Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 219
Filter
1.
Nature ; 612(7938): 132-140, 2022 12.
Article in English | MEDLINE | ID: mdl-36385533

ABSTRACT

Bacteria have evolved diverse immunity mechanisms to protect themselves against the constant onslaught of bacteriophages1-3. Similar to how eukaryotic innate immune systems sense foreign invaders through pathogen-associated molecular patterns4 (PAMPs), many bacterial immune systems that respond to bacteriophage infection require phage-specific triggers to be activated. However, the identities of such triggers and the sensing mechanisms remain largely unknown. Here we identify and investigate the anti-phage function of CapRelSJ46, a fused toxin-antitoxin system that protects Escherichia coli against diverse phages. Using genetic, biochemical and structural analyses, we demonstrate that the C-terminal domain of CapRelSJ46 regulates the toxic N-terminal region, serving as both antitoxin and phage infection sensor. Following infection by certain phages, newly synthesized major capsid protein binds directly to the C-terminal domain of CapRelSJ46 to relieve autoinhibition, enabling the toxin domain to pyrophosphorylate tRNAs, which blocks translation to restrict viral infection. Collectively, our results reveal the molecular mechanism by which a bacterial immune system directly senses a conserved, essential component of phages, suggesting a PAMP-like sensing model for toxin-antitoxin-mediated innate immunity in bacteria. We provide evidence that CapRels and their phage-encoded triggers are engaged in a 'Red Queen conflict'5, revealing a new front in the intense coevolutionary battle between phages and bacteria. Given that capsid proteins of some eukaryotic viruses are known to stimulate innate immune signalling in mammalian hosts6-10, our results reveal a deeply conserved facet of immunity.


Subject(s)
Bacteriophages , Capsid Proteins , Escherichia coli , Immunity, Innate , Animals , Antitoxins/immunology , Bacteriophages/immunology , Capsid Proteins/immunology , Escherichia coli/immunology , Escherichia coli/virology , Eukaryota/immunology , Pathogen-Associated Molecular Pattern Molecules/immunology
2.
Front Immunol ; 13: 831536, 2022.
Article in English | MEDLINE | ID: mdl-35185923

ABSTRACT

Abrin, a type-II ribosome inactivating protein from the seed of Abrus precatorius, is classified as a Category B bioterrorism warfare agent. Due to its high toxicity, ingestion by animals or humans will lead to death from multiple organ failure. Currently, no effective agents have been reported to treat abrin poisoning. In this study, a novel anti-abrin neutralizing antibody (S008) was humanized using computer-aided design, which possessed lower immunogenicity. Similar to the parent antibody, a mouse anti-abrin monoclonal antibody, S008 possessed high affinity and showed a protective effect against abrin both in vitro and in vivo, and protected mice that S008 was administered 6 hours after abrin. S008 was found that it did not inhibit entry of abrin into cells, suggesting an intracellular blockade capacity against the toxin. In conclusion, this work demonstrates that S008 is a high affinity anti-abrin antibody with both a neutralizing and protective effect and may be an excellent candidate for clinical treatment of abrin poisoning.


Subject(s)
Abrin/immunology , Abrin/toxicity , Antibodies, Monoclonal, Humanized/immunology , Antitoxins/immunology , Poisoning/prevention & control , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized/administration & dosage , Antitoxins/administration & dosage , Female , Mice , Mice, Inbred BALB C , Survival Rate
3.
Toxins (Basel) ; 13(9)2021 09 21.
Article in English | MEDLINE | ID: mdl-34564675

ABSTRACT

Botulinum neurotoxins (BoNT) are some of the most toxic proteins known and can induce respiratory failure requiring long-term intensive care. Treatment of botulism includes the administration of antitoxins. Monoclonal antibodies (mAbs) hold considerable promise as BoNT therapeutics and prophylactics, due to their potency and safety. A three-mAb combination has been developed that specifically neutralizes BoNT serotype A (BoNT/A), and a separate three mAb combination has been developed that specifically neutralizes BoNT serotype B (BoNT/B). A six mAb cocktail, designated G03-52-01, has been developed that combines the anti-BoNT/A and anti-BoNT/B mAbs. The pharmacokinetics and neutralizing antibody concentration (NAC) of G03-52-01 has been determined in guinea pigs, and these parameters were correlated with protection against an inhalation challenge of BoNT/A1 or BoNT/B1. Previously, it was shown that each antibody demonstrated a dose-dependent mAb serum concentration and reached maximum circulating concentrations within 48 h after intramuscular (IM) or intraperitoneal (IP) injection and that a single IM injection of G03-52-01 administered 48 h pre-exposure protected guinea pigs against an inhalation challenge of up to 93 LD50s of BoNT/A1 and 116 LD50s of BoNT/B1. The data presented here advance our understanding of the relationship of the neutralizing NAC to the measured circulating antibody concentration and provide additional support that a single IM or intravenous (IV) administration of G03-52-01 will provide pre-exposure prophylaxis against botulism from an aerosol exposure of BoNT/A and BoNT/B.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/therapeutic use , Antitoxins/therapeutic use , Botulinum Toxins/toxicity , Botulism/drug therapy , Clostridium botulinum/genetics , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antitoxins/immunology , Disease Models, Animal , Drug Combinations , Guinea Pigs , Mice , Serogroup
4.
Anaerobe ; 71: 102415, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34298152

