immunity explored through studies of LCMV infection. Curr Top Microbiol Immunol 2002; 263:67-81. [PMID: 11987820 DOI: 10.1007/978-3-642-56055-2_5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Affiliation(s)
- M K Slifka
- OHSU Vaccine and Gene Therapy Institute, 505 NW 185th Avenue, Beaverton, OR 97006, USA
| |
Collapse
|
20
|
Kunz S, Borrow P, Oldstone MBA. Receptor structure, binding, and cell entry of arenaviruses. Curr Top Microbiol Immunol 2002; 262:111-37. [PMID: 11987803 DOI: 10.1007/978-3-642-56029-3_5] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- S Kunz
- Department of Neuropharmacology, Division of Virology, Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
21
|
Liebert UG. Slow and persistent virus infections of neurones--a compromise for neuronal survival. Curr Top Microbiol Immunol 2001; 253:35-60. [PMID: 11417139 DOI: 10.1007/978-3-662-10356-2_3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Affiliation(s)
- U G Liebert
- Institute of Virology, University of Leipzig, Johannisallee 30, 04103 Leipzig, Germany
| |
Collapse
|
22
|
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
|
23
|
Seiler P, Senn BM, Klenerman P, Kalinke U, Hengartner H, Zinkernagel RM. Additive effect of neutralizing antibody and antiviral drug treatment in preventing virus escape and persistence. J Virol 2000; 74:5896-901. [PMID: 10846070 PMCID: PMC112085 DOI: 10.1128/jvi.74.13.5896-5901.2000] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Poorly cytopathic or noncytopathic viruses can escape immune surveillance and establish a chronic infection. Here we exploited the strategy of combining antiviral drug treatment with the induction of a neutralizing antibody response to avoid the appearance of neutralization-resistant virus variants. Despite the fact that H25 immunoglobulin transgenic mice infected with lymphocytic choriomeningitis virus mounted an early neutralizing antibody response, the virus escaped from neutralization and persisted. After ribavirin treatment of H25 transgenic mice, the appearance of neutralization-resistant virus was prevented and virus was cleared. Thus, the combination of virus-neutralizing antibodies and chemotherapy efficiently controlled the infection, whereas each defense line alone did not. Similar additive effects may be unexpectedly efficient and beneficial in humans after infections with persistent viruses such as hepatitis C virus and hepatitis B virus and possibly human immunodeficiency virus.
Collapse
Affiliation(s)
- P Seiler
- Department of Pathology, Institute of Experimental Immunology, University of Zurich, Switzerland.
| | | | | | | | | | | |
Collapse
|
24
|
López-Macías C, Kalinke U, Cascalho M, Wabl M, Hengartner H, Zinkernagel RM, Lamarre A. Secondary rearrangements and hypermutation generate sufficient B cell diversity to mount protective antiviral immunoglobulin responses. J Exp Med 1999; 189:1791-8. [PMID: 10359583 PMCID: PMC2193076 DOI: 10.1084/jem.189.11.1791] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Variable (V) region gene replacement was recently implicated in B cell repertoire diversification, but the contribution of this mechanism to antibody responses is still unknown. To investigate the role of V gene replacements in the generation of antigen-specific antibodies, we analyzed antiviral immunoglobulin responses of "quasimonoclonal" (QM) mice. The B cells of QM mice are genetically committed to exclusively express the anti-(4-hydroxy-3-nitrophenyl) acetyl specificity. However, approximately 20% of the peripheral B cells of QM mice undergo secondary rearrangements and thereby potentially acquire new specificities. QM mice infected with vesicular stomatitis virus (VSV), lymphocytic choriomeningitis virus, or poliovirus mounted virus-specific neutralizing antibody responses. In general, kinetics of the antiviral immunoglobulin responses were delayed in QM mice; however, titers similar to control animals were eventually produced that were sufficient to protect against VSV-induced lethal disease. VSV neutralizing single-chain Fv fragments isolated from phage display libraries constructed from QM mice showed VH gene replacements and extensive hypermutation. Thus, our data demonstrate that secondary rearrangements and hypermutation can generate sufficient B cell diversity in QM mice to mount protective antiviral antibody responses, suggesting that these mechanisms might also contribute to the diversification of the B cell repertoire of normal mice.
Collapse
Affiliation(s)
- C López-Macías
- Institute of Experimental Immunology, Department of Pathology, University Hospital, CH-8091 Zürich, Switzerland
| | | | | | | | | | | | | |
Collapse
|
25
|
Gall JG, Crystal RG, Falck-Pedersen E. Construction and characterization of hexon-chimeric adenoviruses: specification of adenovirus serotype. J Virol 1998; 72:10260-4. [PMID: 9811772 PMCID: PMC110610 DOI: 10.1128/jvi.72.12.10260-10264.1998] [Citation(s) in RCA: 88] [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
This study has used the strategy of gene replacement to characterize the contribution of the adenovirus (Ad) capsid protein hexon to serotype definition. By replacing the Ad type 5 (Ad5) hexon gene with sequences from Ad2, we have changed the type specificity of the chimeric virus. The type-determining epitopes are primarily associated with loop 1 of hexon and, to a much lesser degree, with loop 2. In spite of the serotype distinctiveness of the chimeric hexon viruses, epitope similarity between the vectors resulted in a low level of cross-reactive neutralizing antibody, which in combination with activated cellular and innate arms of the immune system is sufficient to suppress gene transduction following readministration in vivo.
