Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
NPJ Vaccines ; 9(1): 103, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38858423

ABSTRACT

Acellular multivalent vaccines for pertussis (DTaP and Tdap) prevent symptomatic disease and infant mortality, but immunity to Bordetella pertussis infection wanes significantly over time resulting in cyclic epidemics of pertussis. The messenger RNA (mRNA) vaccine platform provides an opportunity to address complex bacterial infections with an adaptable approach providing Th1-biased responses. In this study, immunogenicity and challenge models were used to evaluate the mRNA platform with multivalent vaccine formulations targeting both B. pertussis antigens and diphtheria and tetanus toxoids. Immunization with mRNA formulations were immunogenetic, induced antigen specific antibodies, as well as Th1 T cell responses. Upon challenge with either historical or contemporary B. pertussis strains, 6 and 10 valent mRNA DTP vaccine provided protection equal to that of 1/20th human doses of either DTaP or whole cell pertussis vaccines. mRNA DTP immunized mice were also protected from pertussis toxin challenge as measured by prevention of lymphocytosis and leukocytosis. Collectively these pre-clinical mouse studies illustrate the potential of the mRNA platform for multivalent bacterial pathogen vaccines.

2.
mBio ; 14(1): e0361122, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36749098

ABSTRACT

Tuberculosis (TB) remains a leading cause of morbidity and mortality worldwide. To date, the mainstay of vaccination involves the use of Mycobacterium bovis bacillus Calmette-Guérin (BCG), a live-attenuated vaccine that confers protection against extrapulmonary disease in infants and children but not against lung disease. Thus, there is an urgent need for novel vaccines. Here, we show that a multicomponent acellular vaccine (TB-MAPS) induces robust antibody responses and long-lived systemic and tissue-resident memory Th1, Th17, and cytotoxic CD4+ and CD8+ T cells, and promotes trained innate immunity mediated by γδT and NKT cells in mice. When tested in a mouse aerosol infection model, TB-MAPS significantly reduced bacterial loads in the lungs and spleens to the same extent as BCG. When used in conjunction with BCG, TB-MAPS further enhanced BCG-mediated protection, especially in the lungs, further supporting this construct as a promising TB vaccine candidate. IMPORTANCE Tuberculosis (TB) remains a leading cause of morbidity and mortality worldwide. Here, we evaluate a novel vaccine which induces a broad immune response to Mycobacterium tuberculosis including robust antibody responses and long-lived systemic and tissue-resident memory Th1, Th17, and cytotoxic CD4+ and CD8+ T cells. When tested in a mouse aerosol infection model, this vaccine significantly reduced bacterial loads in the lungs and spleens to the same extent as BCG. When used in conjunction with BCG, TB-MAPS further enhanced BCG-mediated protection, especially in the lungs, further supporting this construct as a promising TB vaccine candidate.


Subject(s)
Mycobacterium bovis , Mycobacterium tuberculosis , Tuberculosis , Animals , Mice , BCG Vaccine , CD8-Positive T-Lymphocytes , Tuberculosis/prevention & control , Antigens, Bacterial
3.
Mucosal Immunol ; 13(1): 172-182, 2020 01.
Article in English | MEDLINE | ID: mdl-31659300

ABSTRACT

The generation of tissue-resident memory T cells (TRM) is an essential aspect of immunity at mucosal surfaces, and it has been suggested that preferential generation of TRM is one of the principal advantages of mucosally administered vaccines. We have previously shown that antigen-specific, IL-17-producing CD4+ T cells can provide capsular antibody-independent protection against nasal carriage of Streptococcus pneumoniae; but whether pneumococcus-responsive TRM are localized within the nasal mucosa and are sufficient for protection from carriage has not been determined. Here, we show that intranasal administration of live or killed pneumococci to mice generates pneumococcus-responsive IL-17A-producing CD4+ mucosal TRM. Furthermore, we show that these cells are sufficient to mediate long-lived, neutrophil-dependent protection against subsequent pneumococcal nasal challenge. Unexpectedly, and in contrast with the prevailing paradigm, we found that parenteral administration of killed pneumococci also generates protective IL-17A+CD4+ TRM in the nasal mucosa. These results demonstrate a critical and sufficient role of TRM in prevention of pneumococcal colonization, and further that these cells can be generated by parenteral immunization. Our findings therefore have important implications regarding the generation of immune protection at mucosal surfaces by vaccination.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Nose/immunology , Pneumococcal Infections/immunology , Streptococcal Vaccines/immunology , Streptococcus pneumoniae/physiology , Animals , Cells, Cultured , Disease Resistance , Humans , Immunologic Memory , Interleukin-17/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Vaccination
4.
Sci Rep ; 6: 22721, 2016 Mar 07.
Article in English | MEDLINE | ID: mdl-26949061