ABSTRACT

OBJECTIVES: To improve the diagnostic accuracy of Clostridioides difficile infection, current U.S. and E.U. guidelines recommend multistep testing that detects the presence of C. difficile and toxin in clinically relevant stool samples to confirm active disease. An accepted gold standard to detect C. difficile toxins is the cell cytotoxicity neutralization assay (CCNA). Although highly sensitive, the traditional CCNA has limitations. One such limitation is the subjective interpretation of an analyst to recognize cytopathic effects in cultured cells exposed to a fecal sample containing toxin. To overcome this limitation, an automated CCNA was developed that replaces most human pipetting steps with robotics and incorporates CellTiterGlo® for a semi-quantitative, non-subjective measure of cell viability instead of microscopy. METHODS: To determine sample positivity and control for non-specific cytopathic effects, two thresholds were defined and validated by evaluating the sample with/without antitoxin antisera (sample-antitoxin/sample + antitoxin): 1) a >70% cell viability threshold was validated with samples containing anti-toxin, and 2) a >1.2-fold difference cut-off where sample results above the cut-off are considered positive. RESULTS: Assay validation demonstrated excellent accuracy, precision, and sample linearity with an LOD of 126.9 pg/mL toxin-B in stool. The positivity cut-offs were clinically validated by comparing 322 diarrheal stool sample results with those run in a predicate, microscopic readout-based CCNA. The automated CCNA demonstrated 96% sensitivity and 100% specificity compared with the predicate CCNA. CONCLUSIONS: Overall, the automated CCNA provides a specific, sensitive, and reproducible tool to support determination of CDI epidemiology or the efficacy of interventions such as vaccines.


Subject(s)
Automation/methods , Clostridioides difficile/isolation & purification , Diarrhea/diagnosis , Diarrhea/microbiology , Feces/microbiology , Neutralization Tests/methods , Antitoxins/analysis , Antitoxins/immunology , Automation/instrumentation , Bacterial Toxins/analysis , Bacterial Toxins/immunology , Bacterial Toxins/toxicity , Cell Culture Techniques , Clostridioides difficile/classification , Clostridioides difficile/genetics , Clostridium Infections/diagnosis , Clostridium Infections/microbiology , Feces/chemistry , Humans , Sensitivity and Specificity
5.
Infect Immun ; 89(10): e0027421, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34097471

ABSTRACT

The pathology associated with Clostridioides difficile disease is caused in large part by TcdB, an intracellular bacterial toxin that inactivates small GTPases. Despite C. difficile causing enteric disease, antitoxin IgG is a clear correlate of protection against infection-associated pathology. Immunization with TcdB-based immunogens or passive transfer of monoclonal antibodies specific for the TcdB carboxy-terminal domain (CTD) confers protection following C. difficile infection. Whether the mechanism by which circulating IgG is delivered to the gut depends on specific receptor-mediated transport or is solely reflective of infection-induced damage to the gut remains unclear. Here, we tested the hypothesis that neonatal Fc receptor (FcRn) is required for the delivery of systemic TcdB-specific IgG to the gut and protection against C. difficile-associated pathology. FcRn-expressing mice and FcRn-deficient littermates were immunized subcutaneously with Alhydrogel adjuvant-adsorbed CTD before challenge with live C. difficile spores. FcRn was required for the delivery of systemic TcdB-specific IgG to the gut and for vaccine-induced protection against C. difficile-associated disease. The lack of FcRn expression had minimal effects on the composition of the gut microbiome and did not affect susceptibility to C. difficile infection in nonimmunized mice. In further experiments, intraperitoneal injection of immune sera in FcRn-deficient mice led to the transport of protective IgG to the gut independently of infection, confirming a reported method of bypassing the FcRn. Our results reveal an FcRn-dependent mechanism by which systemic immunization-induced IgG protects the gut during enteric C. difficile infection. These findings may be beneficial for the targeting of C. difficile-specific IgG to the gut.


