Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 559
Filter
1.
Genes (Basel) ; 15(4)2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38674361

ABSTRACT

Bacillus anthracis is the bacterium responsible for causing the zoonotic disease called anthrax. The disease presents itself in different forms like gastrointestinal, inhalation, and cutaneous. Bacterial spores are tremendously adaptable, can persist for extended periods and occasionally endanger human health. The Anthrax Toxin Receptor-2 (ANTXR2) gene acts as membrane receptor and facilitates the entry of the anthrax toxin into host cells. Additionally, mutations in the ANTXR2 gene have been linked to various autoimmune diseases, including Hyaline Fibromatosis Syndrome (HFS), Ankylosing Spondylitis (AS), Juvenile Hyaline Fibromatosis (JHF), and Infantile Systemic Hyalinosis (ISH). This study delves into the genetic landscape of ANTXR2, aiming to comprehend its associations with diverse disorders, elucidate the impacts of its mutations, and pinpoint minimal non-pathogenic mutations capable of reducing the binding affinity of the ANTXR2 gene with the protective antigen. Recognizing the pivotal role of single-nucleotide polymorphisms (SNPs) in shaping genetic diversity, we conducted computational analyses to discern highly deleterious and tolerated non-synonymous SNPs (nsSNPs) in the ANTXR2 gene. The Mutpred2 server determined that the Arg465Trp alteration in the ANTXR2 gene leads to altered DNA binding (p = 0.22) with a probability of a deleterious mutation of 0.808; notably, among the identified deleterious SNPs, rs368288611 (Arg465Trp) stands out due to its significant impact on altering the DNA-binding ability of ANTXR2. We propose these SNPs as potential candidates for hypertension linked to the ANTXR2 gene, which is implicated in blood pressure regulation. Noteworthy among the tolerated substitutions is rs200536829 (Ala33Ser), recognized as less pathogenic; this highlights its potential as a valuable biomarker, potentially reducing side effects on the host while also reducing binding with the protective antigen protein. Investigating these SNPs holds the potential to correlate with several autoimmune disorders and mitigate the impact of anthrax disease in humans.


Subject(s)
Anthrax , Antigens, Bacterial , Mutation , Polymorphism, Single Nucleotide , Receptors, Peptide , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Humans , Anthrax/microbiology , Anthrax/genetics , Anthrax/immunology , Receptors, Peptide/genetics , Bacterial Toxins/genetics , Bacillus anthracis/genetics , Bacillus anthracis/pathogenicity , Hyaline Fibromatosis Syndrome/genetics , Hyaline Fibromatosis Syndrome/microbiology , Spondylitis, Ankylosing/genetics , Spondylitis, Ankylosing/immunology , Spondylitis, Ankylosing/microbiology , Disease Resistance/genetics , Receptors, Cell Surface/genetics , Protein Binding
2.
PLoS One ; 16(12): e0260202, 2021.
Article in English | MEDLINE | ID: mdl-34928976

ABSTRACT

Live anthrax vaccine containing spores from attenuated strains STI-1 of Bacillus anthracis is used in Russia and former CIS (Commonwealth of Independent States) to prevent anthrax. In this paper we studied the duration of circulation of antibodies specific to spore antigens, the protective antigen (PA), the lethal factor (LF) and their domains (D) in donors' blood at different times after their immunization with live anthrax vaccine. The relationship between the toxin neutralization activity level and the level of antibodies to PA, LF and their domains was tested. The effect of age, gender and number of vaccinations on the level of adaptive post-vaccination immune response has been studied. It was shown that antibodies against PA-D1 circulate in the blood of donors for 1 year or more after immunization with live anthrax vaccine. Antibodies against all domains of LF and PA-D4 were detected in 11 months after vaccination. Antibodies against the spores were detected in 8 months after vaccination. A moderate positive correlation was found between the titers of antibodies to PA, LF, or their domains, and the TNA of the samples of blood serum from the donors.


Subject(s)
Adaptive Immunity , Anthrax Vaccines/immunology , Anthrax/immunology , Anthrax/prevention & control , Anthrax Vaccines/administration & dosage , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Bacterial Toxins/immunology , Blood Donors , Humans , Neutralization Tests , Russia , Spores, Bacterial/immunology , Vaccination
3.
PLoS One ; 16(10): e0258317, 2021.
Article in English | MEDLINE | ID: mdl-34634075

ABSTRACT

Anthrax is a zoonotic disease caused by the gram-positive spore-forming bacterium Bacillus anthracis. Detecting naturally acquired antibodies against anthrax sublethal exposure in animals is essential for anthrax surveillance and effective control measures. Serological assays based on protective antigen (PA) of B. anthracis are mainly used for anthrax surveillance and vaccine evaluation. Although the assay is reliable, it is challenging to distinguish the naturally acquired antibodies from vaccine-induced immunity in animals because PA is cross-reactive to both antibodies. Although additional data on the vaccination history of animals could bypass this problem, such data are not readily accessible in many cases. In this study, we established a new enzyme-linked immunosorbent assay (ELISA) specific to antibodies against capsule biosynthesis protein CapA antigen of B. anthracis, which is non-cross-reactive to vaccine-induced antibodies in horses. Using in silico analyses, we screened coding sequences encoded on pXO2 plasmid, which is absent in the veterinary vaccine strain Sterne 34F2 but present in virulent strains of B. anthracis. Among the 8 selected antigen candidates, capsule biosynthesis protein CapA (GBAA_RS28240) and peptide ABC transporter substrate-binding protein (GBAA_RS28340) were detected by antibodies in infected horse sera. Of these, CapA has not yet been identified as immunoreactive in other studies to the best of our knowledge. Considering the protein solubility and specificity of B. anthracis, we prepared the C-terminus region of CapA, named CapA322, and developed CapA322-ELISA based on a horse model. Comparative analysis of the CapA322-ELISA and PAD1-ELISA (ELISA uses domain one of the PA) showed that CapA322-ELISA could detect anti-CapA antibodies in sera from infected horses but was non-reactive to sera from vaccinated horses. The CapA322-ELISA could contribute to the anthrax surveillance in endemic areas, and two immunoreactive proteins identified in this study could be additives to the improvement of current or future vaccine development.


