Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Jul 18.
Published in final edited form as: Vaccine. 2011 May 25;29(32):5094–5106. doi: 10.1016/j.vaccine.2011.05.022

Mouse models to assess the efficacy of non-typhoidal Salmonella vaccines: revisiting the role of host innate susceptibility and routes of challenge

Raphael Simon a,b, Sharon M Tennant a,b, James E Galen a,b, Myron M Levine a,b,c,*
PMCID: PMC3152302  NIHMSID: NIHMS304179  PMID: 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.

Keywords: Salmonella Typhimurium, Salmonella Enteritidis, mouse model, vaccine efficacy, live attenuated vaccine, conjugate vaccine

1. Introduction

Certain serovars of Salmonella enterica sub-species enterica that cause invasive infections in humans pose a public health burden worldwide [1]. Among the most important are the etiologic agents of typhoid and paratyphoid fever, Salmonella Typhi and Salmonella Paratyphi A and B (and occasionally C), often referred to collectively as the enteric fever serovars. S. Typhimurium and S. Enteritidis are the two most common NTS serovars associated with gastroenteritis [2] and invasive disease [3]. Whereas in industrialized countries it has long been recognized that NTS serovars can occasionally cause severe invasive infections accompanied by high case fatality rates when they infect young infants or the elderly, only recently has systematic, blood culture-based surveillance of febrile infants and young children in sub-Saharan Africa revealed an enormous burden of invasive disease associated with NTS. Although many of the surveillance activities in Africa were originally undertaken to quantify the burden of invasive Haemophilus influenzae type b (Hib) and Streptococcus pneumoniae infections, the studies unexpectedly revealed that invasive NTS were as commonly isolated from bacteremic patients as Hib and pneumococcus [413]. Notably, 75–95% of these invasive NTS infections in Africa are due to S. Typhimurium and S. Enteritidis [414]. Four features differentiate the pediatric invasive NTS infections in sub-Saharan Africa from the invasive pediatric NTS infections observed in the USA and Europe. First, the sub-Saharan Africa infections are clinically severe and accompanied by case fatality rates of ~15–20%. Second, two-thirds of the African invasive NTS cases neither present with gastroenteritis nor have a history of gastroenteritis [15]. Third, full genome sequencing of an invasive prototype S. Typhimurium strain from Malawi surprisingly revealed that the strain manifested considerable genomic degradation, including complete loss of some genes and the presence of multiple pseudogenes, some of which are also found in S. Typhi or S. Paratyphi A [16]. Lastly, the few epidemiological investigations carried out so far have failed to identify an animal reservoir for these emerging “invasive” NTS strains [17].

Oral challenge of mice with the human host-restricted serovars S. Typhi and S. Paratyphi does not lead to a productive invasive infection. In contrast, oral challenge of mice with S. Typhimurium and S. Enteritidis generates an invasive, generalized infection of the gut-associated lymphoid tissue and reticuloendothelial system (RES) that recapitulates many aspects of typhoid fever in humans [18, 19]. Accordingly, these “mouse typhoid” models have been used to study Salmonella pathogenesis, to identify attenuating genetic lesions possibly applicable to S. Typhi, and to assess the immunogenicity and efficacy of different candidate vaccines.

The burgeoning interest in developing NTS vaccines to control invasive NTS disease in sub-Saharan Africa has renewed interest in mouse models of S. Typhimurium and S. Enteritidis, since such pre-clinical models may be invaluable for examining the efficacy of candidate NTS vaccines intended for human use.

2. Mouse Models of Salmonella Pathogenesis

Since the pathogenesis of Salmonella infection is reviewed extensively elsewhere [1823], it will only be briefly summarized here. Following oral inoculation in mice, Salmonella bacteria initiate infection in the small intestine through penetration of the Peyer’s patches [24, 25]. Invasion and colonization of the Peyer’s patches is soon followed by migration within phagocytic cells to mesenteric lymph nodes, followed by a primary bacteremia and dissemination to the organs of the reticuloendothelial system (RES) (spleen, liver, bone marrow, etc.), onset of a systemic febrile illness, and, finally death [19, 23]. Mice can also be infected by parenteral inoculation via the intraperitoneal (IP), intravenous (IV) and subcutaneous (SC) routes, resulting in similar patterns of infection of the RES and death [2628]. Of these various routes, inoculation with Salmonella by the oral route most closely resembles the natural route of infection in humans [18, 26], but requires the highest number of viable bacteria in order to induce lethal infection. However, oral infection allows more accurate control of the administered dose, since orally administered Salmonella do not immediately enter a rapid growth phase following inoculation, as can happen with IP infections [18, 20, 26, 28].

3. Literature Search

3.1. Methods

A review of the literature was performed by searching the PubMed database with no time limits, using search terms including “Salmonella”, “Typhimurium”, “Enteritidis”, “vaccine”, “mice”, “porin”, “conjugate”, “live attenuated”, “killed”, “humoral”, “cell mediated”, as well as the references of included papers. Our search was limited to studies published in the English language. To be selected for inclusion, a publication had to provide detailed information on 1) well characterized vaccine candidates; 2) route and schedule of immunization; 3) the degree of virulence of the wild-type Salmonella challenge strain; 4) the interval from last vaccine dose until challenge; 5) size of the challenge inoculum; 6) route of challenge; and 7) genetic background of the mouse strain with defined or evident NTS susceptibility.

