Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
EcoSal Plus ; : eesp00042023, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39023252

ABSTRACT

Salmonella enterica is a diverse species that infects both humans and animals. S. enterica subspecies enterica consists of more than 1,500 serovars. Unlike typhoidal Salmonella serovars which are human host-restricted, non-typhoidal Salmonella (NTS) serovars are associated with foodborne illnesses worldwide and are transmitted via the food chain. Additionally, NTS serovars can cause disease in livestock animals causing significant economic losses. Salmonella is a well-studied model organism that is easy to manipulate and evaluate in animal models of infection. Advances in genetic engineering approaches in recent years have led to the development of Salmonella vaccines for both humans and animals. In this review, we focus on current progress of recombinant live-attenuated Salmonella vaccines, their use as a source of antigens for parenteral vaccines, their use as live-vector vaccines to deliver foreign antigens, and their use as therapeutic cancer vaccines in humans. We also describe development of live-attenuated Salmonella vaccines and live-vector vaccines for use in animals.

2.
Vaccines (Basel) ; 9(2)2021 Feb 17.
Article in English | MEDLINE | ID: mdl-33671124

ABSTRACT

The use of live-attenuated bacterial vaccines as carriers for the mucosal delivery of foreign antigens to stimulate the mucosal immune system was first proposed over three decades ago. This novel strategy aimed to induce immunity against at least two distinct pathogens using a single bivalent carrier vaccine. It was first tested using a live-attenuated Salmonella enterica serovar Typhi strain in clinical trials in 1984, with excellent humoral immune responses against the carrier strain but only modest responses elicited against the foreign antigen. Since then, clinical trials with additional Salmonella-based carrier vaccines have been conducted. As with the original trial, only modest foreign antigen-specific immunity was achieved in most cases, despite the incorporation of incremental improvements in antigen expression technologies and carrier design over the years. In this review, we will attempt to deconstruct carrier vaccine immunogenicity in humans by examining the basis of bacterial immunity in the human gastrointestinal tract and how the gut detects and responds to pathogens versus benign commensal organisms. Carrier vaccine design will then be explored to determine the feasibility of retaining as many characteristics of a pathogen as possible to elicit robust carrier and foreign antigen-specific immunity, while avoiding over-stimulation of unacceptably reactogenic inflammatory responses.

3.
Front Immunol ; 9: 2543, 2018.
Article in English | MEDLINE | ID: mdl-30443257

ABSTRACT

Although immunity induced by typhoid fever is moderated and short-lived, typhoid vaccination with the attenuated Ty21a oral vaccine generates long-lasting protection rates reaching up to 92%. Thus, there are important differences on how wild-type Salmonella and typhoid vaccine strains stimulate host immunity. We hypothesize that vaccine strains with different mutations might affect gut inflammation and intestinal permeability by different mechanisms. To test this hypothesis, we used an in vitro organotypic model of the human intestinal mucosa composed of human intestinal epithelial cells, lymphocytes/monocytes, endothelial cells, and fibroblasts. We also used six Salmonella enterica serovar Typhi (S. Typhi) strains: the licensed Ty21a oral vaccine, four typhoid vaccine candidates (i.e., CVD 908, CVD 909, CVD 910, and CVD 915) and the wild-type Ty2 strain. We found that genetically engineered S. Typhi vaccine strains elicit differential host changes not only in the intestinal permeability and secretion of inflammatory cytokines, but also in the phenotype and activation pathways of innate cells. These changes were distinct from those elicited by the parent wild-type S. Typhi and depended on the genetic manipulation. In sum, these results emphasize the importance of carefully selecting specific manipulations of the Salmonella genome in the development of typhoid vaccines.


Subject(s)
Intestinal Mucosa/immunology , Polysaccharides, Bacterial/immunology , Salmonella typhi/genetics , Typhoid Fever/immunology , Typhoid-Paratyphoid Vaccines/immunology , Antibodies, Bacterial/blood , Cells, Cultured , Cytokines/metabolism , Gene Expression Regulation, Bacterial , Humans , Immunity, Innate , Inflammation Mediators/metabolism , Organ Culture Techniques , Tight Junctions/metabolism , Vaccination , Vaccines, Attenuated
4.
PLoS Negl Trop Dis ; 11(4): e0005493, 2017 04.
Article in English | MEDLINE | ID: mdl-28388624