Subject(s)
Clostridioides difficile/immunology , Clostridium Infections/immunology , Digestive System/immunology , Digestive System/microbiology , Disease Susceptibility/immunology , Histocompatibility Antigens Class I/immunology , Immunoglobulin G/immunology , Receptors, Fc/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antitoxins/immunology , Bacterial Toxins/immunology , Clostridium Infections/microbiology , Disease Susceptibility/microbiology , Enterotoxins/immunology , Female , Immunity/immunology , Immunization/methods , Male , Mice , Mice, Inbred C57BL , Vaccination/methods
6.
Nat Microbiol ; 6(3): 313-326, 2021 03.
Article in English | MEDLINE | ID: mdl-33462434

ABSTRACT

Fungi of the order Mucorales cause mucormycosis, a lethal infection with an incompletely understood pathogenesis. We demonstrate that Mucorales fungi produce a toxin, which plays a central role in virulence. Polyclonal antibodies against this toxin inhibit its ability to damage human cells in vitro and prevent hypovolemic shock, organ necrosis and death in mice with mucormycosis. Inhibition of the toxin in Rhizopus delemar through RNA interference compromises the ability of the fungus to damage host cells and attenuates virulence in mice. This 17 kDa toxin has structural and functional features of the plant toxin ricin, including the ability to inhibit protein synthesis through its N-glycosylase activity, the existence of a motif that mediates vascular leak and a lectin sequence. Antibodies against the toxin inhibit R. delemar- or toxin-mediated vascular permeability in vitro and cross react with ricin. A monoclonal anti-ricin B chain antibody binds to the toxin and also inhibits its ability to cause vascular permeability. Therefore, we propose the name 'mucoricin' for this toxin. Not only is mucoricin important in the pathogenesis of mucormycosis but our data suggest that a ricin-like toxin is produced by organisms beyond the plant and bacterial kingdoms. Importantly, mucoricin should be a promising therapeutic target.


Subject(s)
Mucorales/pathogenicity , Mucormycosis/pathology , Mycotoxins/metabolism , Ricin/metabolism , Animals , Antitoxins/immunology , Antitoxins/pharmacology , Antitoxins/therapeutic use , Apoptosis , Capillary Permeability , Cells, Cultured , Cross Reactions , Humans , Hyphae/chemistry , Hyphae/pathogenicity , Lectins/metabolism , Mice , Mucorales/chemistry , Mucorales/classification , Mucorales/genetics , Mucormycosis/microbiology , Mucormycosis/prevention & control , Mycotoxins/chemistry , Mycotoxins/genetics , Mycotoxins/immunology , Necrosis , RNA Interference , Rhizopus/chemistry , Rhizopus/genetics , Rhizopus/pathogenicity , Ribosome Inactivating Proteins/metabolism , Ricin/chemistry , Ricin/immunology , Virulence/drug effects , Virulence/genetics
7.
Cell Host Microbe ; 27(6): 937-949.e6, 2020 06 10.
Article in English | MEDLINE | ID: mdl-32396840

ABSTRACT

Typhoidal and non-typhoidal Salmonelleae (NTS) cause typhoid fever and gastroenteritis, respectively, in humans. Salmonella typhoid toxin contributes to typhoid disease progression and chronic infection, but little is known about the role of its NTS ortholog. We found that typhoid toxin and its NTS ortholog induce different clinical presentations. The PltB subunit of each toxin exhibits different glycan-binding preferences that correlate with glycan expression profiles of host cells targeted by each bacterium at the primary infection or intoxication sites. Through co-crystal structures of PltB subunits bound to specific glycan receptor moieties, we show that they induce markedly different glycan-binding preferences and virulence outcomes. Furthermore, immunization with the NTS S. Javiana or its toxin offers cross-reactive protection against lethal-dose typhoid toxin challenge. Cumulatively, these results offer insights into the evolution of host adaptations in Salmonella AB toxins, their cell and tissue tropisms, and the design for improved typhoid vaccines and therapeutics.


Subject(s)
Bacterial Proteins/toxicity , Bacterial Toxins/toxicity , Endotoxins/toxicity , Host Adaptation/drug effects , Host Adaptation/physiology , Salmonella typhi/metabolism , Amino Acid Sequence , Animals , Antitoxins/immunology , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Bacterial Toxins/metabolism , Cross Reactions/immunology , Endotoxins/genetics , Endotoxins/immunology , Endotoxins/metabolism , Female , HEK293 Cells , Humans , Male , Mice, Knockout , Polysaccharides/biosynthesis , Salmonella , Salmonella typhi/immunology , Salmonella typhi/pathogenicity , Typhoid Fever/microbiology , Typhoid Fever/prevention & control , Typhoid-Paratyphoid Vaccines/immunology , Virulence
8.
J Bacteriol ; 202(7)2020 03 11.
Article in English | MEDLINE | ID: mdl-31932311

ABSTRACT

Type II toxin-antitoxin (TA) systems are small genetic elements composed of a toxic protein and its cognate antitoxin protein, the latter counteracting the toxicity of the former. While TA systems were initially discovered on plasmids, functioning as addiction modules through a phenomenon called postsegregational killing, they were later shown to be massively present in bacterial chromosomes, often in association with mobile genetic elements. Extensive research has been conducted in recent decades to better understand the physiological roles of these chromosomally encoded modules and to characterize the conditions leading to their activation. The diversity of their proposed roles, ranging from genomic stabilization and abortive phage infection to stress modulation and antibiotic persistence, in conjunction with the poor understanding of TA system regulation, resulted in the generation of simplistic models, often refuted by contradictory results. This review provides an epistemological and critical retrospective on TA modules and highlights fundamental questions concerning their roles and regulations that still remain unanswered.