Collapse
Affiliation(s)
- J G Gall
- Department of Microbiology, W. R. Hearst Research Foundation, Cornell University Medical College, New York, New York 10021, USA
| | | | | |
Collapse
|
26
|
Homann D, Tishon A, Berger DP, Weigle WO, von Herrath MG, Oldstone MB. Evidence for an underlying CD4 helper and CD8 T-cell defect in B-cell-deficient mice: failure to clear persistent virus infection after adoptive immunotherapy with virus-specific memory cells from muMT/muMT mice. J Virol 1998; 72:9208-16. [PMID: 9765468 PMCID: PMC110340 DOI: 10.1128/jvi.72.11.9208-9216.1998] [Citation(s) in RCA: 140] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Adoptive transfer of virus-specific memory lymphocytes can be used to identify factors and mechanisms involved in the clearance of persistent virus infections. To analyze the role of B cells in clearing persistent infection with lymphocytic choriomeningitis virus (LCMV), we used B-cell-deficient muMT/muMT (B-/-) mice. B-/- mice controlled an acute LCMV infection with the same kinetics and efficiency as B-cell-competent (B+/+) mice via virus-specific, major histocompatibility complex (MHC) class I-restricted CD8(+) cytotoxic T lymphocytes (CTL). CTL from B-/- and B+/+ mice were equivalent in affinity to known LCMV CTL epitopes and had similar CTL precursor frequencies (pCTL). Adoptive transfer of memory cells from B+/+ mice led to virus clearance from persistently infected B+/+ recipients even after in vitro depletion of B cells, indicating that B cells or immunoglobulins are not required in the transfer population. In contrast, transfer of memory splenocytes from B-/- mice failed to clear virus. Control of virus was restored neither by transferring higher numbers of pCTL nor by supplementing B-/- memory splenocytes with LCMV-immune B cells or immune sera. Instead, B-/- mice were found to have a profound CD4 helper defect. Furthermore, compared to cultured splenocytes from B+/+ mice, those from B-/- mice secreted less gamma interferon (IFN-gamma) and interleukin 2, with differences most pronounced for CD8 T cells. While emphasizing the importance of CD4 T-cell help and IFN-gamma in the control of persistent infections, the CD4 T-helper and CD8 T-cell defects in B-/- mice suggest that B cells contribute to the induction of competent T effector cells.
Collapse
Affiliation(s)
- D Homann
- Division of Virology, Department of Neuropharmacology, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | | | | | |
Collapse
|
27
|
Hooper DC, Morimoto K, Bette M, Weihe E, Koprowski H, Dietzschold B. Collaboration of antibody and inflammation in clearance of rabies virus from the central nervous system. J Virol 1998; 72:3711-9. [PMID: 9557653 PMCID: PMC109593 DOI: 10.1128/jvi.72.5.3711-3719.1998] [Citation(s) in RCA: 170] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/1997] [Accepted: 01/23/1998] [Indexed: 02/07/2023] Open
Abstract
To investigate the involvement of various cellular and humoral aspects of immunity in the clearance of rabies virus from the central nervous system, (CNS), we studied the development of clinical signs and virus clearance from the CNS in knockout mice lacking either B and T cells, CD8+ cytotoxic T cells, B cells, alpha/beta interferon (IFN-alpha/beta) receptors, IFN-gamma receptors, or complement components C3 and C4. Following intranasal infection with the attenuated rabies virus CVS-F3, normal adult mice of different genetic backgrounds developed a transient disease characterized by loss of body weight and appetite depression which peaked at 13 days postinfection (p.i.). While these animals had completely recovered by day 21 p.i., mice lacking either B and T cells or B cells alone developed a progressive disease and succumbed to infection. Mice lacking either CD8+ T cells, IFN receptors, or complement components C3 and C4 showed no significant differences in the development of clinical signs by comparison with intact counterparts having the same genetic background. However, while infectious virus and viral RNA could be detected in normal control mice only until day 8 p.i., in all of the gene knockout mice studied except those lacking C3 and C4, virus infection persisted through day 21 p.i. Analysis of rabies virus-specific antibody production together with histological assessment of brain inflammation in infected animals revealed that clearance of CVS-F3 by 21 days p.i. correlated with both a strong inflammatory response in the CNS early in the infection (day 8 p.i.), and the rapid (day 10 p.i.) production of significant levels of virus-neutralizing antibody (VNA). These studies confirm that rabies VNA is an absolute requirement for clearance of an established rabies virus infection. However, for the latter to occur in a timely fashion, collaboration between VNA and inflammatory mechanisms is necessary.