ABSTRACT

Invasive infections associated with non-typhoidal Salmonella (NTS) serovars Enteritidis (SE), Typhimurium (STm) and monophasic variant 1,4,[5],12:i:- are a major health problem in infants and young children in sub-Saharan Africa, and currently, there are no approved human NTS vaccines. NTS O-polysaccharides and flagellin proteins are protective antigens in animal models of invasive NTS infection. Conjugates of SE core and O-polysaccharide (COPS) chemically linked to SE flagellin have enhanced the anti-COPS immune response and protected mice against fatal challenge with a Malian SE blood isolate. We report herein the development of a STm glycoconjugate vaccine comprised of STm COPS conjugated to the homologous serovar phase 1 flagellin protein (FliC) with assessment of the role of COPS O-acetyls for functional immunity. Sun-type COPS conjugates linked through the polysaccharide reducing end to FliC were more immunogenic and protective in mice challenged with a Malian STm blood isolate than multipoint lattice conjugates (>95% vaccine efficacy [VE] versus 30-43% VE). Immunization with de-O-acetylated STm-COPS conjugated to CRM197 provided significant but reduced protection against STm challenge compared to mice immunized with native STm-COPS:CRM197 (63-74% VE versus 100% VE). Although OPS O-acetyls were highly immunogenic, post-vaccination sera that contained various O-acetyl epitope-specific antibody profiles displayed similar in vitro bactericidal activity when equivalent titers of anti-COPS IgG were assayed. In-silico molecular modeling further indicated that STm OPS forms a single dominant conformation, irrespective of O-acetylation, in which O-acetyls extend outward and are highly solvent exposed. These preclinical results establish important quality attributes for an STm vaccine that could be co-formulated with an SE-COPS:FliC glycoconjugate as a bivalent NTS vaccine for use in sub-Saharan Africa.


Subject(s)
Salmonella Infections/prevention & control , Salmonella Vaccines/therapeutic use , Salmonella typhimurium , Africa South of the Sahara , Animals , Antibodies, Bacterial/blood , Disease Models, Animal , Female , Flagellin/immunology , Flagellin/therapeutic use , Glycoconjugates/immunology , Glycoconjugates/therapeutic use , Humans , Immunoglobulin G/blood , Mice , O Antigens/immunology , O Antigens/therapeutic use , Regression Analysis , Salmonella Infections/immunology , Salmonella Vaccines/immunology , Vaccines, Attenuated/immunology , Vaccines, Attenuated/therapeutic use
5.
EcoSal Plus ; 7(1)2016 11.
Article in English | MEDLINE | ID: mdl-27809955

ABSTRACT

Salmonellosis, caused by members of the genus Salmonella, is responsible for considerable global morbidity and mortality in both animals and humans. In this review, we will discuss the pathogenesis of Salmonella enterica serovar Typhi and Salmonella enterica serovar Typhimurium, focusing on human Salmonella infections. We will trace the path of Salmonella through the body, including host entry sites, tissues and organs affected, and mechanisms involved in both pathogenesis and stimulation of host immunity. Careful consideration of the natural progression of disease provides an important context in which attenuated live oral vaccines can be rationally designed and developed. With this in mind, we will describe a series of attenuated live oral vaccines that have been successfully tested in clinical trials and demonstrated to be both safe and highly immunogenic. The attenuation strategies summarized in this review offer important insights into further development of attenuated vaccines against other Salmonella for which live oral candidates are currently unavailable.


Subject(s)
Salmonella Infections/immunology , Salmonella Infections/microbiology , Salmonella Vaccines/immunology , Salmonella typhi/immunology , Salmonella typhimurium/immunology , Administration, Oral , Animals , Antibodies, Bacterial/biosynthesis , Antibodies, Bacterial/immunology , Disease Models, Animal , Humans , Immunogenicity, Vaccine , Salmonella Infections, Animal , Salmonella typhi/pathogenicity , Salmonella typhimurium/pathogenicity , Serogroup , Vaccination , Vaccines, Attenuated/immunology
6.
Pathog Dis ; 74(8)2016 11.
Article in English | MEDLINE | ID: mdl-27630185

ABSTRACT

Space exploration programs have long been interested in the effects of spaceflight on biology. This research is important not only in its relevance to future deep space exploration, but also because it has allowed investigators to ask questions about how gravity impacts cell behavior here on Earth. In the 1980s, scientists designed and built the first rotating wall vessel, capable of mimicking the low shear environment found in space. This vessel has since been used to investigate growth of both microorganisms and human tissue cells in low shear modeled microgravity conditions. Bacterial behavior has been shown to be altered both in space and under simulated microgravity conditions. In some cases, bacteria appear attenuated, whereas in others virulence is enhanced. This has consequences not only for manned spaceflight, but poses larger questions about the ability of bacteria to sense the world around them. By using the microgravity environment as a tool, we can exploit this phenomenon in the search for new therapeutics and preventatives against pathogenic bacteria for use both in space and on Earth.