ABSTRACT

Ricin is a member of the A-B family of bacterial and plant toxins that exploit retrograde trafficking to the Golgi apparatus and endoplasmic reticulum (ER) as a means to deliver their cytotoxic enzymatic subunits into the cytoplasm of mammalian cells. In this study we demonstrate that R70 and SyH7, two well-characterized monoclonal antibodies (mAbs) directed against distinct epitopes on the surface of ricin's enzymatic subunit (RTA), interfere with toxin transport from the plasma membrane to the trans Golgi network. Toxin-mAb complexes formed on the cell surface delayed ricin's egress from EEA-1(+) and Rab7(+) vesicles and enhanced toxin accumulation in LAMP-1(+) vesicles, suggesting the complexes were destined for degradation in lysosomes. Three other RTA-specific neutralizing mAbs against different epitopes were similar to R70 and SyH7 in terms of their effects on ricin retrograde transport. We conclude that interference with toxin retrograde transport may be a hallmark of toxin-neutralizing antibodies directed against disparate epitopes on RTA.


Subject(s)
Antibodies, Monoclonal/metabolism , Antitoxins/metabolism , Chemical Warfare Agents/metabolism , Immunologic Factors/metabolism , Ricin/metabolism , Animals , Antibodies, Monoclonal/immunology , Antitoxins/immunology , Chlorocebus aethiops , Epitopes/immunology , HeLa Cells , Humans , Immunologic Factors/immunology , Protein Binding , Protein Transport , Ricin/immunology , Vero Cells
5.
Immunol Lett ; 158(1-2): 7-13, 2014.
Article in English | MEDLINE | ID: mdl-24269767

ABSTRACT

Efforts to develop a vaccine for ricin toxin are focused on identifying highly immunogenic, safe, and thermostable recombinant derivatives of ricin's enzymatic A subunit (RTA). As a means to guide vaccine design, we have embarked on an effort to generate a comprehensive neutralizing and non-neutralizing B cell epitope map of RTA. In a series of previous studies, we identified three spatially distinct linear (continuous), neutralizing epitopes on RTA, as defined by monoclonal antibodies (mAbs) PB10 (and R70), SyH7, and GD12. In this report we now describe a new collection of 19 toxin-neutralizing mAbs that bind non-linear epitopes on RTA. The most potent toxin-neutralizing mAbs in this new collection, namely WECB2, TB12, PA1, PH12 and IB2 each had nanamolar (or sub-nanomolar) affinities for ricin and were each capable of passively protecting mice against a 5-10xLD50 toxin challenge. Competitive binding assays by surface plasmon resonance revealed that WECB2 binds an epitope that overlaps with PB10 and R70; TB12, PA1, PH12 recognize epitope(s) close to or overlapping with SyH7's epitope; and GD12 and IB2 recognize epitopes that are spatially distinct from all other toxin-neutralizing mAbs. We estimate that we have now accounted for ∼75% of the predicted epitopes on the surface of RTA and that toxin-neutralizing mAbs are directed against a very limited number of these epitopes. Having this information provides a framework for further refinement of RTA mutagenesis and vaccine design.