Subject(s)
Anthrax/immunology , Antibodies, Bacterial/immunology , Bacillus anthracis/immunology , Bacterial Capsules/immunology , Bacterial Proteins/immunology , Enzyme-Linked Immunosorbent Assay/methods , Heat-Shock Proteins/immunology , Animals , Anthrax Vaccines/immunology , Antigens, Bacterial/immunology , Bacterial Proteins/isolation & purification , Heat-Shock Proteins/isolation & purification , Horses , Immunoglobulin G/immunology , Plasmids/metabolism , Sequence Homology, Amino Acid , Spores, Bacterial/immunology
4.
Front Immunol ; 12: 688257, 2021.
Article in English | MEDLINE | ID: mdl-34497601

ABSTRACT

We present a stochastic mathematical model of the intracellular infection dynamics of Bacillus anthracis in macrophages. Following inhalation of B. anthracis spores, these are ingested by alveolar phagocytes. Ingested spores then begin to germinate and divide intracellularly. This can lead to the eventual death of the host cell and the extracellular release of bacterial progeny. Some macrophages successfully eliminate the intracellular bacteria and will recover. Here, a stochastic birth-and-death process with catastrophe is proposed, which includes the mechanism of spore germination and maturation of B. anthracis. The resulting model is used to explore the potential for heterogeneity in the spore germination rate, with the consideration of two extreme cases for the rate distribution: continuous Gaussian and discrete Bernoulli. We make use of approximate Bayesian computation to calibrate our model using experimental measurements from in vitro infection of murine peritoneal macrophages with spores of the Sterne 34F2 strain of B. anthracis. The calibrated stochastic model allows us to compute the probability of rupture, mean time to rupture, and rupture size distribution, of a macrophage that has been infected with one spore. We also obtain the mean spore and bacterial loads over time for a population of cells, each assumed to be initially infected with a single spore. Our results support the existence of significant heterogeneity in the germination rate, with a subset of spores expected to germinate much later than the majority. Furthermore, in agreement with experimental evidence, our results suggest that most of the spores taken up by macrophages are likely to be eliminated by the host cell, but a few germinated spores may survive phagocytosis and lead to the death of the infected cell. Finally, we discuss how this stochastic modelling approach, together with dose-response data, allows us to quantify and predict individual infection risk following exposure.


Subject(s)
Anthrax/microbiology , Bacillus anthracis/pathogenicity , Macrophages, Peritoneal/microbiology , Models, Biological , Spores, Bacterial/pathogenicity , Animals , Anthrax/immunology , Anthrax/pathology , Bacillus anthracis/growth & development , Bacillus anthracis/immunology , Bayes Theorem , Cell Death , Computer Simulation , Disease Models, Animal , Host-Pathogen Interactions , Inhalation Exposure , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/pathology , Mice , Microbial Viability , Phagocytosis , Population Density , Spores, Bacterial/growth & development , Spores, Bacterial/immunology , Stochastic Processes , Time Factors
5.
Acta Crystallogr D Struct Biol ; 77(Pt 9): 1197-1205, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-34473089

ABSTRACT

Anthrax infection is associated with severe illness and high mortality. Protective antigen (PA) is the central component of the anthrax toxin, which is one of two major virulence factors of Bacillus anthracis, the causative agent of anthrax disease. Upon endocytosis, PA opens a pore in the membranes of endosomes, through which the cytotoxic enzymes of the toxin are extruded. The PA pore is formed by a cooperative conformational change in which the membrane-penetrating loops of PA associate, forming a hydrophobic rim that pierces the membrane. Due to its crucial role in anthrax progression, PA is an important target for monoclonal antibody-based therapy. cAb29 is a highly effective neutralizing antibody against PA. Here, the cryo-EM structure of PA in complex with the Fab portion of cAb29 was determined. It was found that cAb29 neutralizes the toxin by clamping the membrane-penetrating loop of PA to the static surface-exposed loop of the D3 domain of the same subunit, thereby preventing pore formation. These results provide the structural basis for the antibody-based neutralization of PA and bring into focus the membrane-penetrating loop of PA as a target for the development of better anti-anthrax vaccines.


Subject(s)
Anthrax , Antibodies, Bacterial/immunology , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antigens, Bacterial/immunology , Bacillus anthracis/immunology , Bacterial Toxins/immunology , Animals , Anthrax/immunology , Anthrax/microbiology , Cryoelectron Microscopy/methods , Humans , Models, Molecular , Protein Binding
6.
Transbound Emerg Dis ; 68(3): 1676-1684, 2021 May.
Article in English | MEDLINE | ID: mdl-32964687