Subject(s)
Bacteria , Bacterial Physiological Phenomena , Host-Pathogen Interactions , Weightlessness , Animals , Bacteria/drug effects , Bacteria/immunology , Bacterial Infections/drug therapy , Bacterial Infections/microbiology , Bacterial Infections/prevention & control , Bacterial Physiological Phenomena/drug effects , Bacterial Vaccines/immunology , Drug Discovery , Host-Pathogen Interactions/drug effects , Humans , Research , Space Flight
7.
Clin Vaccine Immunol ; 23(6): 520-523, 2016 06.
Article in English | MEDLINE | ID: mdl-27030587

ABSTRACT

Nontyphoidal Salmonella (NTS) invasive infections are an important cause of morbidity and mortality in sub-Saharan Africa. Several vaccines are in development to prevent these infections. We describe an NTS opsonophagocytic killing assay that uses HL-60 cells and baby rabbit complement to quantify functional antibodies elicited by candidate NTS vaccines.


Subject(s)
Antibodies, Bacterial/immunology , Immunogenicity, Vaccine , Phagocytosis , Salmonella Vaccines/immunology , Salmonella/immunology , Animals , Antibodies, Bacterial/blood , Child, Preschool , Complement System Proteins/immunology , HL-60 Cells , Humans , Immunoglobulin G/blood , Mice , Rabbits , Salmonella Infections/immunology , Salmonella Infections/microbiology , Salmonella Infections/prevention & control , Salmonella Infections, Animal/immunology , Salmonella Infections, Animal/microbiology , Salmonella Infections, Animal/prevention & control , Salmonella typhimurium/immunology
8.
Infect Immun ; 83(12): 4504-12, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26351285

ABSTRACT

Invasive nontyphoidal Salmonella (NTS) infections constitute a major health problem among infants and toddlers in sub-Saharan Africa; these infections also occur in infants and the elderly in developed countries. We genetically engineered a Salmonella enterica serovar Typhimurium strain of multilocus sequence type 313, the predominant genotype circulating in sub-Saharan Africa. We evaluated the capacities of S. Typhimurium and Salmonella enterica serovar Enteritidis ΔguaBA ΔclpX live oral vaccines to protect mice against a highly lethal challenge dose of the homologous serovar and determined protection against other group B and D serovars circulating in sub-Saharan Africa. The vaccines S. Typhimurium CVD 1931 and S. Enteritidis CVD 1944 were immunogenic and protected BALB/c mice against 10,000 50% lethal doses (LD50) of S. Typhimurium or S. Enteritidis, respectively. S. Typhimurium CVD 1931 protected mice against the group B serovar Salmonella enterica serovar Stanleyville (91% vaccine efficacy), and S. Enteritidis CVD 1944 protected mice against the group D serovar Salmonella enterica serovar Dublin (85% vaccine efficacy). High rates of survival were observed when mice were infected 12 weeks postimmunization, indicating that the vaccines elicited long-lived protective immunity. Whereas CVD 1931 did not protect against S. Enteritidis R11, CVD 1944 did mediate protection against S. Typhimurium D65 (81% efficacy). These findings suggest that a bivalent (S. Typhimurium and S. Enteritidis) vaccine would provide broad protection against the majority of invasive NTS infections in sub-Saharan Africa.


Subject(s)
Antibodies, Bacterial/biosynthesis , Antigens, Bacterial/immunology , Salmonella Infections, Animal/prevention & control , Salmonella Vaccines/immunology , Salmonella enteritidis/immunology , Salmonella typhimurium/immunology , Administration, Oral , Africa South of the Sahara , Animals , Antigens, Bacterial/administration & dosage , Cross Protection , Female , Genotype , Humans , Lethal Dose 50 , Mice , Mice, Inbred BALB C , Salmonella Infections, Animal/immunology , Salmonella Infections, Animal/microbiology , Salmonella Infections, Animal/mortality , Salmonella Vaccines/administration & dosage , Salmonella enteritidis/drug effects , Salmonella enteritidis/genetics , Salmonella typhimurium/drug effects , Salmonella typhimurium/genetics , Serotyping , Survival Analysis , Vaccination , Vaccines, Attenuated
9.
Infect Immun ; 83(1): 161-72, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25332120

ABSTRACT

Live attenuated bacteria hold great promise as multivalent mucosal vaccines against a variety of pathogens. A major challenge of this approach has been the successful delivery of sufficient amounts of vaccine antigens to adequately prime the immune system without overattenuating the live vaccine. Here we used a live attenuated Salmonella enterica serovar Typhi strain to create a bivalent mucosal plague vaccine that produces both the protective F1 capsular antigen of Yersinia pestis and the LcrV protein required for secretion of virulence effector proteins. To reduce the metabolic burden associated with the coexpression of F1 and LcrV within the live vector, we balanced expression of both antigens by combining plasmid-based expression of F1 with chromosomal expression of LcrV from three independent loci. The immunogenicity and protective efficacy of this novel vaccine were assessed in mice by using a heterologous prime-boost immunization strategy and compared to those of a conventional strain in which F1 and LcrV were expressed from a single low-copy-number plasmid. The serum antibody responses to lipopolysaccharide (LPS) induced by the optimized bivalent vaccine were indistinguishable from those elicited by the parent strain, suggesting an adequate immunogenic capacity maintained through preservation of bacterial fitness; in contrast, LPS titers were 10-fold lower in mice immunized with the conventional vaccine strain. Importantly, mice receiving the optimized bivalent vaccine were fully protected against lethal pulmonary challenge. These results demonstrate the feasibility of distributing foreign antigen expression across both chromosomal and plasmid locations within a single vaccine organism for induction of protective immunity.