Subject(s)
Antibodies, Blocking/immunology , B-Lymphocytes/immunology , Epitopes, B-Lymphocyte/metabolism , Hemorrhage/prevention & control , Ricin/metabolism , Animals , Catalytic Domain/immunology , Epitope Mapping , Epitopes, B-Lymphocyte/chemistry , Epitopes, B-Lymphocyte/immunology , Female , Hemorrhage/chemically induced , Hemorrhage/immunology , Humans , Hybridomas , Mice , Mice, Inbred BALB C , Molecular Structure , Ricin/chemistry , Ricin/immunology , Vaccines
6.
PLoS One ; 8(8): e69678, 2013.
Article in English | MEDLINE | ID: mdl-23936344

ABSTRACT

Currently, there is a shortage of adjuvants that can be employed with protein subunit vaccines to enhance protection against biological threats. LT-IIb(T13I) is an engineered nontoxic derivative of LT-IIb, a member of the type II subfamily of heat labile enterotoxins expressed by Escherichia coli, that possesses potent mucosal adjuvant properties. In this study we evaluated the capacity of LT-IIb(T13I) to augment the potency of RiVax, a recombinant ricin toxin A subunit vaccine, when co-administered to mice via the intradermal (i.d.) and intranasal (i.n.) routes. We report that co-administration of RiVax with LT-IIb(T13I) by the i.d. route enhanced the levels of RiVax-specific serum IgG antibodies (Ab) and elevated the ratio of ricin-neutralizing to non-neutralizing Ab, as compared to RiVax alone. Protection against a lethal ricin challenge was also augmented by LT-IIb(T13I). While local inflammatory responses elicited by LT-IIb(T13I) were comparable to those elicited by aluminum salts (Imject®), LT-IIb(T13I) was more effective than aluminum salts at augmenting production of RiVax-specific serum IgG. Finally, i.n. administration of RiVax with LT-IIb(T13I) also increased levels of RiVax-specific serum and mucosal Ab and enhanced protection against ricin challenge. Collectively, these data highlight the potential of LT-IIb(T13I) as an effective next-generation i.d., or possibly i.n. adjuvant for enhancing the immunogenicity of subunit vaccines for biodefense.


Subject(s)
Antibodies, Neutralizing/immunology , Bacterial Toxins/administration & dosage , Enterotoxins/administration & dosage , Escherichia coli Proteins/administration & dosage , Inflammation/prevention & control , Skin/immunology , Vaccines, Subunit/therapeutic use , Vaccines, Synthetic/therapeutic use , Vaccines/administration & dosage , Adjuvants, Immunologic , Administration, Intranasal , Animals , Antibodies, Neutralizing/therapeutic use , Bacterial Toxins/immunology , Bacterial Toxins/metabolism , Drug Synergism , Enterotoxins/immunology , Enterotoxins/metabolism , Escherichia coli Proteins/immunology , Escherichia coli Proteins/metabolism , Female , Immunity, Mucosal , Immunization , Inflammation/immunology , Inflammation/metabolism , Mice , Mice, Inbred BALB C , Skin/metabolism , Vaccines/immunology , Vaccines/metabolism
7.
J Immunol Methods ; 395(1-2): 71-8, 2013 Sep 30.
Article in English | MEDLINE | ID: mdl-23774033

ABSTRACT

The penultimate event in the intoxication of mammalian cells by ricin toxin is the reduction, in the endoplasmic reticulum (ER), of the intermolecular disulfide bond that links ricin's enzymatic (RTA) and binding (RTB) subunits. In this report we adapted an in vitro protein disulfide isomerase (PDI)-mediated reduction assay to test the hypothesis that the RTA-specific neutralizing monoclonal antibody (mAb) IB2 interferes with the liberation of RTA from RTB. IB2 recognizes an epitope located near the interface between RTA and RTB and, like a number of other RTA-specific neutralizing mAbs, is proposed to neutralize ricin intracellularly. In this study, we found that IB2 virtually eliminated the reduction of ricin holotoxin into RTA and RTB in vitro. Surprisingly, three other neutralizing mAbs (GD12, R70 and SyH7) that bind epitopes at considerable distance from ricin's disulfide bond were as effective (or nearly as effective) as IB2 in interfering with PDI-mediated liberation of RTA from RTB. By contrast, two non-neutralizing RTA-specific mAbs, FGA12 and SB1, did not affect PDI-mediated reduction of ricin. These data reveal a possible mechanism by which RTA-specific antibodies may neutralize ricin intracellularly, provided they are capable of trafficking in association with ricin from the cell surface to the ER.


