Abstract
Salmonellosis remains an important cause of human disease worldwide. While there are several licensed vaccines for Salmonella enterica serovar Typhi, these vaccines are generally ineffective against other Salmonella serovars. Vaccines that target paratyphoid and nontyphoidal Salmonella serovars are very much in need. Preclinical evaluation of candidate vaccines is highly dependent on the availability of appropriate scientific tools, particularly animal models. Many different animal models exist for various Salmonella serovars, from whole-animal models to smaller models, such as those recently established in insects. Here, we discuss various mouse, rat, rabbit, calf, primate, and insect models for Salmonella infection, all of which have their place in research. However, choosing the right model is imperative in selecting the best vaccine candidates for further clinical testing. In this minireview, we summarize the various animal models that are used to assess salmonellosis, highlight some of the advantages and disadvantages of each, and discuss their value in vaccine development.
INTRODUCTION
Animal models are indispensable tools for assessing candidate vaccines, with preclinical animal safety, immunogenicity, and efficacy data being prerequisite for regulatory agencies such as the U.S. Food and Drug Administration (FDA) prior to undertaking early-stage clinical trials. However, not all animal models are created equal. While some models are highly robust and closely mimic clinical infection, others are contrived and far removed from clinical relevance. Choosing the right animal model for the vaccine under investigation is imperative in determining its safety and/or effectiveness.
Salmonella spp. are often used as model organisms to study bacterial pathogenesis and host-microbe interactions, due to the ability of certain Salmonella serovars to readily infect animals. As such, there are a myriad of animal models available to vaccine developers. These range from colonization or lethality models to those involving complex surgical techniques (Table 1). The choice of model is often related not only to relevance but also cost, ethics, housing requirements, and the availability of appropriate technical expertise. While most researchers utilize one model or another, integration of data from multiple animal models can provide a more complete understanding of the safety of a candidate vaccine and/or predict how a potential vaccine may perform in humans. In this minireview, we provide background on Salmonella clinical syndromes and pathogenesis and then summarize the various animal models that have been used for Salmonella vaccine research. We include a brief discussion of the technical aspects, advantages, and limitations of each model, followed by a review of the literature surrounding its use in vaccine evaluation. While there has been considerable development for veterinary Salmonella vaccines, here we will focus solely on vaccines and models for human salmonellosis.
TABLE 1.
Model | Salmonella serovar(s) | Disease modeled | Symptoms and readouts | Vaccine safety and efficacy testing? | Reference(s) |
---|---|---|---|---|---|
Immunocompetent mouse | Typhimurium, Enteritidis, Dublin, Newport, Choleraesuis, Typhi, Paratyphi A, Paratyphi B | Invasive salmonellosis | Fever, malaise, wt loss, systemic spread to organs, death | Yes | Summarized in reference 36 |
Immunodeficient mouse | Typhimurium | Invasive salmonellosis | Fever, malaise, wt loss, systemic spread to organs, death | Yes | 56, 62–65 |
Humanized mouse | Typhi | Typhoid fever | Colonization, systemic spread to organs, persistence | No | 70, 71 |
Opium- treated guinea pig | Typhimurium | Invasive salmonellosis | Intestinal pathology, systemic spread to organs, death | No | 73, 74 |
Rat | Typhimurium, Enteritidis | Invasive salmonellosis | Weight loss, malaise, systemic spread to organs, death | Yes | 75 |
Rabbit | Enteritidis, Paratyphi A | Invasive salmonellosis | Systemic spread, fever, wt loss, death | No | 81, 82 |
Streptomycin-treated mouse | Typhimurium, Enteritidis, Dublin, Pullorum | Gastroenteritis | Colonization, systemic spread to organs, intestinal pathology | Yes | 143 |
Suckling mouse | Typhimurium | Gastroenteritis | Fluid accumulation | No | 90 |
Calf | Typhimurium, Dublin | Gastroenteritis | Fever, wt loss, diarrhea, intestinal pathology, death | Yes | 101 |
Ileal loop (calf, rabbit, pig, primate) | Typhimurium, Dublin | Gastroenteritis | Fluid accumulation, intestinal pathology | No | 15, 106–108 |
Chimpanzee | Typhi | Typhoid fever | Fever, malaise, wt loss, diarrhea, intestinal pathology | Yes | 117, 118 |
Rhesus macaque | Typhimurium | Gastroenteritis | Fever, malaise, wt loss, diarrhea, intestinal pathology | Yes | 98, 115, 116 |
C. elegans | Typhimurium, Enteritidis, Dublin | Invasive salmonellosis | Persistence, death | No | 124–126 |
G. mellonella | Typhimurium | Invasive salmonellosis | Death | No | 130 |
Zebrafish | Typhimurium | Invasive salmonellosis | Persistence, death | No | 132 |
SALMONELLA CLINICAL SYNDROMES
Salmonella enterica subsp. enterica is a Gram-negative intracellular bacterium that is the etiological agent of two clinical syndromes: enteric fever and gastroenteritis. Enteric fever is caused by three serovars, Typhi, Paratyphi A, and Paratyphi B. The clinical syndrome is characterized by fever, coughing, nausea, vomiting, and headache (1). Patients may also suffer from enlargement of the spleen and liver and bradycardia. Typhoid fever most commonly affects children, young adults up to 25 years of age, and the elderly (2). Nontyphoidal Salmonella (NTS), such as Salmonella enterica serovar Typhimurium and Salmonella enterica serovar Enteritidis, typically cause gastroenteritis in healthy human adults. Symptoms manifest as fever, abdominal cramping, and diarrhea (3). In the United States and other developed countries, NTS is commonly associated with large foodborne outbreaks (4). In Africa, NTS is highly associated with invasive disease and causes septicemia in young infants and HIV-infected adults and children (5, 6).
SALMONELLA PATHOGENESIS
Salmonella spp. are ingested through contaminated food and water. While typhoidal Salmonella serovars are human host restricted, NTS serovars have a broad host tropism and can colonize many animals, including chickens, pigs, cows, reptiles, and household pets (7). The bacterium possesses effective acid tolerance mechanisms and upon ingestion will survive passage through the low-pH conditions of the stomach. In the small intestine, the bacterium adheres to and invades the intestinal epithelial cells. The bacterium can then be transported through the mucosa, largely via microfold (M) cells, to access the submucosa and underlying lymphoid tissue (8). Macrophages within the lymphoid tissue engulf Salmonella cells but are unable to kill them due to the ability of the bacteria to interfere with phagosome-lysosome fusion (9). Salmonella then resides and proliferates in these immune cells (10). Invasive infection proceeds by dissemination from the intestinal mucosa, causing bacteremia and growth in distant organs, such as the spleen, liver, and gallbladder (1). Noninvasive NTS infections remain localized to the gastrointestinal tract, causing inflammation of the mucosa and secretory diarrhea (3, 11).
The pathogenic elements used by Salmonella to establish and propagate infection are relatively well known and are found in large part on Salmonella pathogenicity islands (SPIs). Twenty-three SPIs have been described to date (12), with some being found universally in Salmonella and others being associated with a subset of strains. Each SPI is associated with certain elements of the bacterial pathogenic process, a subset of which are described below. The ability of Salmonella to manipulate host cell function is accomplished by expression of type III secretion systems (TTSS), which are capable of injecting bacterial proteins directly into the host cell cytoplasm. SPI-1 encodes a TTSS that enables invasion of epithelial cells. Once the bacteria have been transported through the mucosa and are localized within macrophages, they employ the TTSS on SPI-2 to evade lysosomal fusion and set up residence in a specialized niche termed the Salmonella-containing vacuole (SCV). These two pathogenicity islands, therefore, encode two of the major virulence properties of these bacteria, providing the capacity for invasion and persistence. The less highly studied SPI-3 and SPI-4 are thought to be involved in intramacrophage survival, although both encode several additional genes of unknown function (13, 14). SPI-5 is involved in gastroenteritis-associated phenotypes, as it mediates an increase in intestinal fluid secretion (15). Finally, SPI-7, which is found in Salmonella Typhi (among others), encodes the Vi polysaccharide, which has been used in several different Salmonella Typhi vaccine formulations (16).
SALMONELLA VACCINES
There are currently three types of licensed vaccines for Salmonella Typhi, but none for the other serovars (17). The oldest of these vaccines is live attenuated Salmonella Typhi Ty21a, which was constructed by chemical mutagenesis and is unable to synthesize galactose due to a mutation in the galE gene (18). The vaccine is currently formulated as enteric-coated capsules which are taken orally on alternate days until three (in most world regions) or four (in the United States) doses have been received. This vaccine is highly efficacious against typhoid fever and shows some cross-protection against Salmonella Paratyphi B, but not against Salmonella Paratyphi A (19, 20). There are also two vaccines that target the Vi capsule polysaccharide: a polysaccharide-only vaccine and a conjugate vaccine. However, these are only effective against Salmonella Typhi and show no efficacy against other Salmonella serovars. As such, there is a need for additional vaccines to protect against paratyphoidal and nontyphoidal Salmonella infection.
The prevalence of Salmonella Paratyphi A has recently spiked in countries where it is endemic (21, 22), which has increased the interest in a bivalent typhoid-paratyphoid vaccine. Only one Salmonella Paratyphi A vaccine, CVD 1902, has been evaluated in a phase 1 clinical trial (registered at ClinicalTrials.gov [https://clinicaltrials.gov] under registration number NCT01129453) (K. Kotloff, personal communication), and it was found to be safe. Several conjugate vaccine approaches are also under investigation in preclinical studies (23–25).
With regard to nontyphoidal Salmonella, the high incidence of invasive disease occurring in sub-Saharan Africa (approximately 227 cases per 100,000 children) (26), as well as the high case fatality rate of 20 to 25% (5, 27), have spurred a number of vaccine approaches targeting these organisms. These have included novel live attenuated vaccine strains, conjugate vaccines, protein subunit vaccines, and the more recent generalized modules for membrane antigens (GMMA) approach (25, 28, 29). There is also renewed interest in a vaccine that can prevent Salmonella gastroenteritis in developed countries. While morbidity and mortality are generally low for these infections, the economic burden of salmonellosis in the United States is estimated to be $3.3 billion annually (30), making a Salmonella gastroenteritis vaccine economically viable. In addition, elderly patients are known to be especially susceptible to Salmonella gastroenteritis (31). Those in long-term-care facilities are particularly vulnerable, with a case-fatality rate up to 70 times greater than the general population (32, 33). As such, a vaccine that could protect this population specifically is also desirable. All of the Salmonella vaccines currently in development will need to be evaluated for safety and immunogenicity in preclinical studies. The animal models that have been used to date and/or are available for future vaccines are described below.