Subject(s)
Drug Carriers , Plague Vaccine/immunology , Plague/prevention & control , Salmonella typhi/genetics , Animals , Antibodies, Bacterial/blood , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Disease Models, Animal , Genetic Vectors , Mice, Inbred BALB C , Plague/immunology , Plague Vaccine/administration & dosage , Plague Vaccine/genetics , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/immunology , Survival Analysis , Vaccination/methods , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
10.
Clin Vaccine Immunol ; 21(5): 712-21, 2014 May.
Article in English | MEDLINE | ID: mdl-24623629

ABSTRACT

Invasive Salmonella infections for which improved or new vaccines are being developed include enteric fever caused by Salmonella enterica serovars Typhi, Paratyphi A, and Paratyphi B and sepsis and meningitis in young children in sub-Saharan Africa caused by nontyphoidal Salmonella (NTS) serovars, particularly S. enterica serovars Typhimurium and Enteritidis. Assays are needed to measure functional antibodies elicited by the new vaccines to assess their immunogenicities and potential protective capacities. We developed in vitro assays to quantify serum bactericidal antibody (SBA) activity induced by S. Typhi, S. Paratyphi A, S. Typhimurium, and S. Enteritidis vaccines in preclinical studies. Complement from various sources was tested in assays designed to measure antibody-dependent complement-mediated killing. Serum from rabbits 3 to 4 weeks of age provided the best complement source compared to serum from pigs, goats, horses, bovine calves, or rabbits 8 to 12 weeks of age. For S. Enteritidis, S. Typhimurium, and S. Typhi SBA assays to be effective, bacteria had to be harvested at log phase. In contrast, S. Paratyphi A was equally susceptible to killing whether it was grown to the stationary or log phase. The typhoidal serovars were more susceptible to complement-mediated killing than were the nontyphoidal serovars. Lastly, the SBA endpoint titers correlated with serum IgG anti-lipopolysaccharide (LPS) titers in mice immunized with mucosally administered S. Typhimurium, S. Enteritidis, and S. Paratyphi A but not S. Typhi live attenuated vaccines. The SBA assay described here is a useful tool for measuring functional antibodies elicited by Salmonella vaccine candidates.


Subject(s)
Antibodies, Bacterial/blood , Blood Bactericidal Activity , Complement System Proteins/immunology , Salmonella Vaccines/immunology , Animals , Humans , Immunoassay/methods , Immunoglobulin G/blood , Lipopolysaccharides/immunology , Meningitis, Bacterial/prevention & control , Salmonella Infections/prevention & control , Salmonella Vaccines/administration & dosage , Sepsis/prevention & control , Typhoid Fever/prevention & control
11.
Vaccine ; 32(35): 4376-4385, 2014 Jul 31.
Article in English | MEDLINE | ID: mdl-24370705

ABSTRACT

Contemporary vaccine development relies less on empirical methods of vaccine construction, and now employs a powerful array of precise engineering strategies to construct immunogenic live vaccines. In this review, we will survey various engineering techniques used to create attenuated vaccines, with an emphasis on recent advances and insights. We will further explore the adaptation of attenuated strains to create multivalent vaccine platforms for immunization against multiple unrelated pathogens. These carrier vaccines are engineered to deliver sufficient levels of protective antigens to appropriate lymphoid inductive sites to elicit both carrier-specific and foreign antigen-specific immunity. Although many of these technologies were originally developed for use in Salmonella vaccines, application of the essential logic of these approaches will be extended to development of other enteric vaccines where possible. A central theme driving our discussion will stress that the ultimate success of an engineered vaccine rests on achieving the proper balance between attenuation and immunogenicity. Achieving this balance will avoid over-activation of inflammatory responses, which results in unacceptable reactogenicity, but will retain sufficient metabolic fitness to enable the live vaccine to reach deep tissue inductive sites and trigger protective immunity. The breadth of examples presented herein will clearly demonstrate that genetic engineering offers the potential for rapidly propelling vaccine development forward into novel applications and therapies which will significantly expand the role of vaccines in public health.