Subject(s)
Antitoxins/genetics , Antitoxins/immunology , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Biological Evolution , Toxin-Antitoxin Systems , Genetic Association Studies , Genome, Bacterial , Phenotype , Toxin-Antitoxin Systems/genetics , Toxin-Antitoxin Systems/immunology
9.
Hum Vaccin Immunother ; 16(1): 100-108, 2020.
Article in English | MEDLINE | ID: mdl-31210561

ABSTRACT

Botulinum neurotoxins (BoNTs) are among the most toxic proteins. Vaccination is an effective strategy to prevent botulism. To generate a vaccine suitable for human use, a recombinant non-His-tagged isoform of the Hc domain of botulinum neurotoxin serotype E (rEHc) was expressed in Escherichia coli and purified by sequential chromatography. The immunogenicity of rEHc was evaluated in mice and dose- and time-dependent immune responses were observed in both antibody titers and protective potency. Then, the pilot-scale expression and purification of rEHc were performed, and its immunological activity was characterized. Our results showed rEHc has good immunogenicity and can elicit strong protective potency against botulinum neurotoxin serotype E (BoNT/E) in mice, indicating that rEHc is an effective botulism vaccine candidate. Further, we developed a novel antitoxin against BoNT/E by purifying F(ab')2 from pepsin-digested serum IgG of rEHc-inoculated horses. The protective effect of the F(ab')2 antitoxin was determined in vitro and in vivo. The results showed that our F(ab')2 antitoxin can prevent botulism in BoNT/E-challenged mice and effectively alleviate the progression of paralysis caused by BoNT/E to achieve therapeutic effects. Therefore, our results provide valuable experimental data for the production of a novel antitoxin, which is a promising candidate for the treatment of BoNT/E-induced botulism.


Subject(s)
Antitoxins/immunology , Bacterial Vaccines/immunology , Botulinum Toxins/immunology , Botulism/prevention & control , Animals , Antibodies, Bacterial/blood , Bacterial Vaccines/genetics , Botulism/therapy , Female , Horses/immunology , Immunogenicity, Vaccine , Mice , Mice, Inbred BALB C , Vaccination , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Vaccines, Synthetic/immunology
10.
Appl Environ Microbiol ; 85(11)2019 06 01.
Article in English | MEDLINE | ID: mdl-30926730

ABSTRACT

Enterotoxigenic Escherichia coli (ETEC) strains that produce immunologically heterogeneous fimbriae and enterotoxins are the primary cause of neonatal diarrhea and postweaning diarrhea in young pigs. A multivalent vaccine inducing protective immunity against ideally all ETEC fimbriae and enterotoxins could be effective against diarrhea in young pigs. However, developing a vaccine to broadly protect against various ETEC virulence determinants has proven challenging. Recently developed structure- and epitope-based multiepitope fusion antigen (MEFA) technology that presents neutralizing epitopes of various virulence determinants at a backbone immunogen and that mimics epitope native immunogenicity suggests the feasibility of developing multivalent vaccines. With neutralizing epitopes from ETEC fimbria F18 and enterotoxins being identified, it becomes urgent to identify protective epitopes of K88 (F4) fimbriae, which play a major role in pig neonatal and postweaning diarrhea. In this study, we identified B-cell immunodominant epitopes in silico from the K88ac fimbrial major subunit (also adhesin) FaeG and embedded each epitope in a heterogeneous carrier for epitope fusions. We then immunized mice with each epitope fusion protein and examined epitope antigenicity and also neutralizing activities of epitope-induced antibodies. Data showed that while all nine FaeG epitope fusions induced antibodies to K88ac fimbria, anti-K88 IgG antibodies derived from epitopes MTGDFNGSVD (ep1), LNDLTNGGTK (ep2), GRTKEAFATP (ep3), ELRKPDGGTN (ep4), PMKNAGGTKVGAVKVN (ep5), and RENMEYTDGT (ep8) significantly inhibited adherence of K88ac fimbrial bacteria to porcine intestinal cell line IPEC-J2, indicating that these peptides were the neutralizing epitopes of K88ac fimbrial major subunit FaeG and suggesting the future application of FaeG epitopes in ETEC vaccine development.IMPORTANCE Enterotoxigenic Escherichia coli (ETEC) strains producing K88ac fimbriae and enterotoxins are a major cause of porcine neonatal diarrhea and postweaning diarrhea in the United States. Currently, there is no vaccine to induce broadly protective antiadhesin and antitoxin immunity against ETEC-associated diarrhea. To develop a broadly effective ETEC vaccine, we need to target the most important if not all ETEC virulence determinants. While conventional vaccinology approaches encounter difficulties at integrating or including heterogeneous ETEC fimbria and toxin antigens into a vaccine product, multiepitope fusion antigen (MEFA) structural vaccinology provides a new platform to combine neutralizing antigenic elements or epitopes from various heterogeneous virulence factors for broad immunity and protection. Identification of the neutralizing epitopes of K88ac fimbria from this study added the last antigens to an MEFA-based multivalent vaccine against ETEC-associated diarrhea in pigs. An effective vaccine against pig diarrhea can significantly improve swine health and well-being and reduce economic losses to the swine industry worldwide.