ABSTRACT

Numerous unknown factors influence anthrax epidemiology in multi-host systems, especially at wildlife/livestock/human interfaces. Serology tests for anti-anthrax antibodies in carnivores are useful tools in identifying the presence or absence of Bacillus anthracis in a range. These were employed to ascertain whether the disease pattern followed the recognized high- and low-risk anthrax zonation in Zimbabwe and also to establish whether anthrax was absent from Hwange National Park in which there have been no reported outbreaks. African lions (Panthera leo) (n = 114) drawn from free-range protected areas and captive game parks located in recognized high- and low-risk zones across Zimbabwe were tested for antibodies to anthrax PA antigen using the ELISA immunoassay. A random selection of 27 lion sera samples comprising 17 seropositive and 10 seronegative sera was further tested in the species-independent toxin neutralization assay (TNA) in order to validate the former as a surveillance tool for anthrax in African lions. Using the ELISA-PA immunoassay, 21.9% (25/114) of the lions tested positive for antibodies to anthrax. Seropositivity was recorded in all study areas, and there was no significant difference (p = .852) in seropositivity between lions in high- and low-risk anthrax zones. Also, there was no significant difference (McNemar's chi-square test = 0.9, p = .343) in the proportion of lions testing positive to anti-PA anthrax antibodies on ELISA-PA immunoassay compared with the TNA, with fair agreement between the two tests [kappa (K) statistic = 0.30; 0.08 < K<0.613]. Results of this study indicate that anthrax could be more widespread than 42 currently realized in Zimbabwe, and present in recognized high- and low-risk zones, including 43 where it has not been reported in over 20 years such as Hwange National Park. This is also the 44 first report documenting the presence of anthrax lethal toxin-neutralizing antibodies in naturally 45 infected carnivores, further confirming exposure to B. anthracis. The research results point to a 46 need for revisiting the currently recognized anthrax risk zones in Zimbabwe. This should be based 47 on improved surveillance of the disease in both wild and domestic animals for better understanding and control of the disease.


Subject(s)
Anthrax/veterinary , Antibodies, Bacterial/blood , Bacillus anthracis/isolation & purification , Lions , Animals , Animals, Wild , Animals, Zoo , Anthrax/epidemiology , Anthrax/immunology , Antibodies, Neutralizing/blood , Enzyme-Linked Immunosorbent Assay/veterinary , Prevalence , Seroepidemiologic Studies , Zimbabwe/epidemiology
7.
PLoS Biol ; 18(12): e3001052, 2020 12.
Article in English | MEDLINE | ID: mdl-33370274

ABSTRACT

Bacillus anthracis, a spore-forming gram-positive bacterium, causes anthrax. The external surface of the exosporium is coated with glycosylated proteins. The sugar additions are capped with the unique monosaccharide anthrose. The West African Group (WAG) B. anthracis have mutations rendering them anthrose deficient. Through genome sequencing, we identified 2 different large chromosomal deletions within the anthrose biosynthetic operon of B. anthracis strains from Chile and Poland. In silico analysis identified an anthrose-deficient strain in the anthrax outbreak among European heroin users. Anthrose-deficient strains are no longer restricted to West Africa so the role of anthrose in physiology and pathogenesis was investigated in B. anthracis Sterne. Loss of anthrose delayed spore germination and enhanced sporulation. Spores without anthrose were phagocytized at higher rates than spores with anthrose, indicating that anthrose may serve an antiphagocytic function on the spore surface. The anthrose mutant had half the LD50 and decreased time to death (TTD) of wild type and complement B. anthracis Sterne in the A/J mouse model. Following infection, anthrose mutant bacteria were more abundant in the spleen, indicating enhanced dissemination of Sterne anthrose mutant. At low sample sizes in the A/J mouse model, the mortality of ΔantC-infected mice challenged by intranasal or subcutaneous routes was 20% greater than wild type. Competitive index (CI) studies indicated that spores without anthrose disseminated to organs more extensively than a complemented mutant. Death process modeling using mouse mortality dynamics suggested that larger sample sizes would lead to significantly higher deaths in anthrose-negative infected animals. The model was tested by infecting Galleria mellonella with spores and confirmed the anthrose mutant was significantly more lethal. Vaccination studies in the A/J mouse model showed that the human vaccine protected against high-dose challenges of the nonencapsulated Sterne-based anthrose mutant. This work begins to identify the physiologic and pathogenic consequences of convergent anthrose mutations in B. anthracis.


Subject(s)
Amino Sugars/genetics , Bacillus anthracis/genetics , Bacillus anthracis/metabolism , Deoxyglucose/analogs & derivatives , Amino Sugars/immunology , Amino Sugars/metabolism , Animals , Anthrax/genetics , Anthrax/immunology , Anthrax/metabolism , Bacillus anthracis/pathogenicity , Biological Evolution , Deoxyglucose/genetics , Deoxyglucose/immunology , Deoxyglucose/metabolism , Disease Models, Animal , Disease Outbreaks , Evolution, Molecular , Female , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred A , Moths/microbiology , Oligosaccharides/genetics , Oligosaccharides/immunology , Oligosaccharides/metabolism , Spores, Bacterial/genetics , Spores, Bacterial/immunology , Spores, Bacterial/metabolism
8.
Expert Opin Biol Ther ; 20(12): 1405-1425, 2020 12.
Article in English | MEDLINE | ID: mdl-32729741

ABSTRACT

INTRODUCTION: Vaccines and therapeutic antibodies are the most crucial components of anthrax prophylaxis (pre- and post-exposure) and treatment. The improvement in the availability and safety profile of vaccines and the therapeutic antibodies has helped immensely in reducing the worldwide burden of anthrax. AREAS COVERED: Current recommendations for anthrax prophylaxis and control, vaccines and therapeutic antibodies, the recent endeavors, particularly, made after 2010 toward making them safer and more efficacious along with our opinion on its future course. Primarily, PubMed and Europe PMC were searched to cover the recent developments in the above-indicated areas. EXPERT OPINION: Some key existing lacunae in our understanding of the working of biologicals-based anthrax-control measures, i.e., vaccines and therapeutic antibodies, should be addressed to improve their overall stability, safety profile, and efficacy. The identification of novel inhibitors targeting different key-molecules and vital-steps contributing to the overall anthrax pathophysiology could make a difference in anthrax control.