Collapse
Affiliation(s)
- D C Hooper
- Center for Neurovirology, Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107-6799, USA
| | | | | | | | | | | |
Collapse
|
28
|
Seiler P, Kalinke U, Rülicke T, Bucher EM, Böse C, Zinkernagel RM, Hengartner H. Enhanced virus clearance by early inducible lymphocytic choriomeningitis virus-neutralizing antibodies in immunoglobulin-transgenic mice. J Virol 1998; 72:2253-8. [PMID: 9499083 PMCID: PMC109522 DOI: 10.1128/jvi.72.3.2253-2258.1998] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Following infection of mice with lymphocytic choriomeningitis virus (LCMV), virus-neutralizing antibodies appear late, after 30 to 60 days. Such neutralizing antibodies play an important role in protection against reinfection. To analyze whether a neutralizing antibody response which developed earlier could contribute to LCMV clearance during the acute phase of infection, we generated transgenic mice expressing LCMV-neutralizing antibodies. Transgenic mice expressing the immunoglobulin mu heavy chain of the LCMV-neutralizing monoclonal antibody KL25 (H25 transgenic mice) mounted LCMV-neutralizing immunoglobulin M (IgM) serum titers within 8 days after infection. This early inducible LCMV-neutralizing antibody response significantly improved the host's capacity to clear the infection and did not cause an enhancement of disease after intracerebral (i.c.) LCMV infection. In contrast, mice which had been passively administered LCMV-neutralizing antibodies and transgenic mice exhibiting spontaneous LCMV-neutralizing IgM serum titers (HL25 transgenic mice expressing the immunoglobulin mu heavy and the kappa light chain) showed an enhancement of disease after i.c. LCMV infection. Thus, early-inducible LCMV-neutralizing antibodies can contribute to viral clearance in the acute phase of the infection and do not cause antibody-dependent enhancement of disease.
Collapse
Affiliation(s)
- P Seiler
- Department of Pathology, Institute of Experimental Immunology, University of Zurich, Switzerland.
| | | | | | | | | | | | | |
Collapse
|
29
|
Bachmann MF, Kalinke U, Althage A, Freer G, Burkhart C, Roost H, Aguet M, Hengartner H, Zinkernagel RM. The role of antibody concentration and avidity in antiviral protection. Science 1997; 276:2024-7. [PMID: 9197261 DOI: 10.1126/science.276.5321.2024] [Citation(s) in RCA: 206] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Neutralizing antibodies are necessary and sufficient for protection against infection with vesicular stomatitis virus (VSV). The in vitro neutralization capacities and in vivo protective capacities of a panel of immunoglobulin G monoclonal antibodies to the glycoprotein of VSV were evaluated. In vitro, neutralizing activity correlated with avidity and with neutralization rate constant, a measure of on-rate. However, in vivo, protection was independent of immunoglobulin subclass, avidity, neutralization rate constant, and in vitro neutralizing activity; above a minimal avidity threshold, protection depended simply on a minimum serum concentration. These two biologically defined thresholds of antibody specificity offer hope for the development of adoptive therapy with neutralizing antibodies.
Collapse
Affiliation(s)
- M F Bachmann
- Institute of Experimental Immunology, Department of Pathology, University Hospital Zürich, Schmelzbergstrasse 12, CH-8091 Zürich, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Baldridge JR, McGraw TS, Paoletti A, Buchmeier MJ. Antibody prevents the establishment of persistent arenavirus infection in synergy with endogenous T cells. J Virol 1997; 71:755-8. [PMID: 8985412 PMCID: PMC191113 DOI: 10.1128/jvi.71.1.755-758.1997] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
A cardinal feature of the biology of lymphocytic choriomeningitis virus (LCMV) is its ability to establish persistent infections in mice. Persistence is usually established by infection of the mouse during the in utero or neonatal period. Susceptibility can be extended to the adult by treatment with immunosuppressive agents or by infection with immunosuppressive strains of LCMV. In this study we investigated the capacity of passively acquired anti-LCMV antibodies to prevent the establishment of persistence in both neonatal and adult mice. Suckling BALB/c mouse pups nursed by mothers immunized against LCMV before pregnancy had higher survival rates following infection than controls and withstood challenge doses of up to 400 PFU without becoming persistently infected. To establish that maternal antibody alone and not maternally derived T cells provided this protection, nonimmune mothers were infused with monoclonal anti-LCMV neutralizing antibodies within 24 h after delivering their pups. Pups nursing on these passively immunized mothers were resistant to persistent LCMV infection. The establishment of persistence in adult BALB/c mice by the immunosuppressive, macrophage-tropic LCMV variant, clone 13 was also prevented by prophylactic treatment with anti-LCMV monoclonal antibodies. However, the protection afforded by passively acquired antibody was found to be incomplete if the recipients lacked functional CD8+ T cells. While 65% of neonatal athymic (nu/nu) mice nursed by immune nu/+ dams resisted low-dose viral challenge (25 PFU), the majority of nude pups challenged with high doses of virus (100 PFU) became persistently infected. Also, protection was incomplete in beta2-microglobulin knockout mice, which lack functional CD8+ T cells, suggesting that a cooperative effect was exerted by the combination of neutralizing antibody and endogenous T cells. These results indicate that antibodies provide an effective barrier to the establishment of persistent infections in immunocompetent mice and reaffirm that vaccines which induce strong humoral responses may provide efficient protection against arenavirus infections.
Collapse
Affiliation(s)
- J R Baldridge
- Department of Neuropharmacology, Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | |
Collapse
|
31
|
Abstract
Neutralizing antiviral B cell responses differ in various aspects from the many usually measured B cell responses specific for protein in adjuvants. In particular, such neutralizing antiviral B cell responses are more rapidly induced, reach higher titers, are longer lived, and are efficiently generated without adjuvants. Evidence is summarized here that the repetitiveness of many viral antigens is a key factor responsible for the efficiency of these B cell responses, amplifying B cells early and rapidly for potent IgM responses and also for efficient switching to IgG. The data reviewed indicate that B cells discriminate antigen patterns via the degree of surface Ig-cross-linking and use antigen repetitiveness as a self/nonself discriminator.