Subject(s)
Adhesins, Bacterial/immunology , Adhesins, Escherichia coli/immunology , Antibodies, Neutralizing/immunology , Enterotoxigenic Escherichia coli/immunology , Epitopes/immunology , Fimbriae, Bacterial/immunology , Adhesins, Escherichia coli/genetics , Amino Acid Sequence , Animals , Antigens, Bacterial/blood , Antitoxins/immunology , Bacterial Toxins/immunology , Diarrhea/microbiology , Enterotoxigenic Escherichia coli/genetics , Enterotoxins/immunology , Epitopes/genetics , Escherichia coli Vaccines/immunology , Female , Fimbriae, Bacterial/chemistry , Gene Expression Regulation, Bacterial , Immunogenicity, Vaccine , Immunoglobulin G , Mice , Mice, Inbred BALB C , Models, Animal , Sequence Analysis, Protein , Swine , Swine Diseases/immunology , Swine Diseases/prevention & control
11.
Pathog Dis ; 76(3)2018 04 01.
Article in English | MEDLINE | ID: mdl-29718183

ABSTRACT

Research on bacterial toxins is closely linked to the birth of immunology. Our understanding of the interaction of bacterial protein toxins with immune cells has helped to decipher immunopathology, develop preventive and curative treatments for infections, and propose anti-cancer immunotherapies. The link started when Behring and Kitasato demonstrated that serotherapy was effective against 'the strangling angel', namely diphtheria, and its dreadful toxin discovered by Roux and Yersin. The antitoxin treatment helped to save thousands of children. Glenny demonstrated the efficacy of the secondary immune response compared to the primary one. Ramon described anatoxins that allowed the elaboration of effective vaccines and discovered the use of adjuvant to boost the antibody response. Similar approaches were later made for the tetanus toxin. Studying antitoxin antibodies Ehrlich demonstrated, for the first time, the transfer of immunity from mother to newborns. In 1989 Marrack and Kappler coined the concept of 'superantigens' to characterize protein toxins that induce T-lymphocyte proliferation, and cytokine release by both T-lymphocytes and antigen presenting cells. More recently, immunotoxins have been designed to kill cancer cells targeted by either specific antibodies or cytokines. Finally, the action of IgE antibodies against toxins may explain their persistence through evolution despite their side effect in allergy.


Subject(s)
Antitoxins/immunology , Bacterial Toxins/immunology , Hypersensitivity/immunology , Immunotherapy/methods , Vaccines/immunology , Adjuvants, Immunologic/therapeutic use , Antibodies, Neutralizing/history , Antibodies, Neutralizing/therapeutic use , Antigen-Presenting Cells/immunology , Antitoxins/chemistry , Antitoxins/history , Antitoxins/therapeutic use , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/chemistry , Bacterial Toxins/history , Cytokines/biosynthesis , Cytokines/metabolism , History, 19th Century , History, 20th Century , Humans , Hypersensitivity/drug therapy , Hypersensitivity/history , Hypersensitivity/physiopathology , Immunotherapy/history , Immunotoxins/chemistry , Immunotoxins/history , Immunotoxins/therapeutic use , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Superantigens/immunology , T-Lymphocytes/immunology , Tetanus/drug therapy , Tetanus/immunology , Tetanus/microbiology , Tetanus/physiopathology , Vaccines/administration & dosage , Vaccines/history
12.
Toxins (Basel) ; 9(10)2017 10 18.
Article in English | MEDLINE | ID: mdl-29057798

ABSTRACT

Ricin is a type II ribosome-inactivating toxin that catalytically inactivates ribosomes ultimately leading to cell death. The toxicity of ricin along with the prevalence of castor beans (its natural source) has led to its increased notoriety and incidences of nefarious use. Despite these concerns, there are no licensed therapies available for treating ricin intoxication. Here, we describe the development of a F(ab')2 polyclonal ovine antitoxin against ricin and demonstrate the efficacy of a single, post-exposure, administration in an in vivo murine model of intoxication against aerosolised ricin. We found that a single dose of antitoxin afforded a wide window of opportunity for effective treatment with 100% protection observed in mice challenged with aerosolised ricin when given 24 h after exposure to the toxin and 75% protection when given at 30 h. Treated mice had reduced weight loss and clinical signs of intoxication compared to the untreated control group. Finally, using imaging flow cytometry, it was found that both cellular uptake and intracellular trafficking of ricin toxin to the Golgi apparatus was reduced in the presence of the antitoxin suggesting both actions can contribute to the therapeutic mechanism of a polyclonal antitoxin. Collectively, the research highlights the significant potential of the ovine F(ab')2 antitoxin as a treatment for ricin intoxication.