4. The Vaccines Used to Prevent Fatal Salmonella Typhimurium and Salmonella Enteritidis Infections in Mice

4.1 Live attenuated vaccine strategies

Live attenuated strains based on lesions in genes encoding products in bacterial metabolic, signaling, and gene regulation pathways have formed the basis of several live attenuated Salmonella vaccine strains that have shown marked vaccine efficacy in mouse challenge experiments [29]. The mutated genes encode aromatic amino acid biosynthesis (aroA, aroC, aroD), galactose metabolism (galE), heat shock proteins (htrA), guanine nucleotide biosynthesis (guaA, guaB), purine nucleotide biosynthesis (purA, purB, purE, purH), adenylate cyclase signaling (cya, crp), two-component regulatory systems (phoP, phoQ), regulators of protein metabolism (clpP, clpX), outer membrane proteins (ompC, ompF, ompR), DNA recombination and repair pathways (recA, recB, recC), dissemination to deep organs of the reticuloendothelial system (cdt) and transcriptional regulators of gene expression (dam) [2938].

4.2 Inactivation and subunit strategies

Killed whole cell vaccines prepared by a variety of methods, including thermal inactivation, or chemical inactivation with acetone, deoxycholate, or formalin, have been tested in mice. In an effort to target individual Salmonella antigens to avoid the reactogenicity associated with parenteral administration of inactivated whole cell Salmonella vaccines, candidate subunit vaccines have been developed. These include protein (purified porins, bulk outer membrane proteins, flagellin protein), lipopolysaccharide or O-polysaccharide and Salmonella O-polysaccharide-protein conjugate vaccines. Conjugate vaccines based on chemical covalent linkage of the Salmonella O-polysaccharide (OPS) with a protein carrier have been utilized to enhance the immunogenicity of the otherwise weakly immunogenic polysaccharide hapten, and provide functional immunological memory [39].

5. Sources of Variability in Murine Models that Test the Efficacy of Salmonella Typhimurium and Salmonella Enteritidis Vaccines

5.1 Importance of the strain of mouse

The development of the mouse model of lethal S. Typhimurium and S. Enteritidis infections has provided a robust platform for testing experimental vaccines against these two serovars and allowed considerable progress to be made towards understanding the immune responses that can mediate protection against these pathogens [19, 23, 4042]. Nevertheless, the systematic analysis that we undertook of the published scientific literature on S. Typhimurium and S. Enteritidis vaccines evaluated in murine models reveals considerable variability and often contradictory results (Table 1, 2). We found that the genetic background of the mouse strain and the virulence of the wild-type Salmonella challenge strain stood out, strikingly, as the two most important fundamental variables [4348], aside from expected differences in efficacy stemming from the type of vaccine. Indeed, since several commonly used mouse strains differ so markedly in their natural susceptibility to Salmonella infection, they are classified as either inherently “resistant” or “susceptible” [46, 4953]. A defining phenotypic trait of susceptible mouse strains is the dramatic reduction in the lethal dose of a given Salmonella strain required to kill 50% of infected mice (LD50), which can be > 1,000-fold lower as compared to resistant mice (Table 3).

Table 1.

Vaccine efficacy mediated by living and non-living NTS vaccines in susceptible mice