Subject(s)
Salmonella Vaccines/immunology , Drug Discovery/methods , Drug Discovery/trends , Genetic Engineering/methods , Humans , Salmonella Vaccines/genetics , Technology, Pharmaceutical/methods , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
12.
PLoS One ; 8(5): e64680, 2013.
Article in English | MEDLINE | ID: mdl-23741368

ABSTRACT

Non-typhoidal Salmonella (NTS) serovars S. Enteritidis and S. Typhimurium are a major cause of invasive bacterial disease (e.g., bacteremia, meningitis) in infants and young children in sub-Saharan Africa and also occasionally cause invasive disease in highly susceptible hosts (young infants, the elderly, and immunocompromised subjects) in industrialized countries. No licensed vaccines exist against human NTS infections. NTS core and O polysaccharide (COPS) and FliC (Phase 1 flagellin subunits) each constitute protective antigens in murine models. S. Enteritidis COPS conjugated to FliC represents a promising vaccine approach that elicits binding and opsonophagocytic antibodies and protects mice against lethal challenge with virulent S. Enteritidis. We examined the protective efficacy of fractional dosages of S. Enteritidis COPS:FliC conjugate vaccines in mice, and also established that protection can be passively transferred to naïve mice by administering sera from mice immunized with conjugate. Mice were immunized with three doses of either 10 µg, 2.5 µg (full dose), 0.25 µg, or 0.025 µg S. Enteritidis COPS:FliC conjugate at 28 day intervals. Antibody titers to COPS and FliC measured by ELISA fell consonant with progressively smaller vaccine dosage levels; anti-FliC IgG responses remained robust at fractional dosages for which anti-COPS serum IgG titers were decreased. Nevertheless, >90% protection against intraperitoneal challenge was observed in mice immunized with fractional dosages of conjugate that elicited diminished titers to both FliC and COPS. Passive transfer of immune sera from mice immunized with the highest dose of COPS:FliC to naïve mice was also protective, demonstrating the role of antibodies in mediating protection. These results provide important insights regarding the potency of Salmonella glycoconjugate vaccines that use flagellin as a carrier protein.


Subject(s)
Flagellin/immunology , Glycoconjugates/immunology , Immunization, Passive , O Antigens/immunology , Salmonella Infections, Animal/prevention & control , Salmonella Vaccines/immunology , Salmonella enteritidis/immunology , Animals , Antibodies, Bacterial/blood , Dose-Response Relationship, Immunologic , Female , Flagellin/chemistry , Glycoconjugates/administration & dosage , Glycoconjugates/chemistry , Humans , Immune Sera/administration & dosage , Immunoglobulin G/blood , Mice , O Antigens/chemistry , Salmonella Infections, Animal/immunology , Salmonella Infections, Animal/microbiology , Salmonella Vaccines/administration & dosage , Salmonella enteritidis/drug effects , Vaccination , Vaccines, Conjugate
13.
Hum Vaccin Immunother ; 9(7): 1558-64, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23406777

ABSTRACT

Bacterial live vector vaccines represent a vaccine development strategy that offers exceptional flexibility. In this approach, genes encoding protective antigens of unrelated bacterial, viral or parasitic pathogens are expressed in an attenuated bacterial vaccine strain that delivers these foreign antigens to the immune system, thereby eliciting relevant immune responses. Rather than expressing these antigens using low copy expression plasmids, here we pursue expression of foreign proteins from the live vector chromosome. Our strategy is designed to compensate for the inherent disadvantage of loss of gene dosage (vs. plasmid-based expression) by integrating antigen-encoding gene cassettes into multiple chromosomal sites already inactivated in an attenuated Salmonella enterica serovar Typhi vaccine candidate. We tested expression of a cassette encoding the green fluorescent protein (GFPuv) integrated separately into native guaBA, htrA or clyA chromosomal loci. Using single integrations, we show that expression levels of GFPuv are significantly affected by the site of integration, regardless of the inclusion of additional strong promoters within the incoming cassette. Using cassettes integrated into both guaBA and htrA, we observe cumulative synthesis levels from two integration sites superior to single integrations. Most importantly, we observe that GFPuv expression increases in a growth phase-dependent manner, suggesting that foreign antigen synthesis may be "tuned" to the physiology of the live vaccine. We expect this novel platform expression technology to prove invaluable in the development of a wide variety of multivalent live vector vaccines, capable of expressing multiple antigens from both chromosomal and plasmid-based expression systems within a single strain.


Subject(s)
Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacterial Vaccines/immunology , Typhoid-Paratyphoid Vaccines/immunology , Vaccines, Attenuated/immunology , Bacterial Proteins/genetics , Bacterial Vaccines/genetics , Chromosomes, Bacterial/genetics , Gene Expression , Genetic Vectors , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Hemolysin Proteins/genetics , Humans , Promoter Regions, Genetic/genetics , Salmonella typhi/immunology , Typhoid-Paratyphoid Vaccines/genetics , Vaccines, Attenuated/genetics
14.
Comp Immunol Microbiol Infect Dis ; 36(2): 113-28, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23195858

