首页 | 本学科首页   官方微博 | 高级检索  
     


Neutralizing Monoclonal Antibodies Directed against Defined Linear Epitopes on Domain 4 of Anthrax Protective Antigen
Authors:Cassandra D. Kelly-Cirino  Nicholas J. Mantis
Affiliation:Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, and Department of Biomedical Sciences, School of Public Health, University at Albany, Albany, New York 12208
Abstract:The anthrax protective antigen (PA) is the receptor-binding subunit common to lethal toxin (LT) and edema toxin (ET), which are responsible for the high mortality rates associated with inhalational Bacillus anthracis infection. Although recombinant PA (rPA) is likely to be an important constituent of any future anthrax vaccine, evaluation of the efficacies of the various candidate rPA vaccines is currently difficult, because the specific B-cell epitopes involved in toxin neutralization have not been completely defined. In this study, we describe the identification and characterization of two murine monoclonal immunoglobulin G1 antibodies (MAbs), 1-F1 and 2-B12, which recognize distinct linear neutralizing epitopes on domain 4 of PA. 1-F1 recognized a 12-mer peptide corresponding to residues 692 to 703; this epitope maps to a region of domain 4 known to interact with the anthrax toxin receptor CMG-2 and within a conformation-dependent epitope recognized by the well-characterized neutralizing MAb 14B7. As expected, 1-F1 blocked PA''s ability to associate with CMG-2 in an in vitro solid-phase binding assay, and it protected murine macrophage cells from intoxication with LT. 2-B12 recognized a 12-mer peptide corresponding to residues 716 to 727, an epitope located immediately adjacent to the core 14B7 binding site and a stretch of amino acids not previously identified as a target of neutralizing antibodies. 2-B12 was as effective as 1-F1 in neutralizing LT in vitro, although it only partially inhibited PA binding to its receptor. Mice passively administered 1-F1 or 2-B12 were partially protected against a lethal challenge with LT. These results advance our fundamental understanding of the mechanisms by which antibodies neutralize anthrax toxin and may have future application in the evaluation of candidate rPA vaccines.Bacillus anthracis has long been recognized as a serious public health threat, given the ease with which B. anthracis spores can be disseminated via aerosol and due to the high mortality rate that accompanies spore inhalation. These fears were realized in the fall of 2001, when B. anthracis spores were circulated through the U.S. postal system, resulting in five deaths, 22 known cases of infection, and the possible exposure of more than 30,000 people (7). Although routine vaccination of civilians against anthrax is neither necessary nor desirable, certain segments of the population, notably emergency first responders and research laboratory personnel, remain at risk of exposure and are in need of an effective preexposure vaccine. In the United States, the only licensed anthrax vaccine, Anthrax Vaccine Adsorbed (AVA) or Biothrax, has been subject to controversy for years, due to safety concerns (3), and is not considered an ideal vaccine due to its protracted vaccination schedule (six injections over 18 months). AVA, which consists of formalin-treated culture filtrate from an attenuated strain of B. anthracis adsorbed to aluminum hydroxide, is also inherently difficult to manufacture and to standardize. A recombinant anthrax vaccine, manufactured by VaxGen, was considered to be a leading candidate to replace AVA and to supply the Strategic National Stockpile with 75 million doses, but it was recently discontinued because of concerns regarding vaccine production and stability. For these reasons, there is now a concerted effort to develop a new anthrax vaccine based on a defined antigenic composition.Current anthrax vaccines are focused on protective antigen (PA), the primary determinant in AVA that is involved in immunity to anthrax infection (21). PA is the receptor-binding subunit common to both lethal factor (LF), and edema factor (EF), which, upon PA binding form lethal toxin (LT) and edema toxin (ET), respectively. The anthrax toxins are responsible for the high mortality rates associated with inhalational B. anthracis infection. PA is secreted by B. anthracis as an 83-kDa protein (PA83) that binds two known anthrax toxin receptors (ATRs), namely, tumor endothelium marker 