SYSTEMIC INFECTION SMALL ANIMAL MODELS
(i) Immunocompetent mouse models.
The mouse model of invasive salmonellosis has been employed as the standard in the Salmonella pathogenesis and vaccine development fields for decades. There are many published reviews on these models, which have listed in great detail the experimental conditions, advantages, and caveats of the various models in use (34–37). For this reason, we will touch only briefly on systemic infection mouse models in this review.
Salmonella Typhimurium (and several other serovars) can infect mice, causing a systemic disease that resembles typhoid fever. Although many Salmonella Typhimurium strains are virulent in mice, the disease in mice (systemic) is vastly different from the clinical syndrome these strains generally produce in humans (gastroenteric). As such, Salmonella Typhimurium has traditionally been used to model human Salmonella Typhi infections, but it cannot be used to model human NTS gastroenteritis. However, using Salmonella Typhimurium in mice as a model for Salmonella Typhi is itself imperfect, as the two strains have diverged genetically and have dissimilar pathogenic processes (38). Regardless, these mouse models have been instrumental in increasing our understanding of Salmonella pathogenesis and the host immune response.
There are two basic archetypes of systemic infection mouse models for Salmonella. One uses mice that are intrinsically susceptible to Salmonella infections, and the other uses mice that are resistant. Susceptibility is bestowed by mutations in genes that are important for innate or acquired immunity to intracellular pathogens. These mutations include those in the divalent cation transporter Nramp, Toll-like receptor 4, and others. Among these susceptible strains, Nramp1-deficient strains (Nramp1S; e.g., BALB/c, C57BL/6) have been used extensively to elucidate Salmonella pathogenesis. The Nramp1 transporter is an intracellular protein that is recruited to the endosome, where it functions as a transporter for divalent cations, including Fe2+ and Mn2+ (39). There are several ways this transporter is thought to impact Salmonella survival, including denying the bacterium access to iron and activating inflammatory responses such as reactive oxygen species (ROS) and reactive nitrogen species (RNS) (40–42). For susceptible (Nramp1S) mice, infection via the orogastric route leads to a systemic infection and is ultimately lethal in several days or up to 2 weeks (depending on the dose). These mice succumb following parenteral infection with very low doses (<10 CFU) of highly virulent strains. Similar results can be produced with resistant (Nramp1R) mice by using intraperitoneal or intravenous infection with moderate bacterial doses (e.g., 105 CFU), or via oral infection with very high bacterial doses (e.g., 108 CFU). In either case, infection leads to a systemic spread of the bacteria that is similar in clinical presentation to typhoid in humans (35). There are pros and cons to both the susceptible and resistant mouse models, as reviewed by Simon et al. (36). When the route of infection is important, susceptible mice may be preferred, as oral infection is the natural route for Salmonella infection. However, some resistant mouse strains, such as CD-1, have the advantage of being outbred stocks, which is useful for evaluating candidate vaccines, as the bias of major histocompatibility complex (MHC) type is diminished relative to that in inbred strains.
While typhoid pathogenesis and immunity can be investigated in the mouse by using Salmonella Typhimurium, for vaccine safety and efficacy studies experiments need to be performed with the ultimate vaccine strain. Salmonella Typhi and Salmonella Paratyphi A are human host restricted and are normally asymptomatic in mice. However, a lethal infection can be produced in mice by suspending the bacteria in 5 to 10% (wt/vol) hog gastric mucin and then injecting the suspension intraperitoneally (43–46). In immunization studies, dosing mice with live attenuated Salmonella Typhi vaccines was ineffective by the orogastric route but highly immunogenic when vaccines were given intranasally (47). By utilizing this combination of intranasal immunization and hog gastric mucin challenge, researchers have assessed the safety and efficacy of several live attenuated vaccines for Salmonella Typhi and Salmonella Paratyphi A. This includes Salmonella Typhi CVD 908, Salmonella Typhi CVD 908-htrA, and Salmonella Paratyphi A CVD 1902, which have been shown to be well tolerated and immunogenic in clinical trials (48–50) (K. Kotloff, personal communication) (clinical trial registration number NCT01129453 [https://clinicaltrials.gov]). However, while a Salmonella Typhi ΔphoP mutant was shown to be virulent in the hog gastric mucin model (51), this same mutation (Salmonella Typhi Ty800) was found to be attenuating and safe in a human clinical trial (52).
Although the systemic mouse model does not accurately recapitulate disease caused by either typhoidal or nontyphoidal Salmonella serovars, it has a number of advantages. First, the model is very robust, and the high lethality of the infection provides a strong endpoint from which vaccine efficacy can be calculated. The ease of working with mice also allows experiments to be performed quickly and with large sample sizes, increasing the statistical power of the studies. This allows vaccine researchers to quickly identify lead vaccine candidates and assess dosage, immunization schedule, and the requirement for adjuvants. Finally, these early experiments in mice may allow researchers to characterize correlates of protection, allowing them to establish whether the vaccine could be effective in humans. Many Salmonella vaccine candidates have been tested in this model, including whole killed, live attenuated, subunit, conjugate, and Salmonella live vector vaccines (28, 36, 53–55).
(ii) Immunodeficient mouse models.
Immunocompromised mice have been extensively used in Salmonella pathogenesis studies to investigate the relative contributions of different elements of the immune system to bacterial clearance (56–61). In vaccine research, these models have been used both to test the safety of live attenuated vaccines and to determine the ability of vaccines to elicit a response in immunodeficient hosts (56, 62–65).
Immunodeficiency is of particular concern for the development of live attenuated vaccines, as Salmonella vaccines are expected to be used in populations where coinfections or underlying pathologies affect immune status, and immunocompromised individuals may be susceptible to even significantly weakened vaccine strains. To address this, Salmonella mutants that were attenuated both in wild-type and ROS-deficient (gp91−/− phox) mice were identified (66). One attenuating mutation, ΔcysE, caused a delayed presentation of symptoms in immunodeficient mice compared to ΔaroC mutants yet was also protective (86 to 97% vaccine efficacy) in vaccination-challenge studies with wild-type mice. By incorporating mutations identified in this manner, researchers can be more confident in the safety of live attenuated vaccines in diverse populations.
One additional model that must be mentioned is the TLR11−/− mouse model. Mathur et al. reported in 2012 that TLR11-deficient mice (tlr11−/−) are highly susceptible to infection with Salmonella Typhi (67). Knockout mice infected perorally with 5 × 108 CFU of Salmonella Typhi Ty2 developed a systemic illness similar to human typhoid. Those authors also showed that tlr11−/− mice could be protected from lethal challenge by intraperitoneal immunization of a heat-killed Salmonella Typhi vaccine. While this model would be a great step forward for Salmonella Typhi vaccine testing, issues have arisen surrounding its reproducibility. It has since been reported that five separate groups at four institutions have been unable to replicate these results (68). While standing by their results, the original authors have also noted high variability in the susceptibility of the tlr11−/− mice over time (69). Thus, whether this represents a useful model for Salmonella Typhi vaccine testing is yet to be established.
(iii) Humanized mouse models.
Looking to the future, humanized mouse strains may become a useful tool for better predicting immune responses to Salmonella vaccines in humans, making the determination of vaccine efficacy and immune correlates simpler and more relevant. Humanized mouse models have recently been developed for Salmonella Typhi but may also be applicable to other Salmonella serovars. In these mice, human hematopoietic stem cells are introduced into an immunodeficient mouse background (70–72). This is sufficient to make the mice susceptible to Salmonella Typhi, permitting dissemination of bacteria throughout the spleen, liver, and gallbladder. Analysis of the immune response to infection identified upregulation of a number of human cytokines, in addition to the intrinsic mouse immune response, indicating that the introduced cells are able to respond to infection. This could be advantageous to vaccinologists who are looking to predict the human immune response to vaccination.
(iv) Other small animal models.
In addition to the mouse model, there are also guinea pig, rat, and rabbit models of systemic Salmonella infection. The guinea pig model is similar to the mouse model in that animals succumb to a systemic illness within approximately 3 days. However, this model also induces intestinal inflammation and pathology, including blunting of the villi and disruption of the epithelial cell brush border (73, 74). Guinea pigs can be made susceptible to oral infection with Salmonella Typhimurium via the administration of opium following infection. While this model was important for initial studies of Salmonella pathogenesis, it is no longer widely used.
The rat model, on the other hand, is still in limited use. This model shares many parallels with the mouse model, including the systemic spread of bacteria, although death is rare unless overwhelming challenge doses are given (75, 76). As with mice, the rat strain that is used affects the outcome of an infectious challenge, with Hooded-Lister rats found to be particularly susceptible to systemic spread (75). Little work has been performed to characterize the contribution of bacterial virulence determinants to pathogenesis in either the guinea pig or rat models, although it is known that flagellum and fimbria genes are not implicitly required for virulence of Salmonella Enteritidis in rats (75). The rat model has been used to determine immune responses to Salmonella vector vaccines (77–79) and has in one case been used to establish immunogenicity and protection of a Salmonella Enteritidis ghost particle vaccine (80).
Two rabbit models have been described for Salmonella: one using peroral and the other intraperitoneal infection. The peroral model has been used to evaluate Salmonella Paratyphi A vaccine candidates. Four vaccine strains carrying mutations in phoPQ were assessed for reactogenicity and immunogenicity in orally immunized New Zealand White rabbits (81). One strain, MGN10028, was particularly well tolerated, even at very high doses (2 × 1010 CFU). Despite being less immunogenic than the other three (more reactogenic) strains, rabbits given two doses of this vaccine strain were protected from the most severe symptoms of challenge, including lethargy, anorexia, and decreased water intake.