Collapse
Affiliation(s)
- M F Bachmann
- Department of Pathology, University of Zürich, Switzerland
| | | |
Collapse
|
32
|
Bründler MA, Aichele P, Bachmann M, Kitamura D, Rajewsky K, Zinkernagel RM. Immunity to viruses in B cell-deficient mice: influence of antibodies on virus persistence and on T cell memory. Eur J Immunol 1996; 26:2257-62. [PMID: 8814275 DOI: 10.1002/eji.1830260943] [Citation(s) in RCA: 81] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Mice rendered B cell deficient by targeted disruption of the immunoglobulin mu chain gene (IgM-/- mice) were used to analyze the role of antibodies and B cells in viral infections; homozygous IgM-/- mice were bred in a way to avoid transmission of maternal antibodies. After infection with vesicular stomatitis virus (VSV), IgM-/- mice developed paralytic disease and subsequently died, whereas C57BL/6 control mice or IgM-/- mice passively protected with VSV-neutralizing antibodies survived. Furthermore, IgM-/- mice showed increased natural killer (NK) activity upon exposure to either lymphocytic choriomeningitis virus (LCMV) or to poly(I).poly(C), while NK activity in untreated IgM-/- mice was within normal ranges. Cytotoxic T cell responses were comparable in IgM-/- and control mice infected either with VSV or with vaccinia virus or with low doses of LCMV (10(2) infectious focus-forming units [ifu]). After intracerebral infection with LCMV-Armstrong, CD8+ T cell-mediated lethal lymphocytic choriomeningitis developed independently of the presence of B cells and antibodies. After infection with high doses (2 x 10(6) - 5 x 10(6) ifu) of LCMV-WE or LCMV-Docile, IgM-/- mice exhibited a reduced capacity to control these primary infections and had elevated virus titers for prolonged times (> 60 days). Nevertheless, the cytotoxic T cell response against LCMV in the early phase of infection was comparable in IgM-/- and control mice, but disappeared in those IgM-/- mice which had a persistent viral infection. Cytotoxic T cell memory was apparently unimpaired in low-dose-primed IgM-/- mice, which were able to control the primary virus infection; both IgM-/- and control mice cleared a high intravenous dose of virus within 2 days after challenge infection. This indicates that an efficient T cell memory against LCMV was established in the absence of B cells.
Collapse
Affiliation(s)
- M A Bründler
- Department of Pathology, University of Zurich, Switzerland
| | | | | | | | | | | |
Collapse
|
33
|
Abstract
Immunological memory is a hallmark of the immune system. Evolution can teach us which effector arms of immunological memory are biologically relevant against which virus. Antibodies appear to be the critical protective mechanism against cytopathic viruses. Since these viruses cause cell damage and disease directly, particularly in the absence of an immune response, mothers protect their offspring during a critical immunoincompetent period (a consequence of MHC- restricted T cell recognition) by passive transfer of neutralizing antibodies. In contrast, CTL appear to be the crucial effector mechanism against noncytopathic viruses. Since MHC polymorphism has made vertical transmission of T cell memory impossible, immunoincompetent offspring are not, and need not be, protected against such noncytopathic viruses. During the primary response and again during secondary infection, the most important function of CTL is to eliminate noncytopathic viruses, which may otherwise cause lethal immunopathology. Increased precursor frequencies of B and T cells appear to remain in the host independent of antigen persistence. However, in order to protect against cytopathic viruses, memory B cells have to produce antibody to maintain protective elevated levels of antibody; B cell differentiation into plasma cells is driven by persisting antigen. Similarly, to protect against infection with a noncytopathic virus, CTL have to recirculate through peripheral organs. Activation and capacity to emigrate into solid tissues as well as cytolytic effector function are also dependent upon, and driven by, persisting antigen. Because no convincing evidence is available yet of the existence of identifiable B or T cells with specialized memory characteristics, the phenotype of immunological memory correlates best with antigen-driven activation of low frequency effector T cells and plasma cells.
Collapse
|
34
|
von Herrath MG, Yokoyama M, Dockter J, Oldstone MB, Whitton JL. CD4-deficient mice have reduced levels of memory cytotoxic T lymphocytes after immunization and show diminished resistance to subsequent virus challenge. J Virol 1996; 70:1072-9. [PMID: 8551565 PMCID: PMC189913 DOI: 10.1128/jvi.70.2.1072-1079.1996] [Citation(s) in RCA: 223] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Although primary antiviral CD8+ cytotoxic T lymphocytes (CTL) can be induced in mice depleted of CD4+ T cells, the role of CD4+ T lymphocytes in the generation and maintenance of antiviral memory CTL is uncertain. This question, and the consequences upon vaccine-mediated protection, were investigated in transgenic CD4 knockout (CD4ko) mice, which lack CD4+ T lymphocytes. Infection of immunocompetent C57BL/6 mice with lymphocytic choriomeningitis virus (LCMV), or with recombinant vaccinia viruses bearing appropriate LCMV sequences, induces long-lasting protective immunity, mediated mainly by antiviral CD8+ CTL. Here we report two important findings. First, LCMV-specific CD8+ memory CTL are maintained at considerably lower levels in CD4ko mice than in normal C57BL/6J mice; we demonstrate a reduction in precursor CTL evident as soon as 30 days postimmunization and declining, by day 120, to levels 1 to 2 log units below those in normal mice. Thus, CD4+ T cells appear to be important to the generation and maintenance of their CD8+ counterparts. Second, this reduction has an important biological consequence; compared with immunocompetent mice, CD4ko mice immunized with vaccinia virus recombinants expressing nucleoprotein or glycoprotein of LCMV are less effectively protected from subsequent LCMV challenge. Thus, this study underscores the potential importance of CD4+ T lymphocytes in generation of appropriate levels of CD(8+)-cell-mediated immunoprotective memory and has implications for vaccine efficacy in individuals with immune defects in which CD4 levels may be reduced, such as AIDS.