Subject(s)
Anthrax Vaccines/therapeutic use , Anthrax/prevention & control , Anthrax/therapy , Post-Exposure Prophylaxis/methods , Animals , Anthrax/epidemiology , Anthrax/immunology , Anthrax Vaccines/immunology , History, 20th Century , History, 21st Century , Humans , Post-Exposure Prophylaxis/history , Post-Exposure Prophylaxis/trends , Pre-Exposure Prophylaxis/history , Pre-Exposure Prophylaxis/methods , Pre-Exposure Prophylaxis/trends
9.
J Vis Exp ; (159)2020 05 19.
Article in English | MEDLINE | ID: mdl-32510489

ABSTRACT

The opsono-adherence assay is a functional assay that enumerates the attachment of bacterial pathogens to professional phagocytes. Because adherence is requisite to phagocytosis and killing, the assay is an alternative method to opsono-phagocytic killing assays. An advantage of the opsono-adherence assay is the option of using inactivated pathogens and mammalian cell lines, which allows standardization across multiple experiments. The use of an inactivated pathogen in the assay also facilitates work with biosafety level 3 infectious agents and other virulent pathogens. In our work, the opsono-adherence assay was used to assess the functional ability of antibodies, from sera of animals immunized with an anthrax capsule-based vaccine, to induce adherence of fixed Bacillus anthracis to a mouse macrophage cell line, RAW 264.7. Automated fluorescence microscopy was used to capture images of bacilli adhering to macrophages. Increased adherence was correlated with the presence of anti-capsule antibodies in the serum. Non-human primates that exhibited high serum anti-capsule antibody concentrations were protected from anthrax challenge. Thus, the opsono-adherence assay can be used to elucidate the biological functions of antigen specific antibodies in sera, to evaluate the efficacy of vaccine candidates and other therapeutics, and to serve as a possible correlate of immunity.


Subject(s)
Anthrax Vaccines/immunology , Anthrax/immunology , Antibodies, Bacterial/immunology , Bacillus anthracis/immunology , Bacterial Adhesion , Opsonin Proteins/immunology , Animals , Anthrax/microbiology , Anthrax/prevention & control , Antigens, Bacterial/immunology , Fluorescein-5-isothiocyanate/metabolism , Fluorescence , Humans , Macrophages/immunology , Mice , Primates/immunology , Primates/microbiology , RAW 264.7 Cells
10.
J Leukoc Biol ; 108(3): 773-786, 2020 09.
Article in English | MEDLINE | ID: mdl-32421904

ABSTRACT

Anthrax lethal toxin (LT) is a protease that activates the NLRP1b inflammasome sensor in certain rodent strains. Unlike better-studied sensors, relatively little is known about the priming requirements for NLRP1b. In this study, we investigate the rapid and striking priming-independent LT-induced release of IL-1ß in mice within hours of toxin challenge. We find IL-1ß release to be a NLRP1b- and caspase-1-dependent, NLRP3 and caspase-11-independent event that requires both neutrophils and peptidyl arginine deiminiase-4 (PAD4) activity. The simultaneous LT-induced IL-18 response is neutrophil-independent. Bone marrow reconstitution experiments in mice show toxin-induced IL-1ß originates from hematopoietic cells. LT treatment of neutrophils in vitro did not induce IL-1ß, neutrophil extracellular traps (NETs), or pyroptosis. Although platelets interact closely with neutrophils and are also a potential source of IL-1ß, they were unable to bind or endocytose LT and did not secrete IL-1ß in response to the toxin. LT-treated mice had higher levels of cell-free DNA and HMGB1 in circulation than PBS-treated controls, and treatment of mice with recombinant DNase reduced the neutrophil- and NLRP1-dependent IL-1ß release. DNA sensor AIM2 deficiency, however, did not impact IL-1ß release. These data, in combination with the findings on PAD4, suggest a possible role for in vivo NETs or cell-free DNA in cytokine induction in response to LT challenge. Our findings suggest a complex interaction of events and/or mediators in LT-treated mice with the neutrophil as a central player in induction of a profound and rapid inflammatory response to toxin.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Antigens, Bacterial/toxicity , Apoptosis Regulatory Proteins/physiology , Bacillus anthracis/pathogenicity , Bacterial Toxins/toxicity , Extracellular Traps/physiology , Interleukin-1beta/metabolism , Neutrophils/metabolism , Protein-Arginine Deiminase Type 4/physiology , Adaptor Proteins, Signal Transducing/deficiency , Animals , Anthrax/immunology , Antigens, Bacterial/pharmacology , Apoptosis Regulatory Proteins/deficiency , Bacillus anthracis/physiology , Bacterial Toxins/pharmacology , Inflammasomes/physiology , Mice , Mice, 129 Strain , Mice, Congenic , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Monocytes/drug effects , Monocytes/physiology , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , Neutrophils/drug effects , Protein-Arginine Deiminase Type 4/deficiency , Pyroptosis/drug effects , Radiation Chimera , Species Specificity , Spores, Bacterial
11.
Infect Immun ; 88(8)2020 07 21.
Article in English | MEDLINE | ID: mdl-32393506