ABSTRACT

Yersinia pestis causes bubonic and pneumonic plague in humans. The pneumonic infection is the most severe and invariably fatal if untreated. Because of its high virulence, ease of delivery and precedent of use in warfare, Y. pestis is considered as a potential bioterror agent. No licensed plague vaccine is currently available in the US. Laboratory research with virulent strains requires appropriate biocontainment (i.e., Biosafety Level 3 (BSL-3) for procedures that generate aerosol/droplets) and secure facilities that comply with federal select agent regulations. To assist in the identification of promising vaccine candidates during the early phases of development, we characterized mouse models of systemic and pneumonic plague infection using the Y. pestis strain EV76, an attenuated human vaccine strain that can be rendered virulent in mice under in vivo iron supplementation. Mice inoculated intranasally or intravenously with Y. pestis EV76 in the presence of iron developed a systemic and pneumonic plague infection that resulted in disease and lethality. Bacteria replicated and severely compromised the spleen, liver and lungs. Susceptibility was age dependent, with younger mice being more vulnerable to pneumonic infection. We used these models of infection to assess the protective capacity of newly developed Salmonella-based plague vaccines. The protective outcome varied depending on the route and dose of infection. Protection was associated with the induction of specific immunological effectors in systemic/mucosal compartments. The models of infection described could serve as safe and practical tools for identifying promising vaccine candidates that warrant further potency evaluation using fully virulent strains in BSL-3 settings.


Subject(s)
Plague/prevention & control , Yersinia pestis/immunology , Administration, Intranasal , Administration, Intravenous , Animals , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Disease Models, Animal , Disease Susceptibility , Female , Immunoglobulin A, Secretory , Immunoglobulin G/blood , Immunoglobulin G/immunology , Lethal Dose 50 , Mice , Plague/mortality , Plague/pathology , Plague Vaccine/administration & dosage , Plague Vaccine/immunology , Plague Vaccine/pharmacokinetics , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/immunology , Recombinant Proteins , Reproducibility of Results , Virulence/genetics , Yersinia pestis/pathogenicity
15.
PLoS Negl Trop Dis ; 5(11): e1373, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22069504

ABSTRACT

BACKGROUND: Antibiotic-resistant Salmonella enterica serovar Paratyphi A, the agent of paratyphoid A fever, poses an emerging public health dilemma in endemic areas of Asia and among travelers, as there is no licensed vaccine. Integral to our efforts to develop a S. Paratyphi A vaccine, we addressed the role of flagella as a potential protective antigen by comparing cell-associated flagella with exported flagellin subunits expressed by attenuated strains. METHODOLOGY: S. Paratyphi A strain ATCC 9150 was first deleted for the chromosomal guaBA locus, creating CVD 1901. Further chromosomal deletions in fliD (CVD 1901D) or flgK (CVD 1901K) were then engineered, resulting in the export of unpolymerized FliC, without impairing its overall expression. The virulence of the resulting isogenic strains was examined using a novel mouse LD(50) model to accommodate the human-host restricted S. Paratyphi A. The immunogenicity of the attenuated strains was then tested using a mouse intranasal model, followed by intraperitoneal challenge with wildtype ATCC 9150. RESULTS: Mucosal (intranasal) immunization of mice with strain CVD 1901 expressing cell-associated flagella conferred superior protection (vaccine efficacy [VE], 90%) against a lethal intraperitoneal challenge, compared with the flagellin monomer-exporting mutants CVD 1901K (30% VE) or CVD 1901D (47% VE). The superior protection induced by CVD 1901 with its cell-attached flagella was associated with an increased IgG2a:IgG1 ratio of FliC-specific antibodies with enhanced opsonophagocytic capacity. CONCLUSIONS: Our results clearly suggest that enhanced anti-FliC antibody-mediated clearance of S. Paratyphi A by phagocytic cells, induced by vaccines expressing cell-associated rather than exported FliC, might be contributing to the vaccine-induced protection from S. Paratyphi A challenge in vivo. We speculate that an excess of IgG1 anti-FliC antibodies induced by the exported FliC may compete with the IgG2a subtype and block binding to specific phagocyte Fc receptors that are critical for clearing an S. Paratyphi A infection.


Subject(s)
Flagella/immunology , Paratyphoid Fever/prevention & control , Salmonella Vaccines/immunology , Salmonella paratyphi A/immunology , Animals , Antibodies, Bacterial/blood , Disease Models, Animal , Female , Flagella/genetics , Gene Deletion , Immunoglobulin G/blood , Lethal Dose 50 , Mice , Mice, Inbred BALB C , Opsonin Proteins/blood , Paratyphoid Fever/immunology , Phagocytosis , Salmonella Vaccines/administration & dosage , Salmonella Vaccines/genetics , Salmonella paratyphi A/genetics , Survival Analysis , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Virulence
16.
Expert Rev Vaccines ; 10(12): 1679-82, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22085171

ABSTRACT

The construction of safe and protective vaccines, derived from human pathogens that have been genetically modified to remove pathogenicity, is often easier to accomplish on paper than it is in the laboratory. Kong and colleagues have pursued a clever strategy to reduce the reactogenicity of attenuated Salmonella oral vaccines by genetically modifying the surface lipopolysaccharide to lower endotoxic activity. The resulting candidate vaccine strains were highly reduced in virulence yet were able to confer protection in a mouse model against challenge with virulent Salmonella. Remarkably, these strains could also be further modified to present foreign antigens from unrelated human pathogens and again confer protection against heterologous challenge. This work brings important new tools to bear on solving the problem of creating efficacious attenuated bacterial vaccines that maximize both safety and immunogenicity in clinical trials.