More recently, an intraperitoneal rabbit model was established for nontyphoidal Salmonella (82). Animals were infected intraperitoneally with high doses (1012 to 1013 CFU) of Salmonella Enteritidis strain CVD J73 and monitored daily for signs of illness, including weight loss, fever, lethargy, and dehydration. There was a 100% attack rate in infected animals, with all showing some clinical signs of disease. Bacteria were also found in distant organs, such as the liver, spleen, heart, lungs, and kidneys. This model may be used in the future to model invasive NTS infections in vaccine studies.
GASTROENTERITIS MODELS
Unlike the systemic models described above, the following models are designed to mimic a natural human infection with NTS. The focus for these models is therefore on gastrointestinal symptoms, including diarrhea, dehydration, and intestinal inflammation and pathology.
(i) Streptomycin mouse model.
To model gastroenteritis in mice, animals are pretreated with streptomycin to deplete the normal flora prior to orogastric infection (83). Several different serovars have been investigated using this model, including serovars Typhimurium, Enteritidis, Dublin, Gallinarum, and Pullorum (84). The depletion of the normal flora allows Salmonella to colonize the cecum and colon, where it grows rapidly to high density. Salmonella then invades the intestinal mucosa, causing localized inflammation, before spreading systemically. In addition to gut pathology, there is some evidence of increased water content in the stool. The response of the intestinal mucosa to infection in this model is similar to the bovine ileal loop and primate orogastric infection models (85). For these reasons, this model has been used to investigate the contribution of different mutations to disease in both the bacterium and the host. While this model is simple and easily implementable, it has some drawbacks, specifically in the relatively limited level of fluid accumulation induced in mice. This makes it difficult to investigate fluid secretion phenotypes in this model. Thus, this model is an excellent tool for investigation of colonization and mucosal inflammation but may not be suitable for evaluation of fluid secretion and other gastroenteritis phenotypes.
The streptomycin mouse model has been used to test safety and efficacy of several live attenuated vaccines (86–88). In one example, researchers examined the efficacy of a Salmonella Typhimurium strain carrying a deletion of the zinc transport operon, znuABC. This vaccine strain had previously been shown to be safe and efficacious in the systemic mouse model (89). Mice immunized orally with 2 × 107 CFU of attenuated strain SA186 were shown to be protected from homologous challenge with virulent Salmonella Typhimurium post-streptomycin treatment (87). In addition to decreased mortality, immunized animals also showed decreased bacterial burden in the spleen and decreased bacterial burden and inflammation in the cecum.
An additional advantage of this model is that it can be performed using immunodeficient mice. In two separate studies, safety of Salmonella Typhimurium vaccine candidates was examined in streptomycin-pretreated mice carrying mutations affecting interferon gamma signaling (ifng1−/−), complement activity (C3−/−), tumor necrosis factor receptor signaling (tnfr−/−), caspase-1 signaling (casp1−/−), B and T cells (rag1−/−), interleukin-10 signaling (IL-10−/−), T cell costimulation (CD40L−/−), and nitric oxide synthase (cybb−/− nos2−/−) (86, 88). While the two vaccine candidates were sufficiently attenuated in some immunodeficient mouse strains (cybb−/− nos2−/−, tnfr−/−), they were still virulent in highly immunodeficient backgrounds, such as rag1−/− and ifng−/−.
(ii) Suckling mouse model.
Complementary to the streptomycin pretreatment approach, the infant mouse model has been used to specifically address fluid accumulation phenotypes for Salmonella mutant strains (90). In this model, suckling mice are infected intragastrically with Salmonella at a dose of up to 5 × 107 CFU. At 2.5 hours postinfection, the alimentary canals are removed and weighed to determine fluid accumulation. Results are presented as the ratio of alimentary weight to total body weight. Compared to mock-infected mice, mice receiving wild-type Salmonella had significantly higher alimentary weight ratios. In contrast, Salmonella pef (fimbrial) mutants were attenuated in their ability to cause fluid accumulation. These results were consistent with those from a mouse ileal loop experiment, suggesting that this model serves as an effective proxy for intestinal fluid accumulation caused by Salmonella. Due to the young age of the mice, this model is not appropriate for vaccine efficacy testing, but it may be useful for establishing vaccine safety.
(iii) Oral calf infection model.
One of the most robust models for Salmonella gastroenteritis is the calf infection model. Cows are natural hosts for Salmonella Typhimurium as well as other serovars, such as Salmonella Dublin and Salmonella Newport (91). Infection of calves with Salmonella Typhimurium causes a limiting gastroenteritis characterized by diarrhea, dehydration, and intestinal pathology. Likewise, Salmonella Dublin infection produces extensive intestinal pathology; however, it also disseminates to sterile sites, such as the spleen and liver, leading to systemic illness (92). Salmonella Dublin is found in both young and adult cows, with subclinical infections in heifers being a cause of abortion (93). In contrast, Salmonella Typhimurium is virulent predominantly in calves under 2 months of age (94, 95). This matches the age-dependent susceptibility observed in humans, where children constitute the majority of invasive cases (4). The incubation period for both serovars is around 48 h in calves, similar to humans and nonhuman primates (96). In addition, the intestinal pathology observed upon infection of calves with Salmonella Typhimurium is similar to that seen in rhesus macaques and humans (97, 98). These factors combined make calves a relevant model for investigation of Salmonella pathogenesis. Many virulence factors for Salmonella have been identified or confirmed in this model (99). Interestingly, there are a number of discrepancies between the genes required for virulence in calves versus those identified in mice. The most striking examples of these are the spv operon, which is absolutely required for systemic spread in mice (100) but dispensable in calves (101), and SPI-2, mutants of which are highly attenuated in mice (102) but only mildly attenuated in calves (101). Both of these examples refer to genes that are important in the systemic phase of infection. In contrast, mutations in genes involved in the intestinal phases of infection, such as pipA, have been shown to be attenuating in calf ileal loop models (15) but not in a systemic mouse model (E. Higginson and S. M. Tennant, unpublished data).
Salmonella Typhimurium live attenuated vaccines carrying mutations in the aro gene locus have been shown to be safe and effective in multiple studies using the calf infection model (103–105). Specifically, ΔaroA and ΔaroA ΔaroD mutants were shown to be well tolerated and only transiently excreted in orally vaccinated calves (103, 105). In these two studies, vaccination was shown to be highly effective at protecting animals from diarrhea and ultimately death. Interestingly, while animals immunized with a heat-inactivated vaccine showed greater antibody responses to lipopolysaccharide (LPS) and outer membrane porins, those given the live attenuated aro mutant vaccines were significantly better protected (104). This model is useful in determining not only protection against gastroenteritis but also key vaccine characteristics, such as shedding and immune correlates.
(iv) Ileal loop model.
The ileal loop model has been optimized for many different species, including cows, rabbits, pigs, and primates (15, 106–108). With this model, animals undergo a surgical procedure in which the ileum is tied into multiple equal loops. These loops are then injected with bacteria, and the animal is left to recover for up to 18 h. Within this closed system, fluid accumulates over the course of the incubation period. The level of fluid that accumulates can be used as a proxy for the induction of diarrhea. This system for measuring fluid secretion has been used for other bacterial pathogens (109–111), as well as toxins that act on the mucosa, such as cholera toxin (112) and toxin A from Clostridium difficile (113).
Besides fluid accumulation, other aspects of the early infection process can be assessed using this model, including inflammation of the mucosa, presence of immune cell infiltrate, and bacterial adherence and invasion (106). The major advantage of this model is the potential to assess multiple experimental conditions in the same animal, thus increasing the number of experimental replicates without increasing the number of animals required. However, the obvious limitation is that it can only model the early stages of the infection process.
This model has been used to assess safety of live attenuated vaccine candidates. Specifically, researchers identified one gene, invA, which when deleted led to significantly decreased fluid secretion, tissue damage, and inflammation in ileal loops, compared to an aroA mutant and the wild-type parental Salmonella Typhimurium strain SL1344 (114).
(v) Nonhuman primate models.
Due to the close genetic relationship with humans, nonhuman primates provide the most relevant model for determination of vaccine efficacy. Salmonella Typhimurium has been evaluated in rhesus macaques (98, 115, 116), while Salmonella Typhi has been tested in chimpanzees (117–119). In both models, animals are infected via the oral route and show symptoms similar to those in humans, such as diarrhea, weight loss, and fever. In a study published in 1970, researchers showed a decrease in clinical symptoms (fever, dehydration, weight loss) upon challenge of chimpanzees immunized with a live attenuated Salmonella Typhi vaccine (119). Unfortunately, due to the small sample size and high biological variation in response to challenge, there was difficulty in determining the relevant endpoint criteria for calculating vaccine efficacy. This remains a significant concern for those looking to use nonhuman primate models, for which group sizes are often restricted due to the high cost of such studies.
More recently, we used the rhesus macaque model to evaluate the live-attenuated Salmonella Typhimurium vaccine candidate CVD 1921. This novel strain was shown to be significantly attenuated, with decreased systemic spread, shedding, and clinical disease manifestations (116). Preliminary analysis of the humoral immune response to vaccination showed seroconversion for both anti-LPS and anti-FliC (flagellin) serum IgG in three (out of three) immunized rhesus macaques. We also showed that the vaccine was safe in simian immunodeficiency (SIV)-infected rhesus macaques. These promising preclinical data provide a strong rationale for advancement of this vaccine candidate. This ability to accurately model human disease and the potential interactions with other infections thus makes the primate model very persuasive in the submission of investigational new drug applications to regulatory authorities, a key step in advancement of promising candidates into clinical investigations.
INSECT, NEMATODE, AND FISH MODELS
Although experiments conducted in higher vertebrates are more medically relevant, there are advantages to lower-order animal models. First, they are favorable from an animal use ethics standpoint. Second, the model organisms can be easily propagated in the laboratory, thereby decreasing costs and increasing the number of replicates. Finally, they can be used in situations where larger models are not practical, such as in space (e.g., on space shuttles or the International Space Station) (120) or when high-throughput analysis is required. It is in this capacity for use in a high-throughput screening assay that these models may be most useful in vaccine research.
Caenorhabditis elegans is the most highly used invertebrate model for bacterial pathogenesis. This free-living nematode has been used for virulence assessment of other pathogens, including Pseudomonas aeruginosa and enteropathogenic Escherichia coli (121–123). Worms fed a diet of Salmonella Typhimurium have significantly shorter time to death than worms fed laboratory E. coli strain OP50 (124, 125). This time-to-death model has allowed researchers to identify several genes that are required for virulence in C. elegans, including genes located in SPI-1, SPI-3, SPI-4, and SPI-5 (126).