ABSTRACT

Bacillus anthracis is the causative agent of anthrax disease, presents with high mortality, and has been at the center of bioweapon efforts. The only currently U.S. FDA-approved vaccine to prevent anthrax in humans is anthrax vaccine adsorbed (AVA), which is protective in several animal models and induces neutralizing antibodies against protective antigen (PA), the cell-binding component of anthrax toxin. However, AVA requires a five-course regimen to induce immunity, along with an annual booster, and is composed of undefined culture supernatants from a PA-secreting strain. In addition, it appears to be ineffective against strains that lack anthrax toxin. Here, we investigated a vaccine formulation consisting of recombinant proteins from a surface-localized heme transport system containing near-iron transporter (NEAT) domains and its efficacy as a vaccine for anthrax disease. The cocktail of five NEAT domains was protective against a lethal challenge of inhaled bacillus spores at 3 and 28 weeks after vaccination. The reduction of the formulation to three NEATs (IsdX1, IsdX2, and Bslk) was as effective as a five-NEAT domain cocktail. The adjuvant alum, approved for use in humans, was as protective as Freund's Adjuvant, and protective vaccination correlated with increased anti-NEAT antibody reactivity and reduced bacterial levels in organs. Finally, the passive transfer of anti-NEAT antisera reduced mortality and disease severity, suggesting the protective component is comprised of antibodies. Collectively, these results provide evidence that a vaccine based upon recombinant NEAT proteins should be considered in the development of a next-generation anthrax vaccine.


Subject(s)
Anthrax Vaccines/immunology , Anthrax/prevention & control , Antibodies, Bacterial/biosynthesis , Antibodies, Neutralizing/biosynthesis , Antigens, Bacterial/immunology , Bacillus anthracis/drug effects , Administration, Inhalation , Alum Compounds/administration & dosage , Animals , Anthrax/immunology , Anthrax/microbiology , Anthrax/mortality , Anthrax Vaccines/administration & dosage , Anthrax Vaccines/genetics , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/genetics , Bacillus anthracis/immunology , Bacillus anthracis/pathogenicity , Bacterial Proteins/administration & dosage , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Carrier Proteins/administration & dosage , Carrier Proteins/genetics , Carrier Proteins/immunology , Complement C5/deficiency , Female , Freund's Adjuvant/administration & dosage , Humans , Immunogenicity, Vaccine , Mice, Knockout , Survival Analysis , Vaccination/methods
12.
Front Immunol ; 11: 462, 2020.
Article in English | MEDLINE | ID: mdl-32296419

ABSTRACT

Bacillus anthracis poly-γ-D-glutamic acid (PGA) capsule is an essential virulent factor that helps the bacterial pathogen to escape host immunity. Like other encapsulated bacterial species, the B. anthracis capsule may also inhibit complement-mediated clearance and ensure bacterial survival in the host. Previous reports suggest that B. anthracis spore proteins inhibit complement activation. However, the mechanism through which the B. anthracis capsule imparts a survival advantage to the active bacteria has not been demonstrated till date. Thus, to evaluate the role of the PGA capsule in evading host immunity, we have undertaken the present head-to-head comparative study of the phagocytosis and complement activation of non-encapsulated and encapsulated B. anthracis strains. The encapsulated virulent strain exhibited resistance toward complement-dependent and complement-independent bacterial phagocytosis by human macrophages. The non-encapsulated Sterne strain was highly susceptible to phagocytosis by THP-1 macrophages, after incubation with normal human serum (NHS), heat-inactivated serum, and serum-free media, thus indicating that the capsule inhibited both complement-dependent and complement-independent opsonic phagocytosis. An increased binding of C3b and its subsequent activation to C3c and C3dg, which functionally act as potent opsonins, were observed with the non-encapsulated Sterne strain compared with the encapsulated strain. Other known mediators of complement fixation, IgG, C-reactive protein (CRP), and serum amyloid P component (SAP), also bound more prominently with the non-encapsulated Sterne strain. Studies with complement pathway-specific, component-deficient serum demonstrated that the classical pathway was primarily involved in mediating C3b binding on the non-encapsulated bacteria. Both strains equally bound the complement regulatory proteins C4BP and factor H. Importantly, we demonstrated that the negative charge of the PGA capsule was responsible for the differential binding of the complement proteins between the non-encapsulated and encapsulated strains. At lower pH closer to the isoelectric point of PGA, the neutralization of the negative charge was associated with an increased binding of C3b and IgG with the encapsulated B. anthracis strain. Overall, our data have demonstrated that the B. anthracis capsule inhibits complement fixation and opsonization resulting in reduced phagocytosis by macrophages, thus allowing the bacterial pathogen to evade host immunity.


Subject(s)
Anthrax/immunology , Bacillus anthracis/physiology , Macrophages/immunology , Polyglutamic Acid/analogs & derivatives , Antigens, Bacterial/immunology , Bacillus anthracis/pathogenicity , Bacterial Capsules/immunology , Bacterial Capsules/metabolism , Complement Activation , Complement C3b/metabolism , Humans , Immune Evasion , Opsonin Proteins/metabolism , Phagocytosis , Polyglutamic Acid/metabolism , Protein Binding , THP-1 Cells , Virulence
13.
PLoS One ; 15(4): e0230782, 2020.
Article in English | MEDLINE | ID: mdl-32294093

ABSTRACT

Understanding immune responses to native antigens in response to natural infections can lead to improved approaches to vaccination. This study sought to characterize the humoral immune response to anthrax toxin components, capsule and spore antigens in individuals (n = 46) from the Kayseri and Malatya regions of Turkey who had recovered from mild or severe forms of cutaneous anthrax infection, compared to regional healthy controls (n = 20). IgG antibodies to each toxin component, the poly-γ-D-glutamic acid capsule, the Bacillus collagen-like protein of anthracis (BclA) spore antigen, and the spore carbohydrate anthrose, were detected in the cases, with anthrax toxin neutralization and responses to Protective Antigen (PA) and Lethal Factor (LF) being higher following severe forms of the disease. Significant correlative relationships among responses to PA, LF, Edema Factor (EF) and capsule were observed among the cases. Though some regional control sera exhibited binding to a subset of the tested antigens, these samples did not neutralize anthrax toxins and lacked correlative relationships among antigen binding specificities observed in the cases. Comparison of serum binding to overlapping decapeptides covering the entire length of PA, LF and EF proteins in 26 cases compared to 8 regional controls revealed that anthrax toxin-neutralizing antibody responses elicited following natural cutaneous anthrax infection are directed to conformational epitopes. These studies support the concept of vaccination approaches that preserve conformational epitopes.