Subject(s)
Antibodies, Monoclonal/metabolism , Antibodies, Neutralizing/metabolism , Ricin/antagonists & inhibitors , Ricin/immunology , Animals , Antibody Specificity , Cell Line , Chlorocebus aethiops , Humans , Immunoglobulin G/metabolism , Mice , Microscopy, Confocal , Models, Molecular , Oxidation-Reduction , Protein Disulfide-Isomerases/metabolism , Protein Subunits/antagonists & inhibitors , Protein Subunits/chemistry , Protein Subunits/immunology , Ricin/chemistry , Toxins, Biological/antagonists & inhibitors , Toxins, Biological/chemistry , Toxins, Biological/immunology , Vero Cells
8.
Hum Vaccin Immunother ; 9(4): 744-52, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23563512

ABSTRACT

There is great interest in the design and development of highly thermostable and immunogenic protein subunit vaccines for biodefense. In this study, we used two orthogonal and complementary computational protein design approaches to generate a series of single-point mutants of RiVax, an attenuated recombinant ricin A chain (RTA) protein subunit vaccine antigen. As assessed by differential scanning calorimetry, the conformational stabilities of the designed mutants ranged from 4°C less stable to 4.5°C more stable than RiVax, depending on solution pH. Two more thermostable (V18P, C171L) and two less thermostable (T13V, S89T) mutants that displayed native-like secondary and tertiary structures (as determined by circular dichroism and fluorescence spectral analysis, respectively) were tested for their capacity to elicit RTA-specific antibodies and toxin-neutralizing activity. Following a prime-boost regimen, we found qualitative differences with respect to specific antibody titers and toxin neutralizing antibody levels induced by the different mutants. Upon a second boost with the more thermostable mutant C171L, a statistically significant increase in RTA-specific antibody titers was observed when compared with RiVax-immunized mice. Notably, the results indicate that single residue changes can be made to the RiVax antigen that increase its thermal stability without adversely impacting the efficacy of the vaccine.


Subject(s)
Point Mutation , Ricin/administration & dosage , Ricin/immunology , Animals , Antibodies, Neutralizing/blood , Antitoxins/blood , Blood Glucose/analysis , Calorimetry , Circular Dichroism , Disease Models, Animal , Drug Stability , Mice , Mice, Inbred BALB C , Mutant Proteins/administration & dosage , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/immunology , Poisoning/prevention & control , Protein Conformation , Protein Stability , Ricin/chemistry , Ricin/genetics , Ricin/toxicity , Spectrometry, Fluorescence , Survival Analysis , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/chemistry , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/chemistry , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
9.
Eur J Pharm Biopharm ; 85(2): 279-86, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23583494

ABSTRACT

Lyophilization was used to prepare dry, glassy solid vaccine formulations of recombinant ricin toxin A-chain containing suspensions of colloidal aluminum hydroxide adjuvant. Four lyophilized formulations were prepared by using combinations of rapid or slow cooling during lyophilization and one of two buffers, histidine or ammonium acetate. Trehalose was used as the stabilizing excipient. Aggregation of the colloidal aluminum hydroxide suspension was reduced in formulations processed with a rapid cooling rate. Aluminum hydroxide particle size distributions, glass transition temperatures, water contents, and immunogenicities of lyophilized vaccines were independent of incubation time at 40 °C for up to 15 weeks. Mice immunized with reconstituted ricin toxin subunit A (RTA) vaccines produced RTA-specific antibodies and toxin-neutralizing antibodies (TNAs) regardless of the length of high temperature vaccine storage or the degree of aluminum adjuvant aggregation that occurred during lyophilization. In murine studies, lyophilized formulations of vaccines conferred protection against exposure to lethal doses of ricin, even after the lyophilized formulations had been stored at 40 °C for 4 weeks. A corresponding liquid formulation of vaccine stored at 40 °C elicited RTA-specific antibody titers but failed to confer immunity during a ricin challenge.