17.
Infect Immun ; 79(10): 4240-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21807909

ABSTRACT

Nontyphoidal Salmonella enterica serovars Enteritidis and Typhimurium are a common cause of gastroenteritis but also cause invasive infections and enteric fever in certain hosts (young children in sub-Saharan Africa, the elderly, and immunocompromised individuals). Salmonella O polysaccharides (OPS) and flagellar proteins are virulence factors and protective antigens. The surface polysaccharides of Salmonella are poorly immunogenic and do not confer immunologic memory, limitations overcome by covalently attaching them to carrier proteins. We conjugated core polysaccharide-OPS (COPS) of Salmonella Enteritidis lipopolysaccharide (LPS) to flagellin protein from the homologous strain. COPS and flagellin were purified from a genetically attenuated (ΔguaBA) "reagent strain" (derived from an isolate from a patient with clinical bacteremia) engineered for increased flagellin production (ΔclpPX). Conjugates were constructed by linking flagellin monomers or polymers at random COPS hydroxyls with various polysaccharide/protein ratios by 1-cyano-4-dimethylaminopyridinium tetrafluoroborate (CDAP) or at the 3-deoxy-d-manno-octulosonic acid (KDO) terminus by thioether chemistry. Mice immunized on days 0, 28, and 56 with COPS-flagellin conjugates mounted higher anti-LPS IgG levels than mice receiving unconjugated COPS and exhibited high antiflagellin IgG; anti-LPS and antiflagellin IgG levels increased following booster doses. Antibodies generated by COPS-flagellin conjugates mediated opsonophagocytosis of S. Enteritidis cells into mouse macrophages. Mice immunized with flagellin alone, COPS-CRM197, or COPS-flagellin conjugates were significantly protected from lethal challenge with wild-type S. Enteritidis (80 to 100% vaccine efficacy).


Subject(s)
Antibodies, Bacterial/blood , Flagellin/immunology , Immunoconjugates/administration & dosage , Macrophages/microbiology , O Antigens/immunology , Salmonella Infections, Animal/prevention & control , Salmonella Vaccines/administration & dosage , Salmonella enteritidis/immunology , Animals , Animals, Outbred Strains , Cell Line , Female , Flagellin/chemistry , Humans , Immunization , Immunoconjugates/immunology , Immunoglobulin G/blood , Mice , Mice, Inbred BALB C , O Antigens/chemistry , Opsonin Proteins/metabolism , Phagocytosis , Salmonella Infections, Animal/immunology , Salmonella Infections, Animal/microbiology , Salmonella Vaccines/immunology , Salmonella enteritidis/pathogenicity
18.
Infect Immun ; 79(10): 4175-85, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21807911

ABSTRACT

While nontyphoidal Salmonella (NTS) has long been recognized as a cause of self-limited gastroenteritis, it is becoming increasingly evident that multiple-antibiotic-resistant strains are also emerging as important causes of invasive bacteremia and focal infections, resulting in hospitalizations and deaths. We have constructed attenuated Salmonella enterica serovar Typhimurium and Salmonella enterica serovar Enteritidis strains that can serve as live oral vaccines and as "reagent strains" for subunit vaccine production in a safe and economical manner. Prototype attenuated vaccine strains CVD 1921 and CVD 1941, derived from the invasive wild-type strains S. Typhimurium I77 and S. Enteritidis R11, respectively, were constructed by deleting guaBA, encoding guanine biosynthesis, and clpP, encoding a master protease regulator. The clpP mutation resulted in a hyperflagellation phenotype. An additional deletion in fliD yielded reagent strains CVD 1923 and CVD 1943, respectively, which export flagellin monomers. Oral 50% lethal dose (LD50) analyses showed that the NTS vaccine strains were all highly attenuated in mice. Oral immunization with CVD 1921 or CVD 1923 protected mice against lethal challenge with wild-type S. Typhimurium I77. Immunization with CVD 1941 but not CVD 1943 protected mice against lethal infection with S. Enteritidis R11. Immune responses induced by these strains included high levels of serum IgG anti-lipopolysaccharide (LPS) and anti-flagellum antibodies, with titers increasing progressively during the immunization schedule. Since S. Typhimurium and S. Enteritidis are the most common NTS serovars associated with invasive disease, these findings can pave the way for development of a highly effective, broad-spectrum vaccine against invasive NTS.