Subject(s)
Anthrax/immunology , Antibodies, Bacterial/immunology , Antibodies, Neutralizing/immunology , Antigens, Bacterial/immunology , Bacterial Toxins/immunology , Epitopes/immunology , Skin Diseases, Bacterial/immunology , Adult , Anthrax Vaccines/immunology , Antibody Specificity/immunology , Bacillus anthracis/immunology , Female , Humans , Immunity, Humoral/immunology , Immunoglobulin G/immunology , Male , Middle Aged , Neutralization Tests/methods , Turkey , Young Adult
14.
J Appl Microbiol ; 129(2): 443-452, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32118336

ABSTRACT

AIM: Category A classified Bacillus anthracis is highly fatal pathogen that causes anthrax and creates challenges for global security and public health. In this study, development of a safe and ideal next-generation subunit anthrax vaccine has been evaluated in mouse model. METHOD AND RESULTS: Protective antigen (PA) and BA3338, a surface layer homology (SLH) domain possessing protein were cloned, expressed in heterologous system and purified by IMAC. Recombinant PA and BA3338 with alum were administered in mouse alone or in combination. The humoral and cell-mediated immune response was measured by ELISA and vaccinated animals were challenged with B. anthracis spores via intraperitoneal route. The circulating IgG antibody titre of anti-PA and anti-BA3338 was found significantly high in the first and second booster sera. A significant enhanced level of IL-4, IFN-γ and IL-12 was observed in antigens stimulated supernatant of splenocytes of PA + BA3338 vaccinated animals. A combination of PA and BA3338 provided 80% protection against 20 LD50 lethal dose of B. anthracis spores. CONCLUSION: Both antigens induced admirable humoral and cellular immune response as well as protective efficacy against B. anthracis spores. SIGNIFICANCE AND IMPACT OF THE STUDY: This study has been evaluated for the first time using BA3338 as a vaccine candidate alone or in combination with well-known anthrax vaccine candidate PA. The findings of this study demonstrated that BA3338 could be a co-vaccine candidate for development of dual subunit vaccine against anthrax.


Subject(s)
Anthrax Vaccines/administration & dosage , Anthrax/prevention & control , Antigens, Bacterial/immunology , Bacillus anthracis/immunology , Bacterial Toxins/immunology , Membrane Glycoproteins/immunology , Adjuvants, Immunologic/administration & dosage , Alum Compounds/administration & dosage , Animals , Anthrax/immunology , Anthrax Vaccines/immunology , Antibodies, Bacterial/blood , Cytokines/metabolism , Disease Models, Animal , Immunization/methods , Mice , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology
15.
Int J Nanomedicine ; 15: 239-252, 2020.
Article in English | MEDLINE | ID: mdl-32021177

ABSTRACT

INTRODUCTION: Aluminum salts, although they have been used as adjuvants in many vaccine formulations since 1926, exclusively induce a Th2-biased immune response, thereby limiting their use against intracellular pathogens like Mycobacterium tuberculosis. METHODS AND RESULTS: Herein, we synthesized amorphous and crystalline forms of aluminum hydroxide nanoparticles (AH nps) of 150-200 nm size range. Using Bacillus anthracis protective antigen domain 4 (D4) as a model antigen, we demonstrated that both amorphous and crystalline forms of AH nps displayed enhanced antigen D4 uptake by THP1 cells as compared to commercial adjuvant aluminum hydroxide gel (AH gel). In a mouse model, both amorphous and crystalline AH nps triggered an enhanced D4-specific Th2- and Th1-type immune response and conferred superior protection against anthrax spore challenge as compared to AH gel. Physicochemical characterization of crystalline and amorphous AH nps revealed stronger antigen D4 binding and release than AH gel. CONCLUSION: These results demonstrate that size and crystallinity of AH nps play important roles in mediating enhanced antigen presenting cells (APCs) activation and potentiating a strong antigen-specific immune response, and are critical parameters for the rational design of alum-based Th1-type adjuvant to induce a more balanced antigen-specific immune response.


Subject(s)
Adjuvants, Immunologic/pharmacology , Aluminum Hydroxide/chemistry , Anthrax/prevention & control , Antigens, Bacterial/immunology , Bacterial Toxins/immunology , Metal Nanoparticles/chemistry , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacokinetics , Aluminum Hydroxide/immunology , Aluminum Hydroxide/pharmacology , Animals , Anthrax/immunology , Anthrax Vaccines/chemistry , Anthrax Vaccines/immunology , Anthrax Vaccines/pharmacology , Cell Line , Disease Models, Animal , Dynamic Light Scattering , Female , Humans , Mice , RAW 264.7 Cells , Spectroscopy, Fourier Transform Infrared , Th1 Cells/immunology
16.
mSphere ; 5(1)2020 01 15.
Article in English | MEDLINE | ID: mdl-31941807