The Galleria mellonella wax moth model has recently been used to evaluate the pathogenesis of bacterial species such as P. aeruginosa, Acinetobacter baumannii, and Listeria monocytogenes, among others (127–129). The wax moth larvae are infected by injecting bacterial suspensions into the hindmost proleg and assessing viability over time. When infected with Salmonella Typhimurium at various doses, larvae showed a clear dose-dependent response, with a 50% lethal dose of 3.6 × 103 CFU (130). This ability to control the bacterial dosage is one advantage over the C. elegans model, which relies on worms grazing on a bacterial lawn.
A second insect model is that of the fruit fly, Drosophila melanogaster. Flies infected by injection of 104 CFU of Salmonella Typhimurium succumbed to infection within 7 to 9 days (131). When infected with Salmonella Typhimurium, strains carrying mutations in SPI-1 (orgA::Tn10) or SPI-2 (ssrA::miniTn5), the time to death for flies was significantly longer; however, the bacterial load in the flies was almost 4-fold higher than for wild-type-infected flies. The authors postulated that the expression of secreted effectors in the wild-type infection activated the fly immune response, limiting bacterial burden but also causing damage and eventually death.
Zebrafish may be even more relevant to vaccine research, as they have both innate and adaptive immune systems. To assess virulence in real time, fluorescently tagged Salmonella Typhimurium cells are injected into the axial vein of zebrafish embryos (132). As with Salmonella infections in humans, the bacterium resides in macrophage-like cells, which are not able to clear the bacterium. Embryos given a comparatively high dose of bacteria (50 CFU) succumbed to infection within 2 days, while a percentage of those given lower doses survived, suggesting that the zebrafish are able to resist infection. When infected with Salmonella LPS mutant strains, the zebrafish embryos were able to quickly kill bacteria, thus showing an active immune defense.
COMORBIDITY MODELS
As a vaccine candidates moves closer to clinical testing, it becomes important to assess how the vaccine will fare in the broader population, where underlying comorbidities, such as HIV infection, may be present. In recent epidemiological studies in Malawi, the prevalence of invasive NTS (iNTS) infections was found to be intrinsically entwined with rates of HIV, malaria, and malnutrition (133). This supports earlier reports from the Gambia and Kenya, where decreases in iNTS infections were noted upon implementation of effective malaria interventions (134, 135). As such, determining the safety and efficacy of vaccines in populations with these underlying conditions represents a new challenge.
It is well known that underlying malnutrition significantly impairs a person's ability to respond effectively to vaccination. Although malnutrition models for other pathogens, such as Cryptosporidium and Leishmania, have been developed (136–138), as of yet there is no such model for Salmonella. These models use combined protein, iron, and zinc depletion to induce a state of malnutrition in mice. Malnourished mice are then compared to healthy controls in their ability to respond to immunization and mount an effective immune response to challenge. This is important because several Salmonella vaccines in development are live attenuated and thus rely on a functional gut immune system. These vaccines will likely be used in areas where malnutrition and other enteropathies are rife. Experience has taught us that oral vaccines often show reduced efficacy when introduced in developing countries (139). Thus, the development of a model to predict vaccine performance in malnourished children (and/or those with environmental enteropathy) is an area of Salmonella research that merits further investigation.
Several groups have examined the effect of malaria coinfection on Salmonella in mice. In one model, researchers used simultaneous intraperitoneal coinfection with Salmonella Typhimurium and Plasmodium berghei to establish bacterial and parasite loads, as well as investigate the impact on the immune response (140). Both the parasite and bacterial loads were found to be significantly increased in coinfection, compared to those from single infections. This increased burden was also associated with higher levels of tissue damage and mortality. The authors postulated that this outcome was driven largely by uncontrolled oxidative stress brought on by an unchecked immune response. In a second study, the authors recapitulated the likely infection process in areas where the pathogens are endemic by first inducing parisitemia and anemia in mice before orogastrically challenging animals with Salmonella Typhimurium (141). In this staggered secondary infection model, researchers similarly observed high systemic levels of bacteria and parasites, with bacterial replication in the liver significantly increased. This unchecked liver replication was linked to parasite-induced IL-10 production, which blunted the immune response to Salmonella.
Finally, HIV-Salmonella coinfection has been modeled using SIV-infected rhesus macaques. Animals primarily infected with SIV and subsequently infected with Salmonella Typhimurium had much greater systemic spread of bacteria than did immunocompetent animals (142). This was linked to the depletion of Th17 cells in SIV-infected animals. The contribution of Th17 cells to the intestinal barrier function was confirmed by those authors by using IL-17 knockout mice (Il17ra−/−), which also showed increased systemic dissemination of Salmonella compared to immunocompetent mice. As described above, the SIV coinfection model has also been used to evaluate safety of a live attenuated Salmonella Typhimurium vaccine candidate, CVD 1921 (116). This vaccine was well tolerated in SIV-infected macaques, with the vaccine strain being cleared within 4 days. One out of three rhesus macaques seroconverted for LPS and flagella, proving that induction of an immune response is possible. This number could potentially be increased by vaccinating with additional doses, as is required for the Salmonella Typhi live attenuated vaccine Ty21a, which is administered in three or four doses.
CONCLUSIONS
The animal models for human salmonellosis are many and varied. Thus, there is a plethora of options for researchers looking to evaluate new vaccine candidates, but care must be taken to select the most appropriate model to address the research question at hand. While mouse models have been, and likely will continue to be, the most highly utilized models, integrating mouse data with results from other models may provide a more complete assessment of vaccine candidates. In addition, implementation of larger animal models and comorbidity models should be prioritized for vaccine candidates that have already been evaluated in smaller animal models, in order to advance these candidates toward clinical trials and to predict their efficacy in target populations.
Funding Statement
This work was funded by Project 4: Vaccine strategy for broad-spectrum protection against non-typhoidal Salmonella (project leaders, Sharon M. Tennant and Raphael Simon) of the NIH/NIAID Centers for Excellence in Translation Research grant U19 AI109776-01 (principal investigator, Myron M. Levine) and also grant NNX13AN80G from the National Aeronautics and Space Administration (principal investigator, Myron M. Levine).
REFERENCES
- 1.Parry CM, Hien TT, Dougan G, White NJ, Farrar JJ. 2002. Typhoid fever. N Engl J Med 347:1770–1782. doi: 10.1056/NEJMra020201. [DOI] [PubMed] [Google Scholar]
- 2.Stuart BM, Pullen RL. 1946. Typhoid: clinical analysis of three hundred and sixty cases. Arch Intern Med 78:629–661. doi: 10.1001/archinte.1946.00220060002001. [DOI] [PubMed] [Google Scholar]
- 3.Hohmann EL. 2001. Nontyphoidal salmonellosis. Clin Infect Dis 32:263–269. doi: 10.1086/318457. [DOI] [PubMed] [Google Scholar]
- 4.CDC. 2014. Foodborne Diseases Active Surveillance Network (FoodNet): FoodNet surveillance report for 2012 (final report). CDC, U.S. Department of Health and Human Services, Atlanta, GA. [Google Scholar]
- 5.Feasey NA, Dougan G, Kingsley RA, Heyderman RS, Gordon MA. 2012. Invasive non-typhoidal Salmonella disease: an emerging and neglected tropical disease in Africa. Lancet 379:2489–2499. doi: 10.1016/S0140-6736(11)61752-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Gordon MA, Graham SM, Walsh AL, Wilson L, Phiri A, Molyneux E, Zijlstra EE, Heyderman RS, Hart CA, Molyneux ME. 2008. Epidemics of invasive Salmonella enterica serovar Enteritidis and S. enterica serovar Typhimurium infection associated with multidrug resistance among adults and children in Malawi. Clin Infect Dis 46:963–969. doi: 10.1086/529146. [DOI] [PubMed] [Google Scholar]
- 7.Hoelzer K, Switt AM, Wiedmann M. 2011. Animal contact as a source of human non-typhoidal salmonellosis. Vet Res 42:34. doi: 10.1186/1297-9716-42-34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Coburn B, Grassl GA, Finlay B. 2007. Salmonella, the host and disease: a brief review. Immunol Cell Biol 85:112–118. doi: 10.1038/sj.icb.7100007. [DOI] [PubMed] [Google Scholar]