Subject(s)
Antitoxins/immunology , Ricin/immunology , Animals , Antibodies, Neutralizing/analysis , Chlorocebus aethiops , Female , Mice, Inbred BALB C , Ricin/pharmacokinetics , Ricin/toxicity , Sheep , Vero Cells
13.
Eur J Pharm Sci ; 106: 404-412, 2017 Aug 30.
Article in English | MEDLINE | ID: mdl-28595875

ABSTRACT

Snakebite envenoming is a major neglected disease related to poverty in developing countries. Treatment involves the administration of a specific antivenom serum and auxiliary therapies, if necessary. The improvement of antibodies is of great importance for the technological advancement of antivenom therapy and to reduce the morbidity and mortality associated with this medical burden. In the present study, adult hens were immunized nine times with 20µg of B. arietans or C. d. terrificus venoms at three-week intervals between immunizations. Developing antibodies presented increasing avidity and affinity to antigenic toxin epitopes along immunization, attaining a plateau after the seventh immunization. Pooled egg yolk-purified IgY antivenom antibodies, subjected to in vitro-in vivo lethality assay using Swiss adult mice, exhibited potent venom lethal neutralizing activity. Taken together, chickens under the described immunization schedule were considered alternative candidates for antivenom production. Lower maintenance costs, a simple antibody manufacturing process and immunization suffering restrictions are additional advantages.


Subject(s)
Antitoxins/immunology , Crotalid Venoms/immunology , Immunoglobulins/immunology , Animals , Bothrops , Chickens , Crotalus , Female , Mice
14.
Diagn Microbiol Infect Dis ; 88(4): 330-334, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28533000

ABSTRACT

BACKGROUND: Clostridium difficile infection (CDI) causes increased morbidity and mortality. Clinical data cannot clearly predict poor CDI outcome. Data on the value of microbiological predictors is scarce. OBJECTIVE: To identify early predictors of poor outcome of CDI. METHODS: We prospectively included patients with CDI aged >2years. Clinical, immunological (Toxin B IgG/Ig A and Toxin A IgG/Ig A), microbiological factors (bacterial load, toxin quantification, sporulation, germination, and metronidazole susceptibility) were evaluated to identify early independent predictors of poor outcome. RESULTS: We identified 204 cases of CDI; outcome was poor in 22.1%. Advanced age, presence of comorbidities, leukocytosis and high toxigenic C. difficile load were independently associated with poor outcome. We could not demonstrate this correlation for antitoxin antibodies. CONCLUSION: We identified high bacterial load as a microbiological predictor of poor outcome. We propose this factor to be included in combined clinical and microbiological prediction rules of poor outcome in CDI.


Subject(s)
Clostridioides difficile/immunology , Clostridium Infections/immunology , Clostridium Infections/microbiology , Aged , Aged, 80 and over , Anti-Bacterial Agents/immunology , Antitoxins/immunology , Bacterial Proteins/immunology , Enterotoxins/immunology , Female , Humans , Male , Metronidazole/immunology , Middle Aged , Prospective Studies
15.
Microbiol Immunol ; 61(7): 272-279, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28543534

ABSTRACT

Enterotoxigenic Escherichia Coli (ETEC) strains are the commonest bacteria causing diarrhea in children in developing countries and travelers to these areas. Colonization factors (CFs) and enterotoxins are the main virulence determinants in ETEC pathogenesis. Heterogeneity of CFs is commonly considered the bottleneck to developing an effective vaccine. It is believed that broad spectrum protection against ETEC would be achieved by induced anti-CF and anti-enterotoxin immunity simultaneously. Here, a fusion antigen strategy was used to construct a quadrivalent recombinant protein called 3CL and composed of CfaB, a structural subunit of CFA/I, and CS6 structural subunits, LTB and STa toxoid of ETEC. Its anti-CF and antitoxin immunogenicity was then assessed. To achieve high-level expression, the 3CL gene was synthesized using E. coli codon bias. Female BALB/C mice were immunized with purified recombinant 3CL. Immunized mice developed antibodies that were capable of detecting each recombinant subunit in addition to native CS6 protein and also protected the mice against ETEC challenge. Moreover, sera from immunized mice also neutralized STa toxin in a suckling mouse assay. These results indicate that 3CL can induce anti-CF and neutralizing antitoxin antibodies along with introducing CFA/I as a platform for epitope insertion.