ABSTRACT

Protective antigen (PA) is a component of anthrax toxin that can elicit toxin-neutralizing antibody responses. PA is also the major antigen in the current vaccine to prevent anthrax, but stability problems with recombinant proteins have complicated the development of new vaccines containing recombinant PA. The relationship between antigen physical stability and immunogenicity is poorly understood, but there are theoretical reasons to think that this parameter can affect immune responses. We investigated the immunogenicity of anthrax PA, in the presence and absence of the soluble von Willebrand factor A domain of the human form of receptor capillary morphogenesis protein 2 (sCMG2), to elicit antibodies to PA in BALB/c mice. Prior studies showed that sCMG2 stabilizes the 83-kDa PA structure to pH, chemical denaturants, temperature, and proteolysis and slows the hydrogen-deuterium exchange rate of histidine residues far from the binding interface. In contrast to a vaccine containing PA without adjuvant, we found that mice immunized with PA in stable complex with sCMG2 showed markedly reduced antibody responses to PA, including toxin-neutralizing antibodies and antibodies to domain 4, which correlated with fewer toxin-neutralizing antibodies. In contrast, mice immunized with PA in concert with a nonbinding mutant of sCMG2 (D50A) showed anti-PA antibody responses similar to those observed with PA alone. Our results suggest that addition of sCMG2 to a PA vaccine formulation is likely to result in a significantly diminished immune response, but we discuss the multitude of factors that could contribute to reduced immunogenicity.IMPORTANCE The anthrax toxin PA is the major immunogen in the current anthrax vaccine (anthrax vaccine adsorbed). Improving the anthrax vaccine for avoidance of a cold chain necessitates improvements in the thermodynamic stability of PA. We address how stabilizing PA using sCMG2 affects PA immunogenicity in BALB/c mice. Although the stability of PA is increased by binding to sCMG2, PA immunogenicity is decreased. This study emphasizes that, while binding of a ligand retains or improves conformational stability without affecting the native sequence, epitope recognition or processing may be affected, abrogating an effective immune response.


Subject(s)
Anthrax Vaccines/immunology , Antigens, Bacterial/metabolism , Bacterial Toxins/metabolism , Immunogenicity, Vaccine , Receptors, Peptide/immunology , von Willebrand Factor/metabolism , Animals , Anthrax/immunology , Anthrax/prevention & control , Antibodies, Bacterial/blood , Antibodies, Neutralizing/blood , Antigens, Bacterial/immunology , Bacterial Toxins/immunology , Epitopes/immunology , Epitopes/metabolism , Female , Humans , Mice , Mice, Inbred BALB C , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/immunology , von Willebrand Factor/immunology
17.
Innate Immun ; 26(4): 301-311, 2020 05.
Article in English | MEDLINE | ID: mdl-31711335

ABSTRACT

Inflammasomes act as sensors of infection or damage to initiate immune responses. While extensively studied in rodents, understanding of livestock inflammasomes is limited. The NLRP1 inflammasome sensor in rodents is activated by Toxoplasma gondii, Bacillus anthracis lethal toxin (LT), and potentially other zoonotic pathogens. LT activates NLRP1 by N-terminal proteolysis, inducing macrophage pyroptosis and a pro-inflammatory cytokine response. In contrast, NLRP1 in macrophages from humans and certain rodent strains is resistant to LT cleavage, and pyroptosis is not induced. Evolution of NLRP1 sequences towards those leading to pyroptosis is of interest in understanding innate immune responses in different hosts. We characterized NLRP1 in cattle (Bos taurus) and American bison (Bison bison). Bovine NLRP1 is not cleaved by LT, and cattle and bison macrophages do not undergo toxin-induced pyroptosis. Additionally, we found a predicted Nlrp1 splicing isoform in cattle macrophages lacking the N-terminal domain. Resistance to LT in bovine and human NLRP1 correlates with evolutionary sequence similarity to rodents. Consistent with LT-resistant rodents, bovine macrophages undergo a slower non-pyroptotic death in the presence of LPS and LT. Overall, our findings support the model that NLRP1 activation by LT requires N-terminal cleavage, and provide novel information on mechanisms underlying immune response diversity.


Subject(s)
Anthrax/immunology , Bacillus anthracis/physiology , Bison/immunology , Cattle/immunology , Inflammasomes/metabolism , Macrophages/immunology , NLR Proteins/metabolism , Toxoplasma/physiology , Toxoplasmosis/immunology , Animals , Antigens, Bacterial/metabolism , Apoptosis , Bacterial Toxins/metabolism , Caspase 1/metabolism , Cells, Cultured , Immunity, Innate , Proteolysis , Species Specificity
18.
Med Microbiol Immunol ; 209(2): 125-137, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31811379

ABSTRACT

The most promising means of controlling anthrax, a lethal zoonotic disease during the early infection stages, entail restricting the resilient infectious form, i.e., the spores from proliferating to replicating bacilli in the host. The extractible antigen (EA1), a major S-layer protein present on the vegetative cells and spores of Bacillus anthracis, is highly immunogenic and protects mice against lethal challenge upon immunization. In the present study, mice were immunized with r-EA1C, the C terminal crystallization domain of EA1, to generate a neutralizing monoclonal antibody EA752-862, that was evaluated for its anti-spore and anti-bacterial properties. The monoclonal antibody EA752-862 had a minimum inhibitory concentration of 0.08 mg/ml, was bactericidal at a concentration of 0.1 mg/ml and resulted in 100% survival of mice against challenge with B. anthracis vegetative cells. Bacterial cell lysis as observed by scanning electron microscopy and nucleic acid leakage assay could be attributed as a possible mechanism for the bactericidal property. The association of mAb EA752-862 with spores inhibits their subsequent germination to vegetative cells in vitro, enhances phagocytosis of the spores and killing of the vegetative cells within the macrophage, and subsequently resulted in 90% survival of mice upon B. anthracis Ames spore challenge. Therefore, owing to its anti-spore and bactericidal properties, the present study demonstrates mAb EA752-862 as an efficient neutralizing antibody that hinders the establishment of early infection before massive multiplication and toxin release takes place.