Collapse
Affiliation(s)
- M G von Herrath
- Department of Neuropharmacology, Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | | | |
Collapse
|
35
|
Bautista JR, Rubin SA, Moran TH, Schwartz GJ, Carbone KM. Developmental injury to the cerebellum following perinatal Borna disease virus infection. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 1995; 90:45-53. [PMID: 8719328 DOI: 10.1016/0165-3806(96)83485-7] [Citation(s) in RCA: 71] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
In rats infected as neonates, Borna disease virus (BDV) infection causes neuroanatomical, behavioral and physiological abnormalities without encephalitis. Neonatal infection with BDV provides a powerful model for studying the effects of virus replication on brain development without inflammation-induced brain damage. Here we report that neonatal BDV infection interfered with cerebellar development in the Lewis rat. Based on cerebellar cross-sectional area measurements, abnormal cerebellar growth was first noted between 7 and 14 days after infection. Reactive astrocytosis was evident by three days after infection, even without encephalitis, and even before identification of viral proteins in the cerebellum. While neonatal BDV infection caused a significant loss in granule cells, infected granule cells were not identified. BDV proteins were readily detected in the Purkinje cells. Thus, persistent BDV infection of Purkinje cells, but not granule cells, was associated with loss of granule cells during cerebellar development, in the absence of encephalitis.
Collapse
Affiliation(s)
- J R Bautista
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | | | |
Collapse
|
36
|
Quinn DG, Zajac AJ, Frelinger JA. The cell-mediated immune response against lymphocytic choriomeningitis virus in beta 2-microglobulin deficient mice. Immunol Rev 1995; 148:151-69. [PMID: 8825286 DOI: 10.1111/j.1600-065x.1995.tb00097.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- D G Quinn
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill 27599-7290, USA
| | | | | |
Collapse
|
37
|
|
38
|
Borrow P, Evans CF, Oldstone MB. Virus-induced immunosuppression: immune system-mediated destruction of virus-infected dendritic cells results in generalized immune suppression. J Virol 1995; 69:1059-70. [PMID: 7815484 PMCID: PMC188677 DOI: 10.1128/jvi.69.2.1059-1070.1995] [Citation(s) in RCA: 247] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Despite the clinical importance of virus-induced immunosuppression, how virus infection may lead to a generalized suppression of the host immune response is poorly understood. To elucidate the principles involved, we analyzed the mechanism by which a lymphocytic choriomeningitis virus (LCMV) variant produces a generalized immune suppression in its natural host, the mouse. Whereas adult mice inoculated intravenously with LCMV Armstrong rapidly clear the infection and remain immunocompetent, inoculation with the Armstrong-derived LCMV variant clone 13, which differs from its parent virus at only two amino acid positions, by contrast results in persistent infection and a generalized deficit in responsiveness to subsequent immune challenge. Here we show that the immune suppression induced by LCMV clone 13 is associated with a CD8-dependent loss of interdigitating dendritic cells from periarteriolar lymphoid sheaths in the spleen and, functionally, with a deficit in the ability of splenocytes from infected mice to stimulate the proliferation of naive T cells in a primary mixed lymphocyte reaction. Dendritic cells are not depleted in immunocompetent Armstrong-infected mice. LCMV Armstrong and clone 13 exhibit differences in their tropism within the spleen, with clone 13 causing a higher level of infection of antigen-presenting cells in the white pulp, including periarterial interdigitating dendritic cells, than Armstrong, thereby rendering these cells targets for destruction by the antiviral CD8+ cytotoxic T-lymphocyte response which is induced at early times following infection with either virus. Our findings illustrate the key role that virus tropism may play in determining pathogenicity and, further, document a mechanism for virus-induced immunosuppression which may contribute to the clinically important immune suppression associated with many virus infections, including human immunodeficiency virus type 1.