Subject(s)
Drug Stability , Recombinant Proteins/chemistry , Ricin/chemistry , Vaccines, Subunit/chemistry , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacology , Adjuvants, Pharmaceutic/chemistry , Adjuvants, Pharmaceutic/pharmacology , Aluminum Hydroxide/chemistry , Animals , Antibodies, Neutralizing/immunology , Antibody Formation/immunology , Buffers , Chemistry, Pharmaceutical/methods , Drug Storage , Excipients/chemistry , Female , Freeze Drying/methods , Hot Temperature , Mice , Particle Size , Recombinant Proteins/immunology , Ricin/immunology , Transition Temperature , Trehalose/chemistry , Vaccines, Subunit/immunology , Water/chemistry
10.
Clin Vaccine Immunol ; 20(6): 789-94, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23515013

ABSTRACT

The two leading ricin toxin vaccine candidates, RVEc and RiVax, are recombinant derivatives of the toxin's 267-amino-acid enzymatic A chain (RTA). RVEc is truncated at the C terminus (residues 199 to 267) to improve protein thermostability, while RiVax has two point mutations (V76M and Y80A) that eliminate the RNA N-glycosidase activity of RTA, as well as its ability to induce vascular leak syndrome. The two vaccines have never been directly compared in terms of their ability to stimulate RTA-specific antibodies (Abs), toxin-neutralizing activity (TNA), or protective immunity. To address this issue, groups of female BALB/c mice were immunized two or three times with Alhydrogel-adsorbed RiVax or RVEc at a range of doses (0.3 to 20 µg) and then challenged with 10 50% lethal doses (LD(50)s) of ricin. We found that the vaccines were equally effective at eliciting protective immunity at the doses tested. There were, however, quantitative differences in the antibody responses. RVEc tended to elicit higher levels of ricin-specific RTA IgG and TNA than did RiVax. Pepscan analysis revealed that serum Abs elicited by RVEc were skewed toward a solvent-exposed immunodominant α-helix known to be the target of potent toxin-neutralizing Abs. Finally, immunodepletion experiments suggest that the majority of toxin-neutralizing Abs elicited by RiVax were confined to residues 1 to 198, possibly explaining the equal effectiveness of RVEc as a vaccine.


Subject(s)
Antibodies, Neutralizing/blood , Antitoxins/blood , Poisoning/prevention & control , Ricin/antagonists & inhibitors , Ricin/immunology , Vaccines, Subunit/immunology , Adjuvants, Immunologic/administration & dosage , Aluminum Hydroxide/administration & dosage , Animals , Epitope Mapping , Female , Mice , Mice, Inbred BALB C , Ricin/toxicity , Survival Analysis , Vaccines, Subunit/administration & dosage
11.
Curr Top Microbiol Immunol ; 357: 209-41, 2012.
Article in English | MEDLINE | ID: mdl-22113742

ABSTRACT

Ricin toxin is an extraordinarily potent inducer of cell death and inflammation. Ricin is also a potent provocateur of the humoral immune system, eliciting a mixture of neutralizing, non-neutralizing and even toxin-enhancing antibodies. The characterization of dozens of monoclonal antibodies (mAbs) against the toxin's enzymatic (RTA) and binding (RTB) subunits has begun to reveal fundamental insights into the underlying mechanisms by which antibodies neutralize (or fail to neutralize) ricin in systemic and mucosal compartments. This information has had immediate applications in the design, development and evaluation of ricin subunit vaccines and immunotherapeutics.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Ricin/immunology , Ricin/metabolism , Animals , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/metabolism , Epitopes, B-Lymphocyte/immunology , Humans , Immunity, Mucosal , Protein Binding/immunology , Ricin/chemistry
12.
Vaccine ; 30(7): 1239-43, 2012 Feb 08.
Article in English | MEDLINE | ID: mdl-22197964