Subject(s)
Antibodies, Bacterial/blood , Genetic Engineering/methods , Salmonella Infections, Animal/prevention & control , Salmonella Vaccines/administration & dosage , Salmonella enteritidis/immunology , Salmonella typhimurium/immunology , Vaccines, Attenuated/administration & dosage , Administration, Oral , Animals , Bacterial Proteins/genetics , Female , Guanine/metabolism , Immunization Schedule , Immunoglobulin G/blood , Lipopolysaccharides/immunology , Mice , Mice, Inbred BALB C , Salmonella Infections, Animal/immunology , Salmonella Infections, Animal/microbiology , Salmonella Vaccines/genetics , Salmonella Vaccines/immunology , Salmonella enteritidis/genetics , Salmonella enteritidis/physiology , Salmonella typhimurium/genetics , Salmonella typhimurium/physiology , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology
19.
Vaccine ; 29(32): 5094-106, 2011 Jul 18.
Article in English | MEDLINE | ID: mdl-21616112

ABSTRACT

Non-typhoidal Salmonella enterica (NTS) serovars Typhimurium and Enteritidis are important causes of bacterial gastroenteritis in the USA and worldwide. In sub-Saharan Africa these two serovars are emerging as agents associated with lethal invasive disease (e.g., bacteremia, meningitis). The development of NTS vaccines, based on mucosally administered live attenuated strains and parenteral non-living antigens, could diminish the NTS disease burden globally. Mouse models of S. Typhimurium and S. Enteritidis invasive disease can accelerate the development of NTS vaccines. Live attenuated NTS vaccines elicit both cellular and humoral immunity in mice and their efficacy is well established. In contrast, non-living vaccines that primarily elicit humoral immunity have demonstrated variable efficacy. An analysis of the reported studies with non-living vaccines against S. Typhimurium and S. Enteritidis reveals that efficacy is influenced by two important independent variables: (1) the innate susceptibility to NTS infection that differs dramatically between commonly used mouse strains and (2) the virulence of the NTS strain used for challenge. Protection by non-living vaccines has generally been seen only in host-pathogen interactions where a sub-lethal infection results, such as challenging resistant mice with either highly virulent or weakly virulent strains or susceptible mice with weakly virulent strains. The immunologic basis of this discrepancy and the implications for human NTS vaccine development are reviewed herein.


Subject(s)
Immunity, Innate , Salmonella Infections/immunology , Salmonella Infections/prevention & control , Salmonella Vaccines/immunology , Salmonella enteritidis/immunology , Salmonella typhimurium/immunology , Animals , Disease Models, Animal , Host-Pathogen Interactions , Mice , Salmonella Vaccines/administration & dosage
20.
PLoS One ; 6(4): e18700, 2011 Apr 08.
Article in English | MEDLINE | ID: mdl-21494634

ABSTRACT

Certain serovars of Salmonella enterica subsp. enterica cause invasive disease (e.g., enteric fever, bacteremia, septicemia, meningitis, etc.) in humans and constitute a global public health problem. A rapid, sensitive diagnostic test is needed to allow prompt initiation of therapy in individual patients and for measuring disease burden at the population level. An innovative and promising new rapid diagnostic technique is microwave-accelerated metal-enhanced fluorescence (MAMEF). We have adapted this assay platform to detect the chromosomal oriC locus common to all Salmonella enterica subsp. enterica serovars. We have shown efficient lysis of biologically relevant concentrations of Salmonella spp. suspended in bacteriological media using microwave-induced lysis. Following lysis and DNA release, as little as 1 CFU of Salmonella in 1 ml of medium can be detected in <30 seconds. Furthermore the assay is sensitive and specific: it can detect oriC from Salmonella serovars Typhi, Paratyphi A, Paratyphi B, Paratyphi C, Typhimurium, Enteritidis and Choleraesuis but does not detect Escherichia coli, Pseudomonas aeruginosa, Klebsiella pneumoniae, Streptococcus pneumoniae, Haemophilus influenzae or Acinetobacter baumanii. We have also performed preliminary experiments using a synthetic Salmonella oriC oligonucleotide suspended in whole human blood and observed rapid detection when the sample was diluted 1:1 with PBS. These pre-clinical data encourage progress to the next step to detect Salmonella in blood (and other ordinarily sterile, clinically relevant body fluids).


Subject(s)
Diagnostic Tests, Routine/methods , Metals/chemistry , Microwaves , Salmonella/isolation & purification , Typhoid Fever/microbiology , Bacteriolysis , Base Sequence , Biological Assay , DNA, Bacterial/analysis , DNA, Bacterial/blood , Fluorescence , Humans , Molecular Sequence Data , Oligonucleotides/genetics , Oligonucleotides/metabolism , Salmonella/genetics , Sensitivity and Specificity , Typhoid Fever/diagnosis
SELECTION OF CITATIONS
SEARCH DETAIL