8 (TEM-8) and capillary morphogenesis protein 2 (CMG-2) (5, 6, 19). Following receptor engagement, the amino-terminal 20-kDa region of the PA (PA20) is proteolytically cleaved by a furinlike protease, exposing a region of PA that allows for the formation of the holotoxin. The membrane-bound 63-kDa PA (PA63) spontaneously oligomerizes into heptamers and then associates either with LF, a zinc-dependent protease specific for certain members of the mitogen-activated protein kinase kinase family (12, 13), or EF, a calmodulin-, Ca2+-dependent adenylate cyclase (20), to form LT and ET.It is well established that animals immunized with recombinant PA, or passively administered anti-PA antiserum, are protected against LT/ET exposure or spore challenge (reviewed in references 16 and 7). For example, we recently reported that LT-challenged mice treated with goat anti-PA antiserum (either alone or in combination with antibiotics) demonstrated increased survival rates compared to untreated mice (18). However, the antibody response to PA is complicated: total anti-PA titers, as measured by enzyme-linked immunosorbent assays (ELISAs), do not correlate well with protection (35). This is likely due to the fact that a large number of anti-PA antibodies are directed against non-neutralizing (or possibly even toxin-enhancing) epitopes (29, 41). Pioneering work by Little et al. (23) suggests that at least 20 antigenic determinants exist on PA. At present, only a few of these epitopes has been described in detail; several have been shown to be targets of monoclonal antibodies (MAbs) capable of effective neutralization in vivo or in vitro (1, 8, 10, 17, 44, 45). The fact that key neutralizing epitopes on PA remain widely unidentified poses a significant barrier to the evaluation of the efficacy of vaccines based on full-length and truncated PA subunits.PAs can be divided into four functional domains. Domain 1 (residues 1 to 258) contains the furin recognition site (164RKKR167), as well as the region of PA that is recognized by LF and EF (30). Domain 2 (residues 259 to 487) and part of domain 3 (residues 488 to 595) are implicated in heptamerization, pore formation, and translocation of EF/LF across endosomal membranes (4, 27, 28, 34). Domain 4 (residues 596 to 735) constitutes the region of the PA involved in receptor recognition and attachment (39). Antibodies against domain 4 are postulated to be the most effective in neutralizing LT and ET (1), since they are proposed to interfere with PA binding to ATR. However, the only neutralizing epitope that has been characterized in detail is the one recognized by MAb 14B7 (24, 37, 40). This MAb contacts the face of domain 4 that is involved in receptor recognition, and has been proposed to encompass residues 671 to 721. Certainly, additional neutralizing epitopes exist on domain 4 (1, 46). In particular, Abboud and Casadevall (1) suggested a linear epitope, immediately adjacent to or possibly overlapping the 14B7 binding site, as the target of neutralizing antibodies. The characterization of antibodies that are both linear epitope dependent and neutralizing will be useful in the evaluation specific correlates of immunity; such MAbs can be incorporated into functional screening assays of potential vaccine candidates.The goal of our study was to identify additional B-cell epitopes, within domain 4 of PA, that constitute the targets of neutralizing MAbs. Toward this end, we screened a collection of B-cell hybridomas, produced from PA83-immunized mice, for MAbs capable of binding to a domain 4-specific peptide array. We identified two domain 4-reactive MAbs, 1-F1 and 2-B12. 1-F1 recognized a peptide spanning residues 692 to 703; this epitope maps to a region of domain 4 that overlaps with the binding site recognized by the well-characterized neutralizing antibody 14B7 (residues 671 to 721) (23, 40). 2-B12, on the other hand, recognized a peptide spanning residues 716 to 727; this epitope maps adjacent to the core 14B7 binding site and at a location not previously identified as being a target of neutralizing antibodies. Both MAbs neutralized LT in vitro and partially protected mice against LT challenge. These results advance our fundamental understanding of the mechanisms by which antibodies neutralize anthrax toxin and may have future application in the evaluation of candidate rPA vaccines.
Keywords:
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号