Collapse
Affiliation(s)
- P Borrow
- Department of Neuropharmacology, Scripps Research Institute, La Jolla, California 92037
| | | | | |
Collapse
|
39
|
Goldman MB, O'Bryan TA, Buckthal DJ, Tetor LM, Goldman JN. Suppression of measles virus expression by noncytolytic antibody in an immortalized macrophage cell line. J Virol 1995; 69:734-40. [PMID: 7815537 PMCID: PMC188636 DOI: 10.1128/jvi.69.2.734-740.1995] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Immune regulation of measles virus (MV) expression was studied in a persistently infected mouse macrophage cell line. Synthesis of both membrane-associated and internal MV antigens was suppressed when infected macrophages were treated with polyclonal rabbit anti-MV antibody that was specific for MV proteins. Persistently infected macrophages were treated for 3, 5, or 7 days with increasing doses of anti-MV antibody. All MV proteins were down-regulated 2 days after treatment was terminated. One week after treatment was terminated, down-regulation was still evident but to a lesser degree. MV protein synthesis was suppressed whether or not complement components were inactivated by heating all serum supplements and antibodies. However, when complement was active, cell lysis accounted for some of the reduced MV protein synthesis. When lytic destruction of infected cells by antibody and complement was prevented by inactivation of complement, antibody alone reduced the cellular synthesis of viral proteins by noncytolytic mechanisms. The absence of cell death in the absence of complement was confirmed by the lack of 51Cr release from labeled cells, the lack of reduction in cell number, and the lack of a decrease in total protein synthesis when radiolabeled infected cells were treated with antibody. It is noteworthy that low doses of antibody were optimal for suppression in the longer-term experiments and did not cause lysis, even in the presence of active complement. Since infected macrophages disseminate virus in measles infection, noncytolytic regulation of these cells by antibody may supplement viral clearance by cytolytic T cells and other immune mechanisms.
Collapse
Affiliation(s)
- M B Goldman
- Department of Medicine, Pennsylvania State University, Hershey 17033
| | | | | | | | | |
Collapse
|
40
|
Battegay M, Kyburz D, Hengartner H, Zinkernagel RM. Enhancement of disease by neutralizing antiviral antibodies in the absence of primed antiviral cytotoxic T cells. Eur J Immunol 1993; 23:3236-41. [PMID: 8258339 DOI: 10.1002/eji.1830231229] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The effects of neutralizing antibodies on the disease course in mice infected with the noncytopathic lymphocytic choriomeningitis virus (LCMV) were evaluated. Whereas non-neutralizing antisera exhibiting high enzyme-linked immunosorbent assay titers had no effect on T cell responses and their consequences, neutralizing antisera modulated them variably. Neutralizing antibodies were able to prevent lethal choriomeningitis after intracerebral infection with a neurotropic LCMV-isolate (ARMSTRONG) although they could not control local virus replication. The same antibodies exhibited little or no protective effect on choriomeningitis induced by LCMV-WE, a viscerotrope isolate. Surprisingly, these antibodies rendered mice much more susceptible to choriomeningitis after intracerebral infection with LCMV DOCILE, a very rapidly spreading lymphocyto-viscerotrope virus; in this situation antibodies prevented overwhelming infection which causes deletion of immunopathogenic cytotoxic T cell responses. Thus preexisting neutralizing antiviral antibodies had little influence on local virus spread in peripheral tissues but they reduced hematogenic spread and infection of antigen-presenting cells; thereby they influenced the primary cytotoxic T cell (CTL) response and indirectly modulated the extent of T cell-mediated immunopathology in peripheral organs. These results may explain why vaccines inducing neutralizing antibodies but no CTL may enhance an immunopathological disease caused by challenge infection with a noncytopathic virus.
Collapse
Affiliation(s)
- M Battegay
- Department of Pathology, University of Zürich, Switzerland
| | | | | | | |
Collapse
|
41
|
Weibel D, Bründler MA, Zinkernagel RM, Battegay M. Analysis of neutralizing antibody specificities of different strains of lymphocytic choriomeningitis virus with strain-specific immune sera. RESEARCH IN VIROLOGY 1993; 144:471-8. [PMID: 8140290 DOI: 10.1016/s0923-2516(06)80062-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The present study was aimed at comparing specificities and cross-reactivities of immune mouse sera obtained from mice infected with a low (2 x 10(2) or 20 immunological infectious focus units (ifu) or high (2 x 10(6) ifu) dose of various strains of lymphocytic choriomeningitis virus (LCMV). Neutralization titres of the various antisera were determined by an infectious focus reduction assay. This assay was performed on MC57G and on Vero E6 cells using the commonly utilized laboratory strains of LCMV WE, Docile, Armstrong, Armstrong Clone 13, Pasteur, Traub and Aggressive. Experiments with immune sera revealed broad cross-neutralization, demonstrating a variable but close serological relationship between the various strains of LCMV.
Collapse
Affiliation(s)
- D Weibel
- Institute of Experimental Immunology, University of Zurich, Switzerland
| | | | | | | |
Collapse
|
42
|
Abstract
The outcome of a viral infection within the nervous system depends on a complex interplay between the virus, its target cell and the immune system. Recent research has elucidated a variety of mechanisms involved in these interactions and their role in the production of disease.