Subject(s)
Antigens, Bacterial/immunology , Enterotoxigenic Escherichia coli/immunology , Escherichia coli Vaccines/immunology , Recombinant Fusion Proteins/immunology , Toxoids/immunology , Animals , Antibodies, Bacterial/blood , Antibodies, Neutralizing/immunology , Antigens, Bacterial/genetics , Antitoxins/immunology , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Colicins/genetics , Colicins/immunology , Enterotoxins/genetics , Enterotoxins/immunology , Enterotoxins/toxicity , Escherichia coli Infections/immunology , Escherichia coli Infections/prevention & control , Escherichia coli Proteins/genetics , Escherichia coli Proteins/immunology , Escherichia coli Vaccines/genetics , Female , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/genetics , Toxoids/genetics
16.
PLoS One ; 12(3): e0174187, 2017.
Article in English | MEDLINE | ID: mdl-28323873

ABSTRACT

Human botulism is primarily caused by botulinum neurotoxin (BoNT) serotypes A, B and E, with around 1% caused by serotype F (BoNT/F). BoNT/F comprises at least seven different subtypes with the amino acid sequence difference between subtypes as high as 36%. The sequence differences present a significant challenge for generating monoclonal antibodies (mAbs) that can bind, detect and neutralize all BoNT/F subtypes. We used repertoire cloning of immune mouse antibody variable (V) regions and yeast display to generate a panel of 33 lead single chain Fv (scFv) mAbs that bound one or more BoNT/F subtypes with a median equilibrium dissociation constant (KD) of 4.06 × 10-9 M. By diversifying the V-regions of the lead mAbs and selecting for cross reactivity we generated five mAbs that bound each of the seven subtypes. Three scFv binding non-overlapping epitopes were converted to IgG that had KD for the different BoNT/F subtypes ranging from 2.2×10-8 M to 1.47×10-12 pM. An equimolar combination of the mAbs was able to potently neutralize BoNT/F1, F2, F4 and F7 in the mouse neutralization assay. The mAbs have potential utility as diagnostics capable of recognizing the known BoNT/F subtypes and could be developed as antitoxins to prevent and treat type F botulism.


Subject(s)
Antibodies, Monoclonal/immunology , Antitoxins/immunology , Botulinum Toxins/immunology , Single-Chain Antibodies/immunology , Amino Acid Sequence , Animals , Antitoxins/genetics , Botulism/diagnosis , Botulism/therapy , Catalytic Domain/immunology , Clostridium botulinum/metabolism , Cross Reactions/immunology , Epitope Mapping , Epitopes/immunology , Escherichia coli/genetics , Immunization , Mice , Saccharomyces cerevisiae/genetics , Single-Chain Antibodies/genetics
17.
Microb Pathog ; 104: 28-31, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28062294

ABSTRACT

The hallmark patogenicity in Pseudomonas aeruginosa (P. aeruginosa) is biofilm formation that is not easy to eradicate, because it has variety mechanisms for antibiotic resistance. In addition, toxin-antitoxin (TA) system may play role in biofilm formation. The current study aimed to evaluate the role of TA loci in biofilm formation. Therefore, 18 P. aeruginosa clinical isolates were collected and evaluated for specific biofilm and TA genes. The analysis by RT-qPCR demonstrated that expression of mazE antitoxin in biofilm formation was increase. On the other hand, mazE antitoxin TA system was used as target for antisense PNA. mazE-PNA was able to influence in biofilm formation and was inhibit at 5,10 and 15 µM concentrations biofilm formation in P. aeruginosa. Therefore, it could be highlighted target for anti-biofilm target to eradicate P. aeruginosa biofilm producer.


Subject(s)
Antitoxins/immunology , Bacterial Proteins/immunology , Biofilms , Immune Sera/immunology , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/immunology , Antitoxins/genetics , Bacterial Proteins/genetics , Gene Expression , Humans , Pseudomonas Infections/microbiology , Quantitative Trait Loci , Stress, Physiological
18.
PLoS One ; 11(6): e0157970, 2016.
Article in English | MEDLINE | ID: mdl-27336843

ABSTRACT

Clostridium difficile (C. difficile) infection (CDI) is the main cause of nosocomial antibiotic-associated colitis and increased incidence of community-associated diarrhea in industrialized countries. At present, the primary treatment of CDI is antibiotic administration, which is effective but often associated with recurrence, especially in the elderly. Pathogenic strains produce enterotoxin, toxin A (TcdA), and cytotoxin, toxin B (TcdB), which are necessary for C. difficile induced diarrhea and gut pathological changes. Administration of anti-toxin antibodies provides an alternative approach to treat CDI, and has shown promising results in preclinical and clinical studies. In the current study, several humanized anti-TcdA and anti-TcdB monoclonal antibodies were generated and their protective potency was characterized in a hamster infection model. The humanized anti-TcdA (CANmAbA4) and anti-TcdB (CANmAbB4 and CANmAbB1) antibodies showed broad spectrum in vitro neutralization of toxins from clinical strains and neutralization in a mouse toxin challenge model. Moreover, co-administration of humanized antibodies (CANmAbA4 and CANmAbB4 cocktail) provided a high level of protection in a dose dependent manner (85% versus 57% survival at day 22 for 50 mg/kg and 20 mg/kg doses, respectively) in a hamster gastrointestinal infection (GI) model. This study describes the protective effects conferred by novel neutralizing anti-toxin monoclonal antibodies against C. difficile toxins and their potential as therapeutic agents in treating CDI.


