37 research outputs found

    Contribution of murine IgG Fc regions to antibody binding to the capsule of Burkholderia pseudomallei.

    Get PDF
    Immunoglobulin G3 (IgG3) is the predominant IgG subclass elicited in response to polysaccharide antigens in mice. This specific subclass has been shown to crosslink its fragment crystallizable (Fc) regions following binding to multivalent polysaccharides. Crosslinking leads to increased affinity through avidity, which theoretically should lead to more effective protection against bacteria and yeast displaying capsular polysaccharides on their surface. To investigate this further we have analyzed the binding characteristics of 2 IgG monoclonal antibody (mAb) subclass families that bind to the capsular polysaccharide (CPS) of Burkholderia pseudomallei. The first subclass family originated from an IgG3 hybridoma cell line (3C5); the second family was generated from an IgG1 cell line (2A5). When the Fc region of the 3C5 IgG3 is removed by proteolytic cleavage, the resulting F(ab')2 fragments exhibit decreased affinity compared to the full-length mAb. Similarly, when the parent IgG3 mAb is subclass-switched to IgG1, IgG2b, and IgG2a, all of these subclasses exhibit decreased affinity. This decrease in affinity is not seen when the 2A5 IgG1 mAb is switched to an IgG2b or IgG2a, strongly suggesting the drop in affinity is related to the IgG3 Fc region

    Polysaccharide Specific Monoclonal Antibodies Provide Passive Protection against Intranasal Challenge with Burkholderia pseudomallei

    Get PDF
    Burkholderia pseudomallei is a Gram-negative bacillus that is the causative agent of melioidosis. The bacterium is inherently resistant to many antibiotics and mortality rates remain high in endemic areas. The lipopolysaccharide (LPS) and capsular polysaccharide (CPS) are two surface-associated antigens that contribute to pathogenesis. We previously developed two monoclonal antibodies (mAbs) specific to the CPS and LPS; the CPS mAb was shown to identify antigen in serum and urine from melioidosis patients. The goal of this study was to determine if passive immunization with CPS and LPS mAbs alone and in combination would protect mice from a lethal challenge with B. pseudomallei. Intranasal (i.n.) challenge experiments were performed with B. pseudomallei strains 1026b and K96423. Both mAbs provided significant protection when administered alone. A combination of mAbs was protective when low doses were administered. In addition, combination therapy provided a significant reduction in spleen colony forming units (cfu) compared to results when either the CPS or LPS mAbs were administered alone

    Protective and Immunochemical Activities of Monoclonal Antibodies Reactive with the Bacillus anthracis Polypeptide Capsule

    No full text
    Bacillus anthracis is surrounded by a polypeptide capsule composed of poly-gamma-d-glutamic acid (γDPGA). In a previous study, we reported that a monoclonal antibody (MAb F26G3) reactive with the capsular polypeptide is protective in a murine model of pulmonary anthrax. The present study examined a library of six MAbs generated from mice immunized with γDPGA. Evaluation of MAb binding to the capsule by a capsular “quellung” type reaction showed a striking diversity in capsular effects. Most MAbs produced a rim type reaction that was characterized by a sharp increase followed directly by a decrease in refractive index at the capsular edge. Some MAbs produced a second capsular reaction well beneath the capsular edge, suggesting complexity in capsular architecture. Binding of MAbs to soluble γDPGA was assessed by a fluorescence perturbation assay in which a change in the MAb intrinsic fluorescence produced by ligand binding was used as a reporter for antigen-antibody interaction. The MAbs differed considerably in the complexity of the binding curves. MAbs producing rim type capsule reactions typically produced the more complex binding isotherms. Finally, the protective activity of the MAbs was compared in a murine model of pulmonary anthrax. One MAb was markedly less protective than the remaining five MAbs. Characteristics of the more protective MAbs included a relatively high affinity, an immunoglobulin G3 isotype, and a complex binding isotherm in the fluorescence perturbation assay. Given the relatively monotonous structure of γDPGA, the results demonstrate a striking diversity in the antigen binding behavior of γDPGA antibodies

    IgG subclass and heavy chain domains contribute to binding and protection by mAbs to the poly Îł-D-glutamic acid capsular antigen of Bacillus anthracis.

    Get PDF
    Bacterial capsules are common targets for antibody-mediated immunity. The capsule of Bacillus anthracis is unusual among capsules because it is composed of a polymer of poly-γ-d-glutamic acid (γdPGA). We previously generated murine IgG3 monoclonal antibodies (mAbs) to γdPGA that were protective in a murine model of pulmonary anthrax. IgG3 antibodies are characteristic of the murine response to polysaccharide antigens. The goal of the present study was to produce subclass switch variants of the γdPGA mAbs (IgG3 → IgG1 → IgG2b → IgG2a) and assess the contribution of subclass to antibody affinity and protection. Subclass switch antibodies had identical variable regions but differed in their heavy chains. The results showed that a switch from the protective IgG3 to IgG1, IgG2b or IgG2a was accompanied by i) a loss of protective activity ii) a change in mAb binding to the capsular matrix, and iii) a loss of affinity. These results identify a role for the heavy chain constant region in mAb binding. Hybrid mAbs were constructed in which the CH1, CH2 or CH3 heavy chain constant domains from a non-protective, low binding IgG2b mAb were swapped into the protective IgG3 mAb. The IgG3 mAb that contained the CH1 domain from IgG2b showed no loss of affinity or protection. In contrast, swapping the CH2 or CH3 domains from IgG2b into IgG3 produced a reduction in affinity and a loss of protection. These studies identify a role for the constant region of IgG heavy chains in affinity and protection against an encapsulated bacterial pathogen