- 9.Buchmeier NA, Heffron F. 1991. Inhibition of macrophage phagosome-lysosome fusion by Salmonella typhimurium. Infect Immun 59:2232–2238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.House D, Bishop A, Parry C, Dougan G, Wain J. 2001. Typhoid fever: pathogenesis and disease. Curr Opin Infect Dis 14:573–578. doi: 10.1097/00001432-200110000-00011. [DOI] [PubMed] [Google Scholar]
- 11.Mandal B, Brennand J. 1988. Bacteraemia in salmonellosis: a 15 year retrospective study from a regional infectious diseases unit. BMJ 297:1242–1243. doi: 10.1136/bmj.297.6658.1242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Hayward MR, Jansen VA, Woodward MJ. 2013. Comparative genomics of Salmonella enterica serovars Derby and Mbandaka, two prevalent serovars associated with different livestock species in the UK. BMC Genomics 14:1. doi: 10.1186/1471-2164-14-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Blanc-Potard A-B, Solomon F, Kayser J, Groisman EA. 1999. The SPI-3 pathogenicity island of Salmonella enterica. J Bacteriol 181:998–1004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Bäumler A, Kusters JG, Stojiljkovic I, Heffron F. 1994. Salmonella typhimurium loci involved in survival within macrophages. Infect Immun 62:1623–1630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Wood MW, Jones MA, Watson PR, Hedges S, Wallis TS, Galyov EE. 1998. Identification of a pathogenicity island required for Salmonella enteropathogenicity. Mol Microbiol 29:883–891. doi: 10.1046/j.1365-2958.1998.00984.x. [DOI] [PubMed] [Google Scholar]
- 16.Parkhill J, Dougan G, James K, Thomson N, Pickard D, Wain J, Churcher C, Mungall K, Bentley S, Holden M. 2001. Complete genome sequence of a multiple drug resistant Salmonella enterica serovar Typhi CT18. Nature 413:848–852. doi: 10.1038/35101607. [DOI] [PubMed] [Google Scholar]
- 17.Tennant SM, Levine MM. 2015. Live attenuated vaccines for invasive Salmonella infections. Vaccine 33(Suppl 3):C36–C41. doi: 10.1016/j.vaccine.2015.04.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Germanier R, Fiirer E. 1975. Isolation and characterization of GalE mutant Ty21a of Salmonella typhi: a candidate strain for a live, oral typhoid vaccine. J Infect Dis 131:553–558. doi: 10.1093/infdis/131.5.553. [DOI] [PubMed] [Google Scholar]
- 19.Black RE, Levine MM, Ferreccio C, Clements ML, Lanata C, Rooney J, Germanier R, Chilean Typhoid Committee. 1990. Efficacy of one or two doses of Ty21a Salmonella typhi vaccine in enteric-coated capsules in a controlled field trial. Vaccine 8:81–84. doi: 10.1016/0264-410X(90)90183-M. [DOI] [PubMed] [Google Scholar]
- 20.Levine MM, Ferreccio C, Black RE, Lagos R, San Martin O, Blackwelder WC. 2007. Ty21a live oral typhoid vaccine and prevention of paratyphoid fever caused by Salmonella enterica serovar Paratyphi B. Clin Infect Dis 45:S24–S28. doi: 10.1086/518141. [DOI] [PubMed] [Google Scholar]
- 21.Ochiai RL, Wang X, Von Seidlein L, Yang J, Bhutta ZA, Bhattacharya SK, Agtini M, Deen JL, Wain J, Kim DR. 2005. Salmonella Paratyphi A rates, Asia. Emerg Infect Dis 11:1764–1766. doi: 10.3201/eid1111.050168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Woods CW, Murdoch DR, Zimmerman MD, Glover WA, Basnyat B, Wolf L, Belbase RH, Reller LB. 2006. Emergence of Salmonella enterica serotype Paratyphi A as a major cause of enteric fever in Kathmandu, Nepal. Trans R Soc Trop Med Hyg 100:1063–1067. doi: 10.1016/j.trstmh.2005.12.011. [DOI] [PubMed] [Google Scholar]
- 23.Konadu EY, Lin F-YC, Hó VA, Thuy NTT, Van Bay P, Thanh TC, Khiem HB, Trach DD, Karpas AB, Li J. 2000. Phase 1 and phase 2 studies of Salmonella enterica serovar Paratyphi A O-specific polysaccharide-tetanus toxoid conjugates in adults, teenagers, and 2-to 4-year-old children in Vietnam. Infect Immun 68:1529–1534. doi: 10.1128/IAI.68.3.1529-1534.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Micoli F, Rondini S, Gavini M, Lanzilao L, Medaglini D, Saul A, Martin LB. 2012. O:2-CRM(197) conjugates against Salmonella Paratyphi A. PLoS One 7:e47039. doi: 10.1371/journal.pone.0047039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.MacLennan CA, Martin LB, Micoli F. 2014. Vaccines against invasive Salmonella disease: current status and future directions. Hum Vaccin Immunother 10:1478–1493. doi: 10.4161/hv.29054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Ao TT, Feasey NA, Gordon MA, Keddy KH, Angulo FJ, Crump JA. 2015. Global burden of invasive nontyphoidal Salmonella disease, 2010. Emerg Infect Dis 21:941–949. doi: 10.3201/eid2106.140999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Levine M, Tapia M, Bornstein K, Tennant S, Sow S, Mandomando I, MacLennan C. 2014. An overview of invasive non-typhoidal Salmonella (iNTS) epidemiology. Int J Infect Dis 21:11. doi: 10.1016/j.ijid.2014.03.427. [DOI] [Google Scholar]
- 28.Simon R, Tennant SM, Wang JY, Schmidlein PJ, Lees A, Ernst RK, Pasetti MF, Galen JE, Levine MM. 2011. Salmonella enterica serovar Enteritidis core O polysaccharide conjugated to H:g,m flagellin as a candidate vaccine for protection against invasive infection with S. Enteritidis. Infect Immun 79:4240–4249. doi: 10.1128/IAI.05484-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Tennant SM, MacLennan CA, Simon R, Martin LB, Khan MI. 2016. Nontyphoidal Salmonella disease: current status of vaccine research and development. Vaccine 34:2907–2910. doi: 10.1016/j.vaccine.2016.03.072. [DOI] [PubMed] [Google Scholar]
- 30.Hoffmann S, Batz MB, Morris JG Jr. 2012. Annual cost of illness and quality-adjusted life year losses in the United States due to 14 foodborne pathogens. J Food Prot 75:1292–1302. doi: 10.4315/0362-028X.JFP-11-417. [DOI] [PubMed] [Google Scholar]
- 31.Vugia DJ, Samuel M, Farley MM, Marcus R, Shiferaw B, Shallow S, Smith K, Angulo FJ, Emerging Infections Program FoodNet Working Group. 2004. Invasive Salmonella infections in the United States, FoodNet, 1996-1999: incidence, serotype distribution, and outcome. Clin Infect Dis 38:S149–S156. doi: 10.1086/381581. [DOI] [PubMed] [Google Scholar]
- 32.Mishu B, Koehler J, Lee LA, Rodrigue D, Brenner FH, Blake P, Tauxe RV. 1994. Outbreaks of Salmonella Enteritidis infections in the United States, 1985-1991. J Infect Dis 169:547–552. doi: 10.1093/infdis/169.3.547. [DOI] [PubMed] [Google Scholar]
- 33.Kirk MD, Veitch MG, Hall GV. 2010. Gastroenteritis and food-borne disease in elderly people living in long-term care. Clin Infect Dis 50:397–404. doi: 10.1086/649878. [DOI] [PubMed] [Google Scholar]
- 34.Mastroeni P, Sheppard M. 2004. Salmonella infections in the mouse model: host resistance factors and in vivo dynamics of bacterial spread and distribution in the tissues. Microbes Infect 6:398–405. doi: 10.1016/j.micinf.2003.12.009. [DOI] [PubMed] [Google Scholar]
- 35.Santos RL, Zhang S, Tsolis RM, Kingsley RA, Adams LG, Bäumler AJ. 2001. Animal models of Salmonella infections: enteritis versus typhoid fever. Microbes Infect 3:1335–1344. doi: 10.1016/S1286-4579(01)01495-2. [DOI] [PubMed] [Google Scholar]
- 36.Simon R, Tennant SM, Galen JE, Levine MM. 2011. Mouse models to assess the efficacy of non-typhoidal Salmonella vaccines: revisiting the role of host innate susceptibility and routes of challenge. Vaccine 29:5094–5106. doi: 10.1016/j.vaccine.2011.05.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Strugnell RA, Scott TA, Wang N, Yang C, Peres N, Bedoui S, Kupz A. 2014. Salmonella vaccines: lessons from the mouse model or bad teaching? Curr Opin Microbiol 17:99–105. doi: 10.1016/j.mib.2013.12.004. [DOI] [PubMed] [Google Scholar]
- 38.Baker S, Dougan G. 2007. The genome of Salmonella enterica serovar Typhi. Clin Infect Dis 45:S29–S33. doi: 10.1086/518143. [DOI] [PubMed] [Google Scholar]
- 39.Caron J, Loredo-Osti JC, Laroche L, Skamene E, Morgan K, Malo D. 2002. Identification of genetic loci controlling bacterial clearance in experimental Salmonella Enteritidis infection: an unexpected role of Nramp1 (Slc11a1) in the persistence of infection in mice. Genes Immun 3:196–204. doi: 10.1038/sj.gene.6363850. [DOI] [PubMed] [Google Scholar]
- 40.Fritsche G, Dlaska M, Barton H, Theurl I, Garimorth K, Weiss G. 2003. Nramp1 functionality increases inducible nitric oxide synthase transcription via stimulation of IFN regulatory factor 1 expression. J Immunol 171:1994–1998. doi: 10.4049/jimmunol.171.4.1994. [DOI] [PubMed] [Google Scholar]
- 41.Fritsche G, Nairz M, Theurl I, Mair S, Bellmann-Weiler R, Barton HC, Weiss G. 2007. Modulation of macrophage iron transport by Nramp1 (Slc11a1). Immunobiology 212:751–757. doi: 10.1016/j.imbio.2007.09.014. [DOI] [PubMed] [Google Scholar]
- 42.Fritsche G, Nairz M, Werner ER, Barton HC, Weiss G. 2008. Nramp1-functionality increases iNOS expression via repression of IL-10 formation. Eur J Immunol 38:3060–3067. doi: 10.1002/eji.200838449. [DOI] [PubMed] [Google Scholar]
- 43.Hone DM, Harris AM, Chatfield S, Dougan G, Levine MM. 1991. Construction of genetically defined double aro mutants of Salmonella Typhi. Vaccine 9:810–816. doi: 10.1016/0264-410X(91)90218-U. [DOI] [PubMed] [Google Scholar]
- 44.Powell CJ, DeSett CR, Lowenthal JP, Berman S. 1980. The effect of adding iron to mucin on the enhancement of virulence for mice of Salmonella Typhi strain TY 2. J Biol Stand 8:79–85. doi: 10.1016/S0092-1157(80)80049-7. [DOI] [PubMed] [Google Scholar]