ABSTRACT

GD12 is a murine monoclonal IgG(1) (mAb) that recognizes an immunodominant linear neutralizing epitope (163-TLARSFIICIQM-174) on the A subunit (RTA) of ricin toxin. With the long-term goal of using GD12 as a potential countermeasure against ricin intoxication, we have produced a chimeric derivative of GD12 (cGD12) in which the murine heavy and light chain variable regions were fused to a human IgG(1) framework. The chimeric mAb, expressed and purified using a Nicotiana-based system demonstrated epitope specificity and ricin neutralizing activity similar to the parental murine mAb. Passive administration of cGD12 (10µg) to mice by intraperitoneal injection protected the animals against a systemic ricin challenge. In a post-exposure setting, the murine and chimeric mAbs administered as much as 6h after toxin challenge were each capable of rescuing mice from toxin-induced death, revealing the potential of GD12 to serve as both a prophylactic and therapeutic for ricin intoxication.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Neutralizing/administration & dosage , Immunoglobulin G/immunology , Nicotiana/metabolism , Ricin/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal, Humanized/biosynthesis , Antibodies, Monoclonal, Humanized/genetics , Antibodies, Neutralizing/biosynthesis , Antibodies, Neutralizing/genetics , Epitopes , Female , Gene Expression , Humans , Immunization, Passive , Immunodominant Epitopes , Immunoglobulin G/chemistry , Injections, Intraperitoneal , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Poisoning/prevention & control , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Ricin/toxicity , Nicotiana/genetics
13.
Toxins (Basel) ; 3(9): 1131-45, 2011 09.
Article in English | MEDLINE | ID: mdl-22069759

ABSTRACT

The entry of ricin toxin into macrophages and certain other cell types in the spleen and liver results in toxin-induced inflammation, tissue damage and organ failure. It has been proposed that uptake of ricin into macrophages is facilitated by the mannose receptor (MR; CD206), a C-type lectin known to recognize the oligosaccharide side chains on ricin's A (RTA) and B (RTB) subunits. In this study, we confirmed that the MR does indeed promote ricin binding, uptake and killing of monocytes in vitro. To assess the role of MR in the pathogenesis of ricin in vivo, MR knockout (MR(-/-)) mice were challenged with the equivalent of 2.5× or 5× LD(50) of ricin by intraperitoneal injection. We found that MR(-/-) mice were significantly more susceptible to toxin-induced death than their age-matched, wild-type control counterparts. These data are consistent with a role for the MR in scavenging and degradation of ricin, not facilitating its uptake and toxicity in vivo.


Subject(s)
Immunity, Innate/drug effects , Lectins, C-Type/physiology , Mannose-Binding Lectins/physiology , Receptors, Cell Surface/physiology , Ricin/toxicity , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cytokines/immunology , Female , Humans , Leukocytes/drug effects , Macrophages/drug effects , Male , Mannose Receptor , Mice , Mice, Knockout , Ricin/blood , Ricin/pharmacokinetics
14.
Vaccine ; 28(43): 7035-46, 2010 Oct 08.
Article in English | MEDLINE | ID: mdl-20727394

ABSTRACT

Efforts to develop an effective vaccine against ricin are focused on the engineering of attenuated and stable recombinant forms of the toxin's enzymatic A subunit (RTA). While several candidate antigens are in development, vaccine design and efficacy studies are being undertaken in the absence of a fundamental understanding of those regions of RTA that are critical in eliciting protective immunity. In this present study, we produced and characterized a collection of monoclonal antibodies (MAbs) directed against five distinct immunodominant regions on RTA, and used these MAbs to identify several key neutralizing epitopes on the toxin. Protective MAbs were directed against α-helices located in RTA folding domains 1 and 2, whereas non-neutralizing antibodies recognized random coils and loops that were primarily confined to folding domain 3. These data offer insights into the immunodominant and structural determinants on RTA that give rise to protective immunity, and for the first time provide an immunological rationale for ricin vaccine design.


Subject(s)
Antibodies, Monoclonal/immunology , Ricin/chemistry , Ricin/immunology , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Neutralizing/immunology , Antibody Affinity , Chlorocebus aethiops , Epitope Mapping , Epitopes, B-Lymphocyte/immunology , Female , Immunodominant Epitopes/immunology , Mice , Mice, Inbred BALB C , Models, Molecular , Protein Folding , Protein Structure, Secondary , Rabbits , Vero Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...