Collapse
Key Words
- adcc, antibody-dependent cell-mediated cytotoxicity
- cns, central nervous system
- csf, cerebrospinal fluid
- ctl, cytotoxic t lymphocytes
- hiv, human immunodeficiency virus
- hsv, herpes simplex virus
- il, interleukin
- lcmv, lymphocytec choriomeningitis virus
- mhc, major histocompatibility complex
- scid, severe combined immune deficiency
- tgf, transforming growth factor
- tnf, tumor necrosis factor
Collapse
Affiliation(s)
- S B Wharton
- Department of Pathology, University of Cambridge, UK
| | | |
Collapse
|
43
|
Zinkernagel RM, Moskophidis D, Kündig T, Oehen S, Pircher H, Hengartner H. Effector T-cell induction and T-cell memory versus peripheral deletion of T cells. Immunol Rev 1993; 133:199-223. [PMID: 8225368 DOI: 10.1111/j.1600-065x.1993.tb01517.x] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- R M Zinkernagel
- Institute of Experimental Immunology, University of Zurich, Switzerland
| | | | | | | | | | | |
Collapse
|
44
|
Abstract
The novel role of antibody in clearing virus from the central nervous system without the help of other immune effectors is an important phenomenon that has only recently been documented. Possible routes for antibodies across the blood-brain barrier and how they work in the CNS are discussed here.
Collapse
Affiliation(s)
- B Dietzschold
- Center of Neurovirology, Thomas Jefferson University, Philadelphia, PA 19107
| |
Collapse
|
45
|
Oehen S, Ohashi PS, Aichele P, Bürki K, Hengartner H, Zinkernagel RM. Vaccination or tolerance to prevent diabetes. Eur J Immunol 1992; 22:3149-53. [PMID: 1446706 DOI: 10.1002/eji.1830221218] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Experiments with transgenic mice expressing the glycoprotein (GP) of lymphocytic choriomeningitis virus (LCMV) under the control of the rat insulin promoter (RIP) have demonstrated that potentially self-reactive T cells that normally ignore self peptides may nevertheless be induced by self peptides or "cross-reactive" foreign (e.g. viral) peptides that arise in the host in an immunogenic form; once activated these potentially self-reactive T cells may cause autoaggressive diseases (e.g. diabetes). The possibility of vaccinating against such T cell-mediated immunopathological diseases was evaluated in the RIP-GP transgenic mouse line Bln. Any attempt to vaccinate with the self antigen itself (e.g. recombinant vaccinia virus expressing LCMV-GP) failed to protect mice from disease. However, immunization with a recombinant vaccinia virus expressing LCMV-nucleoprotein (vacc-NP) as a non-GP LCMV vaccine was able to modulate the immune response and prevented autoaggressive disease in a MHC-dependent fashion. In contrast, tolerance induction neonatally or, more generally applicable, by lethal irradiation and reconstitution with neo-self antigen-expressing bone marrow cells always resulted in prevention of virally induced diabetes in this model situation.
Collapse
Affiliation(s)
- S Oehen
- Department of Pathology, University of Zurich, Switzerland
| | | | | | | | | | | |
Collapse
|
46
|
Borrow P, Oldstone MB. Characterization of lymphocytic choriomeningitis virus-binding protein(s): a candidate cellular receptor for the virus. J Virol 1992; 66:7270-81. [PMID: 1331520 PMCID: PMC240431 DOI: 10.1128/jvi.66.12.7270-7281.1992] [Citation(s) in RCA: 102] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The attachment of lymphocytic choriomeningitis virus (LCMV) to murine and primate cell lines was quantitated by a fluorescence-activated cell sorter assay in which binding of biotinylated virus was detected with streptavidin-fluorescein isothiocyanate. Cell lines that were readily infected by LCMV (e.g., MC57, Rin, BHK, Vero, and HeLa) bound virus in a dose-dependent manner, whereas no significant binding was observed to lymphocytic cell lines (e.g., RMA and WIL 2) that were not readily infected. Binding was specific and competitively blocked by nonbiotinylated LCMV. It was also blocked by LCMV-specific antiserum and a neutralizing monoclonal antibody to the virus glycoprotein GP-1 but not by antibodies specific for GP-2, indicating that attachment was likely mediated by GP-1. Treatment of cells with any of several proteases abolished LCMV binding, whereas phospholipases including phosphatidylinositol-specific phospholipase C had no effect, indicating that one or more membrane proteins were involved in virus attachment. These proteins were characterized with a virus overlay protein blot assay. Virus bound to protein(s) with a molecular mass of 120 to 140 kDa in membranes from cell lines permissive for LCMV but not from nonpermissive cell lines. Binding was specific, since unlabeled LCMV, but not the unrelated enveloped virus herpes simplex virus type 1, competed with 125I-labeled LCMV for binding to the 120- to 140-kDa band. The proteinaceous nature of the LCMV-binding substance was confirmed by the lack of virus binding to proteinase K-treated membrane components. By contrast, glycosidase treatment of membranes did not abolish virus binding. However, in membranes treated with endoglycosidase F/N-glycosidase F, and/or neuraminidase and in membranes from cells grown in tunicamycin, the molecular mass of the LCMV-binding entity was reduced. Hence, LCMV attachment to rodent fibroblastic cell lines is mediated by a glycoprotein(s) with a molecular mass of 120 to 140 kDa, with complex N-linked sugars that are not involved in virus binding.