    <i>In vivo</i> Distribution and Clearance of Purified Capsular Polysaccharide from <i>Burkholderia pseudomallei</i> in a Murine Model

    No full text
    <div><p><i>Burkholderia pseudomallei</i> is the causative agent of melioidosis, a severe infection prominent in northern Australia and Southeast Asia. The “gold standard” for melioidosis diagnosis is bacterial isolation, which takes several days to complete. The resulting delay in diagnosis leads to delayed treatments, which could result in death. In an attempt to develop better methods for early diagnosis of melioidosis, <i>B</i>. <i>pseudomallei</i> capsular polysaccharide (CPS) was identified as an important diagnostic biomarker. A rapid lateral flow immunoassay utilizing CPS-specific monoclonal antibody was developed and tested in endemic regions worldwide. However, the <i>in vivo</i> fate and clearance of CPS has never been thoroughly investigated. Here, we injected mice with purified CPS intravenously and determined CPS concentrations in serum, urine, and major organs at various intervals. The results indicate that CPS is predominantly eliminated through urine and no CPS accumulation occurs in the major organs. Immunoblot analysis demonstrated that intact CPS was excreted through urine. To understand how a large molecule like CPS was eliminated without degradation, a 3-dimenational structure of CPS was modeled. The predicted CPS structure has a rod-like shape with a small diameter that could allow it to flow through the glomerulus of the kidney. CPS clearance was determined using exponential decay models and the corrected Akaike Information Criterion. The results show that CPS has a relatively short serum half-life of 2.9 to 4.4 hours. Therefore, the presence of CPS in the serum and/or urine suggests active melioidosis infection and provides a marker to monitor treatment of melioidosis.</p></div

    Contribution of murine IgG Fc regions to antibody binding to the capsule of Burkholderia pseudomallei

    No full text
    Immunoglobulin G3 (IgG3) is the predominant IgG subclass elicited in response to polysaccharide antigens in mice. This specific subclass has been shown to crosslink its fragment crystallizable (Fc) regions following binding to multivalent polysaccharides. Crosslinking leads to increased affinity through avidity, which theoretically should lead to more effective protection against bacteria and yeast displaying capsular polysaccharides on their surface. To investigate this further we have analyzed the binding characteristics of 2 IgG monoclonal antibody (mAb) subclass families that bind to the capsular polysaccharide (CPS) of Burkholderia pseudomallei. The first subclass family originated from an IgG3 hybridoma cell line (3C5)the second family was generated from an IgG1 cell line (2A5). When the Fc region of the 3C5 IgG3 is removed by proteolytic cleavage, the resulting F(ab')(2) fragments exhibit decreased affinity compared to the full-length mAb. Similarly, when the parent IgG3 mAb is subclass-switched to IgG1, IgG2b, and IgG2a, all of these subclasses exhibit decreased affinity. This decrease in affinity is not seen when the 2A5 IgG1 mAb is switched to an IgG2b or IgG2a, strongly suggesting the drop in affinity is related to the IgG3 Fc region

    Kinetics for clearance of CPS from serum.

    No full text
    <p>Mice were intravenously injected with 100 μg, 20 μg, or 4 μg of CPS. Blood samples were collected at the designated time points, and CPS concentrations in serum samples were determined using quantitative sandwich ELISA. The data were best described by a two-parameter monophasic exponential decay model (<i>y = ae</i><sup><i>-bx</i></sup><i>)</i>, where <i>a</i> is the <i>Y</i> intercept and <i>b</i> is the rate constant for clearance. Data shown are mean ± standard deviation for five mice per dose per time point. Half-life (t<sub>1/2</sub>) values calculated from the elimination rate constant (<i>b</i>) derived from the model fitting were similar for all three doses of CPS. The results demonstrate that CPS is eliminated rapidly from serum with a half-life of 4 hours, 4.4 hours, or 2.9 hours for the doses of 100 μg, 20 μg, or 4 μg CPS, respectively.</p

    Organ distribution of CPS.

    No full text
    <p>Mice were intravenously injected with 100 μg CPS per mouse. Internal organs (lungs, liver, spleen, and kidneys) were collected at various time points post-injection. The organs were homogenized in PBS. CPS amount per organ was determined by quantitative sandwich ELISA. The amount of CPS in blood samples was calculated from the CPS concentration in serum as shown in <a href="http://www.plosntds.org/article/info:doi/10.1371/journal.pntd.0005217#pntd.0005217.g001" target="_blank">Fig 1</a>. Data shown are mean ± standard deviation for five mice per time point. The negative values after subtraction of CPS amounts from serum found in each organ were adjusted to zero. The results showed that no significant amount of CPS accumulated in any of the colletced organs.</p

    Excreted CPS detection by AMD<sup>™</sup> LFI.

    No full text
    <p>A urine sample from a CPS-treated mouse was serially diluted in mouse control urine to the indicated CPS concentrations. Each concentration of the urine sample then was tested with AMD<sup>™</sup> LFI. The tests were assessed by four examiners in a randomized, semi-blinded manner (panel A), and by using a lateral flow reader (panel B). The results demonstrated that AMD<sup>™</sup> LFI could detect excreted CPS as low as 0.2 ng/mL.</p
    corecore