- 45.Gat O, Galen JE, Tennant S, Simon R, Blackwelder WC, Silverman DJ, Pasetti MF, Levine MM. 2011. Cell-associated flagella enhance the protection conferred by mucosally-administered attenuated Salmonella Paratyphi A vaccines. PLoS Negl Trop Dis 5:e1373. doi: 10.1371/journal.pntd.0001373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Wang JY, Pasetti MF, Noriega FR, Anderson RJ, Wasserman SS, Galen JE, Sztein MB, Levine MM. 2001. Construction, genotypic and phenotypic characterization, and immunogenicity of attenuated ΔguaBA Salmonella enterica serovar Typhi strain CVD 915. Infect Immun 69:4734–4741. doi: 10.1128/IAI.69.8.4734-4741.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Galen JE, Gomez-Duarte OG, Losonsky GA, Halpern JL, Lauderbaugh CS, Kaintuck S, Reymann MK, Levine MM. 1997. A murine model of intranasal immunization to assess the immunogenicity of attenuated Salmonella Typhi live vector vaccines in stimulating serum antibody responses to expressed foreign antigens. Vaccine 15:700–708. doi: 10.1016/S0264-410X(96)00227-7. [DOI] [PubMed] [Google Scholar]
- 48.Tacket CO, Sztein MB, Losonsky GA, Wasserman SS, Nataro JP, Edelman R, Pickard D, Dougan G, Chatfield SN, Levine MM. 1997. Safety of live oral Salmonella typhi vaccine strains with deletions in htrA and aroC aroD and immune response in humans. Infect Immun 65:452–456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Tacket CO, Sztein MB, Wasserman SS, Losonsky G, Kotloff KL, Wyant TL, Nataro JP, Edelman R, Perry J, Bedford P, Brown D, Chatfield S, Dougan G, Levine MM. 2000. Phase 2 clinical trial of attenuated Salmonella enterica serovar Typhi oral live vector vaccine CVD 908-htrA in U.S. volunteers. Infect Immun 68:1196–1201. doi: 10.1128/IAI.68.3.1196-1201.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Tacket CO, Hone DM, Losonsky GA, Guers L, Edelman R, Levine MM. 1992. Clinical acceptability and immunogenicity of CVD 908 Salmonella typhi vaccine strain. Vaccine 10:443–446. doi: 10.1016/0264-410X(92)90392-W. [DOI] [PubMed] [Google Scholar]
- 51.Baker SJ, Daniels C, Morona R. 1997. PhoP/Q regulated genes in Salmonella typhi: identification of melittin sensitive mutants. Microb Pathog 22:165–179. doi: 10.1006/mpat.1996.0099. [DOI] [PubMed] [Google Scholar]
- 52.Hohmann EL, Oletta CA, Killeen KP, Miller SI. 1996. phoP/phoQ-deleted Salmonella typhi (Ty800) is a safe and immunogenic single dose typhoid fever vaccine in volunteers. J Infect Dis 173:1408–1414. doi: 10.1093/infdis/173.6.1408. [DOI] [PubMed] [Google Scholar]
- 53.Matsui H, Isshiki Y, Eguchi M, Ogawa Y, Shimoji Y. 2015. Evaluation of the live vaccine efficacy of virulence plasmid-cured, and phoP- or aroA-deficient Salmonella enterica serovar Typhimurium in mice. J Vet Med Sci 77:181. doi: 10.1292/jvms.14-0013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Tennant SM, Schmidlein P, Simon R, Pasetti MF, Galen JE, Levine MM. 2015. Refined live-attenuated Salmonella enterica serovar Typhimurium and Enteritidis vaccines mediate homologous and heterologous serogroup protection in mice. Infect Immun 83:4504–4512. doi: 10.1128/IAI.00924-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Galen JE, Wang JY, Carrasco JA, Lloyd SA, Mellado-Sanchez G, Diaz-McNair J, Franco O, Buskirk AD, Nataro JP, Pasetti MF. 2015. A bivalent typhoid live vector vaccine expressing both chromosome- and plasmid-encoded Yersinia pestis antigens fully protects against murine lethal pulmonary plague infection. Infect Immun 83:161–172. doi: 10.1128/IAI.02443-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Hess J, Ladel C, Miko D, Kaufmann S. 1996. Salmonella typhimurium aroA-infection in gene-targeted immunodeficient mice: major role of CD4+ TCR-alpha beta cells and IFN-gamma in bacterial clearance independent of intracellular location. J Immunol 156:3321–3326. [PubMed] [Google Scholar]
- 57.Mastroeni P, Simmons C, Fowler R, Hormaeche C, Dougan G. 2000. Igh-6−/− (B-cell-deficient) mice fail to mount solid acquired resistance to oral challenge with virulent Salmonella enterica serovar Typhimurium and show impaired Th1 T-cell responses to Salmonella antigens. Infect Immun 68:46–53. doi: 10.1128/IAI.68.1.46-53.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Vazquez-Torres A, Vallance BA, Bergman MA, Finlay BB, Cookson BT, Jones-Carson J, Fang FC. 2004. Toll-like receptor 4 dependence of innate and adaptive immunity to Salmonella: importance of the Kupffer cell network. J Immunol 172:6202–6208. doi: 10.4049/jimmunol.172.10.6202. [DOI] [PubMed] [Google Scholar]
- 59.Weintraub BC, Eckmann L, Okamoto S, Hense M, Hedrick SM, Fierer J. 1997. Role of αβ and γδ T cells in the host response to Salmonella infection as demonstrated in T-cell-receptor-deficient mice of defined Ity genotypes. Infect Immun 65:2306–2312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Shiloh MU, MacMicking JD, Nicholson S, Brause JE, Potter S, Marino M, Fang F, Dinauer M, Nathan C. 1999. Phenotype of mice and macrophages deficient in both phagocyte oxidase and inducible nitric oxide synthase. Immunity 10:29–38. doi: 10.1016/S1074-7613(00)80004-7. [DOI] [PubMed] [Google Scholar]
- 61.Vazquez-Torres A, Jones-Carson J, Baumler AJ, Falkow S, Valdivia R, Brown W, Le M, Berggren R, Parks WT, Fang FC. 1999. Extraintestinal dissemination of Salmonella by CD18-expressing phagocytes. Nature 401:804–808. doi: 10.1038/44593. [DOI] [PubMed] [Google Scholar]
- 62.Everest P, Roberts M, Dougan G. 1998. Susceptibility to Salmonella typhimurium infection and effectiveness of vaccination in mice deficient in the tumor necrosis factor alpha p55 receptor. Infect Immun 66:3355–3364. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Izhar M, DeSilva L, Joysey HS, Hormaeche CE. 1990. Moderate immunodeficiency does not increase susceptibility to Salmonella typhimurium aroA live vaccines in mice. Infect Immun 58:2258–2261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Mastroeni P, Harrison J, Robinson J, Clare S, Khan S, Maskell D, Dougan G, Hormaeche C. 1998. Interleukin-12 is required for control of the growth of attenuated aromatic-compound-dependent salmonellae in BALB/c mice: role of gamma interferon and macrophage activation. Infect Immun 66:4767–4776. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Sinha K, Mastroeni P, Harrison J, de Hormaeche RD, Hormaeche CE. 1997. Salmonella typhimurium aroA, htrA, and aroD htrA mutants cause progressive infections in athymic (nu/nu) BALB/c mice. Infect Immun 65:1566–1569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Grant AJ, Oshota O, Chaudhuri RR, Mayho M, Peters SE, Clare S, Maskell DJ, Mastroeni P. 2016. Genes required for the fitness of Salmonella enterica serovar Typhimurium during infection of immunodeficient gp91−/− phox mice. Infect Immun 84:989–997. doi: 10.1128/IAI.01423-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Mathur R, Oh H, Zhang D, Park S-G, Seo J, Koblansky A, Hayden MS, Ghosh S. 2012. A mouse model of Salmonella typhi infection. Cell 151:590–602. doi: 10.1016/j.cell.2012.08.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Song J, Wilhelm CL, Wangdi T, Maira-Litran T, Lee S-J, Raetz M, Sturge CR, Mirpuri J, Pei J, Grishin NV. 2016. Absence of TLR11 in mice does not confer susceptibility to Salmonella Typhi. Cell 164:827–828. doi: 10.1016/j.cell.2016.02.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Mathur R, Zeng W, Hayden MS, Ghosh S. 2016. Mice lacking TLR11 exhibit variable Salmonella Typhi susceptibility. Cell 164:829–830. doi: 10.1016/j.cell.2016.02.020. [DOI] [PubMed] [Google Scholar]
- 70.Song J, Willinger T, Rongvaux A, Eynon EE, Stevens S, Manz MG, Flavell RA, Galán JE. 2010. A mouse model for the human pathogen Salmonella typhi. Cell Host Microbe 8:369–376. doi: 10.1016/j.chom.2010.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Libby SJ, Brehm MA, Greiner DL, Shultz LD, McClelland M, Smith KD, Cookson BT, Karlinsey JE, Kinkel TL, Porwollik S. 2010. Humanized nonobese diabetic-scid IL2rγ null mice are susceptible to lethal Salmonella Typhi infection. Proc Natl Acad Sci U S A 107:15589–15594. doi: 10.1073/pnas.1005566107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Firoz Mian M, Pek EA, Chenoweth MJ, Ashkar AA. 2011. Humanized mice are susceptible to Salmonella typhi infection. Cell Mol Immunol 8:83–87. doi: 10.1038/cmi.2010.52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Takeuchi A. 1967. Electron microscope studies of experimental Salmonella infection. I. Penetration into the intestinal epithelium by Salmonella typhimurium. Am J Pathol 50:109. [PMC free article] [PubMed] [Google Scholar]
- 74.Takeuchi A, Sprinz H. 1967. Electron-microscope studies of experimental Salmonella infection in the preconditioned guinea pig. II. Response of the intestinal mucosa to the invasion by Salmonella typhimurium. Am J Pathol 51:137. [PMC free article] [PubMed] [Google Scholar]
- 75.Robertson JM, McKenzie NH, Duncan M, Allen-Vercoe E, Woodward MJ, Flint HJ, Grant G. 2003. Lack of flagella disadvantages Salmonella enterica serovar Enteritidis during the early stages of infection in the rat. J Med Microbiol 52:91–99. doi: 10.1099/jmm.0.04901-0. [DOI] [PubMed] [Google Scholar]