Collapse
Affiliation(s)
- P Borrow
- Department of Neuropharmacology, Scripps Research Institute, La Jolla, California 92037
| | | |
Collapse
|
47
|
Dietzschold B, Kao M, Zheng YM, Chen ZY, Maul G, Fu ZF, Rupprecht CE, Koprowski H. Delineation of putative mechanisms involved in antibody-mediated clearance of rabies virus from the central nervous system. Proc Natl Acad Sci U S A 1992; 89:7252-6. [PMID: 1496020 PMCID: PMC49684 DOI: 10.1073/pnas.89.15.7252] [Citation(s) in RCA: 111] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The in vitro biological activities of several rabies virus-neutralizing monoclonal antibodies (mAbs) were compared with their ability to prevent a lethal rabies virus encephalomyelitis. The protective activity of a particular mAb in vivo did not correlate with its virus-neutralizing activity in vitro; rather it was related to the mAb's ability to inhibit virus spread from cell to cell and to restrict rabies virus RNA transcription. Since treatment of rabies virus-infected cells with virus-neutralizing mAbs results in an endocytosis of the antibody, we hypothesize that an antibody may exert its inhibitory activity even after uptake by the cell. Post-exposure treatment of rats with a mAb that inhibited both virus spread and virus RNA transcription in vitro resulted in viral clearance from the central nervous system and protected the animals against a lethal rabies virus infection.
Collapse
MESH Headings
- Actins/genetics
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Viral/immunology
- Base Sequence
- Blotting, Northern
- Blotting, Southern
- Brain/microbiology
- Capsid/genetics
- Cell Line
- Female
- Mice
- Mice, Inbred ICR
- Molecular Sequence Data
- Neuroblastoma
- Neutralization Tests
- Oligonucleotide Probes
- Polymerase Chain Reaction/methods
- RNA, Messenger/analysis
- RNA, Messenger/genetics
- RNA, Viral/genetics
- RNA, Viral/isolation & purification
- Rabies/immunology
- Rabies virus/genetics
- Rabies virus/immunology
- Rabies virus/isolation & purification
- Rats
- Rats, Inbred Lew
- Transcription, Genetic
- Viral Core Proteins/genetics
Collapse
Affiliation(s)
- B Dietzschold
- Wistar Institute of Anatomy and Biology, Philadelphia, PA 19109
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Baldridge JR, Buchmeier MJ. Mechanisms of antibody-mediated protection against lymphocytic choriomeningitis virus infection: mother-to-baby transfer of humoral protection. J Virol 1992; 66:4252-7. [PMID: 1376367 PMCID: PMC241229 DOI: 10.1128/jvi.66.7.4252-4257.1992] [Citation(s) in RCA: 74] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The role of antiviral antibodies in resistance to lymphocytic choriomeningitis virus (LCMV) infection was explored. Immune serum and monoclonal antibodies prevented fatal T-cell-mediated immunopathology following acute LCMV infections. In addition, 10- and 14-day-old mice that received maternally derived anti-LCMV antibodies through nursing were protected from an otherwise lethal LCMV challenge. Detailed investigation of the mechanism(s) by which these antiviral antibodies provided was carried out by using anti-LCMV monoclonal antibodies. Protection correlated directly with the ability of the antibodies to reduce viral titers in the tissues of conventional (K. E. Wright and M. J. Buchmeier, J. Virol. 65:3001-3006, 1991) and nude mice. However, this reduction was not simply a reflection of virus neutralizing activity, since not all antibodies which neutralized in vitro were protective. A correlation was also found between immunoglobulin isotype and protection: all of the protective antibodies were immunoglobulin G2a (IgG2a), while IgG1 antibodies mapping to the same epitopes were not. Protection appeared to be associated with events controlled by the Fc region. Functional F(ab')2 fragments which retained in vitro neutralizing activity were not protective in vivo. Furthermore, this Fc-associated function was not related to complement-mediated cell lysis, since C5-deficient mouse strains were also protected. These results suggest a role for antibody in protection from arenavirus infections and indicate that a distinct immunoglobulin subclass, IgG2a, may be essential for this protection.
Collapse
Affiliation(s)
- J R Baldridge
- Department of Neuropharmacology, Scripps Research Institute, La Jolla, California 92037
| | | |
Collapse
|
49
|
Graham BS, Bunton LA, Rowland J, Wright PF, Karzon DT. Respiratory syncytial virus infection in anti-mu-treated mice. J Virol 1991; 65:4936-42. [PMID: 1908028 PMCID: PMC248955 DOI: 10.1128/jvi.65.9.4936-4942.1991] [Citation(s) in RCA: 60] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BALB/c mice were depleted of B cells by anti-mu treatment to investigate the pathogenesis of respiratory syncytial virus (RSV) infection in the absence of antibody. Termination of RSV replication after primary infection occurred with the same kinetics in anti-mu-treated mice as in phosphate-buffered saline (PBS)-treated controls. Yet, when rechallenged, anti-mu-treated mice were more permissive to RSV replication than PBS-treated controls. Anti-mu-treated mice also experienced greater illness than PBS-treated controls during both primary infection and rechallenge. Passive transfer of RSV-specific immune serum to anti-mu-treated mice before rechallenge reconstituted complete protection from RSV replication and diminished illness. Thus, RSV-specific antibody is not required to terminate RSV replication in primary infection, but without antibody, only partial immunity against rechallenge is induced. While it is unknown whether the mechanism is a direct effect on RSV titer or modulation of the illness-causing cellular immune response, the presence of RSV-specific antibody reduces illness in both primary RSV infection and rechallenge of mice.
Collapse
Affiliation(s)
- B S Graham
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-2605
| | | | | | | | | |
Collapse
|