Subject(s)
Anthrax/prevention & control , Anti-Bacterial Agents/therapeutic use , Antibodies, Monoclonal/therapeutic use , Bacillus anthracis/immunology , Spores, Bacterial/immunology , Animals , Anthrax/immunology , Anti-Bacterial Agents/biosynthesis , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Antibodies, Bacterial/biosynthesis , Antibodies, Bacterial/chemistry , Antibodies, Bacterial/isolation & purification , Antibodies, Bacterial/pharmacology , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/biosynthesis , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/isolation & purification , Antibodies, Neutralizing/pharmacology , Antigens, Bacterial/immunology , Bacillus anthracis/drug effects , Binding Sites , Female , Immunization , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Microscopy, Electron, Scanning , Phagocytosis/drug effects , Phagocytosis/immunology , Spores, Bacterial/drug effects
19.
Morb. mort. wkly. rep. (Print) ; 68(4): 1-14, Dec. 13, 2019. tab
Article in English | AIM (Africa), BIGG - GRADE guidelines | ID: biblio-1397576

ABSTRACT

This report updates the 2009 recommendations from the CDC Advisory Committee on Immunization Practices (ACIP) regarding use of anthrax vaccine in the United States (Wright JG, Quinn CP, Shadomy S, Messonnier N. Use of anthrax vaccine in the United States: recommendations of the Advisory Committee on Immunization Practices [ACIP)], 2009. MMWR Recomm Rep 2010;59[No. RR-6]). The report 1) summarizes data on estimated efficacy in humans using a correlates of protection model and safety data published since the last ACIP review, 2) provides updated guidance for use of anthrax vaccine adsorbed (AVA) for preexposure prophylaxis (PrEP) and in conjunction with antimicrobials for postexposure prophylaxis (PEP), 3) provides updated guidance regarding PrEP vaccination of emergency and other responders, 4) summarizes the available data on an investigational anthrax vaccine (AV7909), and 5) discusses the use of anthrax antitoxins for PEP. Changes from previous guidance in this report include the following: 1) a booster dose of AVA for PrEP can be given every 3 years instead of annually to persons not at high risk for exposure to Bacillus anthracis who have previously received the initial AVA 3-dose priming and 2-dose booster series and want to maintain protection; 2) during a large-scale emergency response, AVA for PEP can be administered using an intramuscular route if the subcutaneous route of administration poses significant materiel, personnel, or clinical challenges that might delay or preclude vaccination; 3) recommendations on dose-sparing AVA PEP regimens if the anthrax vaccine supply is insufficient to vaccinate all potentially exposed persons; and 4) clarification on the duration of antimicrobial therapy when used in conjunction with vaccine for PEP. These updated recommendations can be used by health care providers and guide emergency preparedness officials and planners who are developing plans to provide anthrax vaccine, including preparations for a wide-area aerosol release of B. anthracis spores. The recommendations also provide guidance on dose-sparing options, if needed, to extend the supply of vaccine to increase the number of persons receiving PEP in a mass casualty event.


Subject(s)
Humans , Adolescent , Adult , Middle Aged , Aged , Centers for Disease Control and Prevention, U.S. , Anthrax Vaccines/therapeutic use , Advisory Committees , Anthrax/immunology , Antitoxins/therapeutic use , Anthrax Vaccines/administration & dosage
20.
BMC Infect Dis ; 19(1): 940, 2019 Nov 07.
Article in English | MEDLINE | ID: mdl-31699037

ABSTRACT

BACKGROUND: Bacillus anthracis causes a highly lethal infectious disease primarily due to toxin-mediated injury. Antibiotics are no longer effective to treat the accumulation of anthrax toxin, thereby new strategies of antibody treatment are essential. Two anti- anthrax protective antigen (PA) antibodies, hmPA6 and PA21, have been reported by our lab previously. METHODS: The mechanisms of the two antibodies were elucidated by Electrophoresis, Competitive Enzyme-linked immune sorbent assay, Western blot analysis and immunoprecipitation test, and in vitro, in vivo (F344 rats) treatment test. The epitopes of the two antibodies were proved by Western blot and Enzyme-linked immune sorbent assay with different domains of PA. RESULTS: In this study, we compared affinity and neutralization of these two antibodies. PA21 was better in protecting cells and rats, whereas hmPA6 had higher affinity. Furthermore, the neutralization mechanisms of the two antibodies and their recognition domains of PA were studied. The results showed that hmPA6 recognized domain IV, thus PA could not bind to cell receptors. Conversely, PA21 recognized domain II, thereby limiting heptamer oligomerization of PA63 in cells. CONCLUSIONS: Our studies elucidated the mechanisms and epitopes of hmPA6 and PA21. The present investigation can advance future use of the two antibodies in anthrax treatment or prophylaxis, and potentially as a combination treatment as the antibodies target different epitopes.


Subject(s)
Antibodies, Bacterial/metabolism , Antibodies, Neutralizing/metabolism , Bacillus anthracis/immunology , Animals , Anthrax/immunology , Antibodies, Bacterial/immunology , Antibodies, Bacterial/pharmacology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/pharmacology , Antigens, Bacterial/toxicity , Bacterial Toxins/toxicity , Cell Line , Cell Survival/drug effects , Electrophoresis , Epitopes/analysis , Epitopes/immunology , Immunoassay , Rats , Rats, Inbred F344
SELECTION OF CITATIONS
SEARCH DETAIL
...