- 76.Havelaar A, Garssen J, Takumi K, Koedam M, Dufrenne J, Van Leusden F, de La Fonteyne L, Bousema J, Vos J. 2001. A rat model for dose-response relationships of Salmonella Enteritidis infection. J Appl Microbiol 91:442–452. doi: 10.1046/j.1365-2672.2001.01399.x. [DOI] [PubMed] [Google Scholar]
- 77.Redman TK, Harmon CC, Lallone RL, Michalek SM. 1995. Oral immunization with recombinant Salmonella typhimurium expressing surface protein antigen A of Streptococcus sobrinus: dose response and induction of protective humoral responses in rats. Infect Immun 63:2004–2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Hui F, Meng C, Guo N, Yang L, Shi F, Mao D. 2014. Evaluation of attenuated Salmonella Choleraesuis-mediated inhibin recombinant DNA vaccine in rats. Genet Mol Res 13:6113–6125. doi: 10.4238/2014.August.7.27. [DOI] [PubMed] [Google Scholar]
- 79.Kuang Y, Yan Y-C, Gao A-W, Zhai Y-M, Miao S-Y, Wang L-F, Koide S. 2000. Immune responses in rats following oral immunization with attenuated Salmonella Typhimurium expressing human sperm antigen. Arch Androl 45:169–180. doi: 10.1080/01485010050193940. [DOI] [PubMed] [Google Scholar]
- 80.Vinod N, Oh S, Kim S, Choi CW, Kim SC, Jung C-H. 2014. Chemically induced Salmonella Enteritidis ghosts as a novel vaccine candidate against virulent challenge in a rat model. Vaccine 32:3249–3255. doi: 10.1016/j.vaccine.2014.03.090. [DOI] [PubMed] [Google Scholar]
- 81.Roland KL, Tinge SA, Kochi SK, Thomas LJ, Killeen KP. 2010. Reactogenicity and immunogenicity of live attenuated Salmonella enterica serovar Paratyphi A enteric fever vaccine candidates. Vaccine 28:3679–3687. doi: 10.1016/j.vaccine.2010.03.019. [DOI] [PubMed] [Google Scholar]
- 82.Panda A, Tatarov I, Masek BJ, Hardick J, Crusan A, Wakefield T, Carroll K, Yang S, Hsieh Y-H, Lipsky MM, McLeod CG, Levine MM, Rothman RE, Gaydos CA, DeTolla LJ. 2014. A rabbit model of non-typhoidal Salmonella bacteremia. Comp Immunol Microbiol Infect Dis 37:211–220. doi: 10.1016/j.cimid.2014.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Barthel M, Hapfelmeier S, Quintanilla-Martínez L, Kremer M, Rohde M, Hogardt M, Pfeffer K, Rüssmann H, Hardt W-D. 2003. Pretreatment of mice with streptomycin provides a Salmonella enterica serovar Typhimurium colitis model that allows analysis of both pathogen and host. Infect Immun 71:2839–2858. doi: 10.1128/IAI.71.5.2839-2858.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Suar M, Jantsch J, Hapfelmeier S, Kremer M, Stallmach T, Barrow PA, Hardt W-D. 2006. Virulence of broad- and narrow-host-range Salmonella enterica serovars in the streptomycin-pretreated mouse model. Infect Immun 74:632–644. doi: 10.1128/IAI.74.1.632-644.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Kaiser P, Diard M, Stecher B, Hardt WD. 2012. The streptomycin mouse model for Salmonella diarrhea: functional analysis of the microbiota, the pathogen's virulence factors, and the host's mucosal immune response. Immunol Rev 245:56–83. doi: 10.1111/j.1600-065X.2011.01070.x. [DOI] [PubMed] [Google Scholar]
- 86.Periaswamy B, Maier L, Vishwakarma V, Slack E, Kremer M, Andrews-Polymenis HL, McClelland M, Grant AJ, Suar M, Hardt W-D. 2012. Live attenuated S. Typhimurium vaccine with improved safety in immuno-compromised mice. PLoS One 7:e45433. doi: 10.1371/journal.pone.0045433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Pesciaroli M, Aloisio F, Ammendola S, Pistoia C, Petrucci P, Tarantino M, Francia M, Battistoni A, Pasquali P. 2011. An attenuated Salmonella enterica serovar Typhimurium strain lacking the ZnuABC transporter induces protection in a mouse intestinal model of Salmonella infection. Vaccine 29:1783–1790. doi: 10.1016/j.vaccine.2010.12.111. [DOI] [PubMed] [Google Scholar]
- 88.Vishwakarma V, Pati NB, Chandel HS, Sahoo SS, Saha B, Suar M. 2012. Evaluation of Salmonella enterica serovar Typhimurium TTSS-2 deficient fur mutant as safe live-attenuated vaccine candidate for immunocompromised mice. PLoS One 7:e52043. doi: 10.1371/journal.pone.0052043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Pasquali P, Ammendola S, Pistoia C, Petrucci P, Tarantino M, Valente C, Marenzoni ML, Rotilio G, Battistoni A. 2008. Attenuated Salmonella enterica serovar Typhimurium lacking the ZnuABC transporter confers immune-based protection against challenge infections in mice. Vaccine 26:3421–3426. doi: 10.1016/j.vaccine.2008.04.036. [DOI] [PubMed] [Google Scholar]
- 90.Bäumler AJ, Tsolis RM, Bowe FA, Kusters JG, Hoffmann S, Heffron F. 1996. The pef fimbrial operon of Salmonella typhimurium mediates adhesion to murine small intestine and is necessary for fluid accumulation in the infant mouse. Infect Immun 64:61–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Smith BP. 2002. Large animal internal medicine, p 775–779. Mosby Inc., St. Louis, MO. [Google Scholar]
- 92.Rings D. 1985. Salmonellosis in calves. Vet Clin North Am Food Anim Pract 1:529–539. doi: 10.1016/S0749-0720(15)31301-3. [DOI] [PubMed] [Google Scholar]
- 93.Hall G, Jones P, Parsons K, Chanter N, Aitken M. 1979. Studies of the virulence of Salmonella Dublin in experimental infections of cattle and rats. Brit Vet J 135:243–248. [DOI] [PubMed] [Google Scholar]
- 94.Sojka W, Field H. 1970. Salmone uosis in England and Wales 1958-1967. Vet Bull (Weybridge) 40:515–531. [Google Scholar]
- 95.Smith B, Habasha F, Reina-Guerra M, Hardy A. 1979. Bovine salmonellosis: experimental production and characterization of the disease in calves, using oral challenge with Salmonella Typhimurium. Am J Vet Res 40:1510–1513. [PubMed] [Google Scholar]
- 96.Rankin J, Taylor R. 1966. The estimation of doses of Salmonella Typhimurium suitable for the experimental production of disease in calves. Vet Rec 78:706–707. doi: 10.1136/vr.78.21.706. [DOI] [PubMed] [Google Scholar]
- 97.Day D, Mandal B, Morson B. 1978. The rectal biopsy appearances in Salmonella colitis. Histopathology 2:117–131. doi: 10.1111/j.1365-2559.1978.tb01700.x. [DOI] [PubMed] [Google Scholar]
- 98.Rout W, Formal S, Dammin G, Giannella R. 1974. Pathophysiology of Salmonella diarrhea in the rhesus monkey: intestinal transport, morphological and bacteriological studies. Gastroenterology 67:59–70. [PubMed] [Google Scholar]
- 99.Costa LF, Paixão TA, Tsolis RM, Bäumler AJ, Santos RL. 2012. Salmonellosis in cattle: advantages of being an experimental model. Res Vet Sci 93:1–6. doi: 10.1016/j.rvsc.2012.03.002. [DOI] [PubMed] [Google Scholar]
- 100.Gulig PA, Curtiss R. 1987. Plasmid-associated virulence of Salmonella Typhimurium. Infect Immun 55:2891–2901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Tsolis RM, Adams LG, Ficht TA, Bäumler AJ. 1999. Contribution of Salmonella typhimurium virulence factors to diarrheal disease in calves. Infect Immun 67:4879–4885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Hensel M, Shea JE, Gleeson C, Jones MD, Dalton E, Holden DW. 1995. Simultaneous identification of bacterial virulence genes by negative selection. Science 269:400–403. doi: 10.1126/science.7618105. [DOI] [PubMed] [Google Scholar]
- 103.Jones P, Dougan G, Hayward C, Mackensie N, Collins P, Chatfield S. 1991. Oral vaccination of calves against experimental salmonellosis using a double aro mutant of Salmonella typhimurium. Vaccine 9:29–34. doi: 10.1016/0264-410X(91)90313-U. [DOI] [PubMed] [Google Scholar]
- 104.Lindberg AA, Robertsson JA. 1983. Salmonella typhimurium infection in calves: cell-mediated and humoral immune reactions before and after challenge with live virulent bacteria in calves given live or inactivated vaccines. Infect Immun 41:751–757. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Robertsson J, Lindberg A, Hoiseth S, Stocker B. 1983. Salmonella typhimurium infection in calves: protection and survival of virulent challenge bacteria after immunization with live or inactivated vaccines. Infect Immun 41:742–750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Giannella RA, Formal SB, Dammin GJ, Collins H. 1973. Pathogenesis of salmonellosis: studies of fluid secretion, mucosal invasion, and morphologic reaction in the rabbit ileum. J Clin Invest 52:441–453. doi: 10.1172/JCI107201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Reed W, Olander H, Thacker H. 1986. Studies on the pathogenesis of Salmonella typhimurium and Salmonella choleraesuis var. Kunzendorf infection in weanling pigs. Am J Vet Res 47:75–83. [PubMed] [Google Scholar]
- 108.Santos R, Almeida A, Xavier M, Paixão T, Wilson R, Dandekar S, Raffatellu M, Bäumler A. 2011. Enteric pathology and Salmonella-induced cell death in healthy and SIV-infected rhesus macaques. Vet Pathol Online 48:933–941. doi: 10.1177/0300985810386468. [DOI] [PubMed] [Google Scholar]
- 109.Formal S, Kundel D, Schneider H, Kunev N, Sprinz H. 1961. Studies with Vibrio cholerae in the ligated loop of the rabbit intestine. Br J Exp Pathol 42:504. [PMC free article] [PubMed] [Google Scholar]
- 110.Spira W, Goepfert J. 1972. Bacillus cereus-induced fluid accumulation in rabbit ileal loops. Appl Microbiol 24:341–348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.De SN, Chatterjee DN. 1953. An experimental study of the mechanism of action of Vibrio cholerae on the intestinal mucous membrane. J Pathol Bacteriol 66:559–562. doi: 10.1002/path.1700660228. [DOI] [PubMed] [Google Scholar]