Subject(s)
Antibodies, Bacterial , Antibodies, Monoclonal, Humanized , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing , Antitoxins/immunology , Bacterial Proteins/immunology , Bacterial Toxins/immunology , Enterotoxins/immunology , Neutralization Tests , Animals , Clostridioides difficile/immunology , Clostridioides difficile/isolation & purification , Clostridium Infections/immunology , Clostridium Infections/microbiology , Clostridium Infections/mortality , Cricetinae , Disease Models, Animal , Humans , Immunoglobulin G/immunology , Mice , Spores, Bacterial
19.
Antimicrob Agents Chemother ; 60(8): 4878-85, 2016 08.
Article in English | MEDLINE | ID: mdl-27270276

ABSTRACT

Protective antigen (PA)-based vaccines are effective in preventing the development of fatal anthrax disease both in humans and in relevant animal models. The Bacillus anthracis toxins lethal toxin (lethal factor [LF] plus PA) and edema toxin (edema factor [EF] plus PA) are essential for the establishment of the infection, as inactivation of these toxins results in attenuation of the pathogen. Since the toxins reach high toxemia levels at the bacteremic stages of the disease, the CDC's recommendations include combining antibiotic treatment with antitoxin (anti-PA) immunotherapy. We demonstrate here that while treatment with a highly potent neutralizing monoclonal antibody was highly efficient as postexposure prophylaxis treatment, it failed to protect rabbits with any detectable bacteremia (≥10 CFU/ml). In addition, we show that while PA vaccination was effective against a subcutaneous spore challenge, it failed to protect rabbits against systemic challenges (intravenous injection of vegetative bacteria) with the wild-type Vollum strain or a toxin-deficient mutant. To test the possibility that additional proteins, which are secreted by the bacteria under pathogenicity-stimulating conditions in vitro, may contribute to the vaccine's potency, we immunized rabbits with a secreted protein fraction from a toxin-null mutant. The antiserum raised against the secreted fraction reacts with the bacteria in an immunofluorescence assay. Immunization with the secreted protein fraction did not protect the rabbits against a systemic challenge with the fully pathogenic bacteria. Full protection was obtained only by a combined vaccination with PA and the secreted protein fraction. Therefore, these results indicate that an effective antiserum treatment in advanced stages of anthrax must include toxin-neutralizing antibodies in combination with antibodies against bacterial cell targets.


Subject(s)
Anthrax/immunology , Antigens, Bacterial/immunology , Antitoxins/immunology , Bacillus anthracis/immunology , Bacterial Toxins/immunology , Animals , Anthrax/microbiology , Anthrax Vaccines/immunology , Antibodies, Bacterial/immunology , Female , Immune Sera/immunology , Rabbits , Spores, Bacterial/immunology , Vaccination/methods
20.
PLoS One ; 11(6): e0156893, 2016.
Article in English | MEDLINE | ID: mdl-27300140

ABSTRACT

JJX12 is an engineered bispecific antibody against ricin, a member of the medically important A-B family of toxins that exploits retrograde transport as means to gain entry into the cytosol of target cells. JJX12 consists of RTA-D10, a camelid single variable domain (VHH) antibody directed against an epitope on ricin's enzymatic subunit (RTA), linked via a 15-mer peptide to RTB-B7, a VHH against ricin's bivalent galactose binding subunit (RTB). We previously reported that JJX12, but not an equimolar mixture of RTA-D10 and RTB-B7 monomers, was able to passively protect mice against a lethal dose ricin challenge, demonstrating that physically linking RTB-B7 and RTA-D10 is critical for toxin-neutralizing activity in vivo. We also reported that JJX12 promotes aggregation of ricin in solution, presumably through the formation of intermolecular crosslinking. In the current study, we now present evidence that JJX12 affects the dynamics of ricin uptake and trafficking in human epithelial cells. Confocal microscopy, as well as live cell imaging coupled with endocytosis pathway-specific inhibitors, revealed that JJX12-toxin complexes are formed on the surfaces of mammalian cells and internalized via a pathway sensitive to amiloride, a known inhibitor of macropinocytosis. Moreover, in the presence of JJX12, retrograde transport of ricin to the trans-Golgi network was significantly reduced, while accumulation of the toxin in late endosomes was significantly enhanced. In summary, we propose that JJX12, by virtue of its ability to crosslink ricin toxin, alters the route of toxin uptake and trafficking within cells.


Subject(s)
Antibodies, Bispecific/pharmacology , Antitoxins/pharmacology , Protein Aggregates/drug effects , Protein Transport/drug effects , Ricin/metabolism , A549 Cells , Animals , Antibodies, Bispecific/immunology , Antitoxins/immunology , Endocytosis/drug effects , Humans , Mice , Ricin/immunology , Ricinus/chemistry , trans-Golgi Network/drug effects , trans-Golgi Network/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...