- 112.Burrows W, Musteikis GM. 1966. Cholera infection and toxin in the rabbit ileal loop. J Infect Dis 162:183–190. [DOI] [PubMed] [Google Scholar]
- 113.Mitchell TJ, Ketley JM, Haslam SC, Stephen J, Burdon DW, Candy DC, Daniel R. 1986. Effect of toxin A and B of Clostridium difficile on rabbit ileum and colon. Gut 27:78–85. doi: 10.1136/gut.27.1.78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Everest P, Ketley J, Hardy S, Douce G, Khan S, Shea J, Holden D, Maskell D, Dougan G. 1999. Evaluation of Salmonella typhimurium mutants in a model of experimental gastroenteritis. Infect Immun 67:2815–2821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Kent TH, Formal S, Labrec E. 1966. Salmonella gastroenteritis in rhesus monkeys. Arch Pathol 82:272–279. [PubMed] [Google Scholar]
- 116.Ault A, Tennant SM, Gorres JP, Eckhaus M, Sandler NG, Roque A, Livio S, Bao S, Foulds KE, Kao S-F. 2013. Safety and tolerability of a live oral Salmonella typhimurium vaccine candidate in SIV-infected nonhuman primates. Vaccine 31:5879–5888. doi: 10.1016/j.vaccine.2013.09.041. [DOI] [PubMed] [Google Scholar]
- 117.Edsall G, Gaines S, Landy M, Tigertt WD, Sprinz H, Trapani R-J, Mandel AD, Benenson AS. 1960. Studies on infection and immunity in experimental typhoid fever. I. Typhoid fever in chimpanzees orally infected with Salmonella typhosa. J Exp Med 112:143–166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Gaines S, Sprinz H, Tully JG, Tigertt WD. 1968. Studies on infection and immunity in experimental typhoid fever. VII. The distribution of Salmonella typhi in chimpanzee tissue following oral challenge, and the relationship between the numbers of bacilli and morphologic lesions. J Infect Dis 118:293–306. [DOI] [PubMed] [Google Scholar]
- 119.Cvjetanović B, Mel DM, Felsenfeld O. 1970. Study of live typhoid vaccine in chimpanzees. Bull World Health Organ 42:499–507. [PMC free article] [PubMed] [Google Scholar]
- 120.Hammond TG, Stodieck L, Birdsall HH, Becker J, Koenig P, Hammond JS, Gunter MA, Allen PL. 2013. Effects of microgravity on the virulence of Salmonella toward Caenorhabditis elegans. New Space 1:123–131. doi: 10.1089/space.2013.0011. [DOI] [Google Scholar]
- 121.Tan M-W, Mahajan-Miklos S, Ausubel FM. 1999. Killing of Caenorhabditis elegans by Pseudomonas aeruginosa used to model mammalian bacterial pathogenesis. Proc Natl Acad Sci U S A 96:715–720. doi: 10.1073/pnas.96.2.715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Mahajan-Miklos S, Tan M-W, Rahme LG, Ausubel FM. 1999. Molecular mechanisms of bacterial virulence elucidated using a Pseudomonas aeruginosa-Caenorhabditis elegans pathogenesis model. Cell 96:47–56. doi: 10.1016/S0092-8674(00)80958-7. [DOI] [PubMed] [Google Scholar]
- 123.Anyanful A, Dolan-Livengood JM, Lewis T, Sheth S, DeZalia MN, Sherman MA, Kalman LV, Benian GM, Kalman D. 2005. Paralysis and killing of Caenorhabditis elegans by enteropathogenic Escherichia coli requires the bacterial tryptophanase gene. Mol Microbiol 57:988–1007. doi: 10.1111/j.1365-2958.2005.04739.x. [DOI] [PubMed] [Google Scholar]
- 124.Aballay A, Yorgey P, Ausubel FM. 2000. Salmonella typhimurium proliferates and establishes a persistent infection in the intestine of Caenorhabditis elegans. Curr Biol 10:1539–1542. doi: 10.1016/S0960-9822(00)00830-7. [DOI] [PubMed] [Google Scholar]
- 125.Labrousse A, Chauvet S, Couillault C, Kurz CL, Ewbank JJ. 2000. Caenorhabditis elegans is a model host for Salmonella Typhimurium. Curr Biol 10:1543–1545. doi: 10.1016/S0960-9822(00)00833-2. [DOI] [PubMed] [Google Scholar]
- 126.Tenor JL, McCormick BA, Ausubel FM, Aballay A. 2004. Caenorhabditis elegans-based screen identifies Salmonella virulence factors required for conserved host-pathogen interactions. Curr Biol 14:1018–1024. doi: 10.1016/j.cub.2004.05.050. [DOI] [PubMed] [Google Scholar]
- 127.Miyata S, Casey M, Frank DW, Ausubel FM, Drenkard E. 2003. Use of the Galleria mellonella caterpillar as a model host to study the role of the type III secretion system in Pseudomonas aeruginosa pathogenesis. Infect Immun 71:2404–2413. doi: 10.1128/IAI.71.5.2404-2413.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Mukherjee K, Altincicek B, Hain T, Domann E, Vilcinskas A, Chakraborty T. 2010. Galleria mellonella as a model system for studying Listeria pathogenesis. Appl Environ Microbiol 76:310–317. doi: 10.1128/AEM.01301-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Peleg AY, Jara S, Monga D, Eliopoulos GM, Moellering RC, Mylonakis E. 2009. Galleria mellonella as a model system to study Acinetobacter baumannii pathogenesis and therapeutics. Antimicrob Agents Chemother 53:2605–2609. doi: 10.1128/AAC.01533-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Bender JK, Wille T, Blank K, Lange A, Gerlach RG. 2013. LPS structure and PhoQ activity are important for Salmonella Typhimurium virulence in the Gallleria mellonella infection model. PLoS One 8:e73287. doi: 10.1371/journal.pone.0073287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Brandt SM, Dionne MS, Khush RS, Pham LN, Vigdal TJ, Schneider DS. 2004. Secreted bacterial effectors and host-produced Eiger/TNF drive death in a Salmonella-infected fruit fly. PLoS Biol 2:e418. doi: 10.1371/journal.pbio.0020418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Van Der Sar AM, Musters RJ, Van Eeden FJ, Appelmelk BJ, Vandenbroucke-Grauls CM, Bitter W. 2003. Zebrafish embryos as a model host for the real time analysis of Salmonella Typhimurium infections. Cell Microbiol 5:601–611. doi: 10.1046/j.1462-5822.2003.00303.x. [DOI] [PubMed] [Google Scholar]
- 133.Feasey NA, Everett D, Faragher EB, Roca-Feltrer A, Kang'ombe A, Denis B, Kerac M, Molyneux E, Molyneux M, Jahn A, Gordon MA, Heyderman RS. 2015. Modelling the contributions of malaria, HIV, malnutrition and rainfall to the decline in paediatric invasive non-typhoidal Salmonella disease in Malawi. PLoS Negl Trop Dis 9:e0003979. doi: 10.1371/journal.pntd.0003979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Mackenzie G, Ceesay SJ, Hill PC, Walther M, Bojang KA, Satoguina J, Enwere G, D'Alessandro U, Saha D, Ikumapayi UNA, O'Dempsey T, Mabey DCW, Corrah T, Conway DJ, Adegbola RA, Greenwood BM. 2010. A decline in the incidence of invasive non-typhoidal Salmonella infection in the Gambia temporally associated with a decline in malaria infection. PLoS One 5:e10568. doi: 10.1371/journal.pone.0010568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Scott JAG, Berkley JA, Mwangi I, Ochola L, Uyoga S, Macharia A, Ndila C, Lowe BS, Mwarumba S, Bauni E, Marsh K, Williams TN. 2011. Relation between falciparum malaria and bacteraemia in Kenyan children: a population-based, case-control study and a longitudinal study. Lancet 378:1316–1323. doi: 10.1016/S0140-6736(11)60888-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Malafaia G, Serafim T, Silva M, Pedrosa M, Rezende S. 2009. Protein-energy malnutrition decreases immune response to Leishmania chagasi vaccine in BALB/c mice. Parasite Immunol 31:41–49. doi: 10.1111/j.1365-3024.2008.01069.x. [DOI] [PubMed] [Google Scholar]
- 137.Roche JK, Rojo AL, Costa LB, Smeltz R, Manque P, Woehlbier U, Bartelt L, Galen J, Buck G, Guerrant RL. 2013. Intranasal vaccination in mice with an attenuated Salmonella enterica serovar 908htrA expressing Cp15 of Cryptosporidium: impact of malnutrition with preservation of cytokine secretion. Vaccine 31:912–918. doi: 10.1016/j.vaccine.2012.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Anstead GM, Chandrasekar B, Zhao W, Yang J, Perez LE, Melby PC. 2001. Malnutrition alters the innate immune response and increases early visceralization following Leishmania donovani infection. Infect Immun 69:4709–4718. doi: 10.1128/IAI.69.8.4709-4718.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Levine MM. 2010. Immunogenicity and efficacy of oral vaccines in developing countries: lessons from a live cholera vaccine. BMC Biol 8:1. doi: 10.1186/1741-7007-8-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Shukla G, Singh D, Sharma L, Koul A, Rishi P. 2009. Effect of Plasmodium and Salmonella co-infection in a murine model. Central Eur J Med 4:340–347. [Google Scholar]
- 141.Lokken KL, Mooney JP, Butler BP, Xavier MN, Chau JY, Schaltenberg N, Begum RH, Müller W, Luckhart S, Tsolis RM. 2014. Malaria parasite infection compromises control of concurrent systemic non-typhoidal Salmonella infection via IL-10-mediated alteration of myeloid cell function. PLoS Pathog 10:e1004049. doi: 10.1371/journal.ppat.1004049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Raffatellu M, Santos RL, Verhoeven DE, George MD, Wilson RP, Winter SE, Godinez I, Sankaran S, Paixao TA, Gordon MA, Kolls JK, Dandekar S, Baumler AJ. 2008. Simian immunodeficiency virus-induced mucosal interleukin-17 deficiency promotes Salmonella dissemination from the gut. Nat Med 14:421–428. doi: 10.1038/nm1743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Coombes BK, Coburn BA, Potter AA, Gomis S, Mirakhur K, Li Y, Finlay BB. 2005. Analysis of the contribution of Salmonella pathogenicity islands 1 and 2 to enteric disease progression using a novel bovine ileal loop model and a murine model of infectious enterocolitis. Infect Immun 73:7161–7169. doi: 10.1128/IAI.73.11.7161-7169.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]