Vaccine type Mouse strain Vaccine (dose) Route of vaccination Number of doses Duration till challenge Route of challenge Challenge strain Reported LD50 Challenge Inoculum size Vaccine Efficacya Reference
Live BALB/c Live attenuated dam S. Typhimurium (109 cfu) Oral 1 5 weeks Oral S. Typhimurium 14028 1 × 105 10,000 × LD50 100% [32, 33]
Live BALB/c Live attenuated crp,cdt S. Typhimurium SL1344 (109 cfu) Oral 1 30 days Oral S. Typhimurium SL1344 6 × 104 > 10,000 × LD50 100% [30]
Live BALB/c Live attenuated crp,cdt S. Typhimurium UK-1 (109 cfu) Oral 2 30 days Oral S. Typhimurium UK-1 8.5 × 103 >10,000 × LD50 100% [30]
Live BALB/c Delayed attenuated phoPQ,crp,rpoS,fur S. Typhimurium (~109 cfu) Oral 1 30 days Oral S. Typhimurium UK-1 1.2 × 104 10,000 × LD50 100% [31]
Live BALB/c Live attenuated aroA S. Typhimurium SL3261 (1010 cfu) Oral 1 8 weeks Oral S. Typhimurium C5 1 × 106 10,000 × LD50 100%c [63]
Live C57Bl/6 Live attenuated aroA S. Typhimurium SL3261 (109 cfu) Oral 1 6 weeks Oral S. Typhimurium SL1344 NRb 1 × 105 cfu 100% [64]
Live BALB/c Delayed attenuated phoPQ,crp,rpoS,fur S. Typhimurium (104 cfu) IP 1 30 days Oral S. Typhimurium UK-1 1.2 × 104 10,000 × LD50 100% [31]
Live BALB/c Live attenuated aroA S. Typhimurium SL3261 (106 cfu) IV 1 8–12 weeks Oral S. Typhimurium C5 1 × 106 10,000 × LD50 100%c [63, 65, 66]
Live BALB/c Live attenuated aroA Se795 S. Enteritidis (106 cfu) IV 1 3 months Oral S. Enteritidis Thirsk 1 × 106 10,000 × LD50 100%c [65]
Live C57Bl/6 Live attenuated aroA S. Typhimurium SL3261 (5 × 105 cfu) IV 1 4 months Oral S. Typhimurium C5 1 × 107 250 × LD50 100% [67]
Live BALB/c Live attenuated aroA S. Typhimurium SL3261 (106 cfu) SC 1 8 weeks Oral S. Typhimurium C5 1 × 106 1,000 × LD50 100%d [63]
Live BALB/c Live attenuated aroA S. Typhimurium SL3261 (5 × 105 cfu) IV 1 6 weeks IP S. Typhimurium SL1344 <10 104 cfu 100% [68]
Live C3HeB/FeJ Live attenuated aroA S. Typhimurium SL3235 (7 × 104 cfu) IP 1 21 days IP S. Typhimurium W118-2 1 cfu > 1,000 × LD50 100% [57]
Live C3H/HeJ Live attenuated aroA S. Typhimurium SL3235 (2.5 × 105 cfu) IP 1 21 days IP S. Typhimurium W118-2 <7 cfu > 1,000 × LD50 100% [47, 57]
Non-living BALB/c Acetone-inactivated attenuated aroA S. Typhimurium SL3261 (108 cfu) SC 2 8 weeks Oral S. Typhimurium C5 1 × 106 10 × LD50 25% [63]
Non-living C57Bl Heat-inactivated S. Enteritidis 5694 (108 cfu) +CFA SC 2 2 weeks Oral S. Enteritidis 5694 5 × 103 2 × LD50 100%e [69, 70]
Non-living C57Bl Heat-inactivated S. Enteritidis 5694 (108 cfu) SC 2 2 weeks Oral S. Enteritidis 5694 5 × 103 2 × LD50 0%f [69, 70]
Non-living C57Bl/6 Flagellin (200 μg) SC 2 1 week Oral S. Typhimurium SL1344 NR 1 × 105 cfu 70% [64]
Non-living BALB/c Porin (50 μg) IP 1 3 weeks IP S. Typhimurium LT2 1 × 105 1,000 × LD50 100%d [71, 72]
Non-living BALB/c OPS-Tetanus Toxoid Conjugate (10 μg) SC 3 1 week IP S. Typhimurium PTCC 1733 1.3 × 104 77 × LD50 50% [73]
Non-living BALB/c OPS (10 μg) SC 3 1 week IP S. Typhimurium PTCC 1733 1.3 × 104 77 × LD50 12.5% [73]
Non-living BALB/c Heat-inactivated S. Typhimurium PTCC 1735 (108 cfu) SC 3 1 week IP S. Typhimurium PTCC 1733 1.3 × 104 77 × LD50 37.5% [73]
Non-living C3H/HeJ Acetone-inactivated S. Typhimurium W118-2 (60 μg) IP 2 21 days IP S. Typhimurium W118-2 <7 cfu 6 cfu 36% [46]
Non-living C3H/HeJ Crude LPS (100 μg) IP 2 21 days IP S. Typhimurium W118-2 <7 cfu 6 cfu 21% [46]
Non-living C3H/HeJ Purified LPS (100 μg) IP 2 21 days IP S. Typhimurium W118-2 <7 cfu 6 cfu 0% [46]
Non-living C3HeB/FeJ Acetone-inactivated S. Typhimurium W118-2 (60 μg) IP 2 21 days IP S. Typhimurium W118-2 1 cfu 24 cfu 100% [46]
Non-living C3HeB/FeJ Crude LPS (100 μg) IP 2 21 days IP S. Typhimurium W118-2 1 cfu 24 cfu 50% [46]
Non-living C3HeB/FeJ Purified LPS (100 μg) IP 2 21 days IP S. Typhimurium W118-2 1 cfu 24 cfu 10% [46]
Non-living C57Bl/6 Porin (20 μg) IP 2 35 days IP S. Typhimurium SL1344 NR 3 × 103 cfu 0%g [74]
Non-living C57Bl/6J Phenol-inactivated S. Typhimurium (3 × 107 cfu) IP 6 7 days IP S. Typhimurium C5 <10 101 – 107 cfu 0% [54]
Non-living BALB/cJ Phenol-inactivated S. Typhimurium (3 × 107 cfu) IP 6 7 days IP S. Typhimurium C5 <10 101 – 107 cfu 0% [54]
Non-living C3H/HeJ Phenol-inactivated S. Typhimurium (3 × 107 cfu) IP 6 7 days IP S. Typhimurium C5 <10 101 – 107 cfu 0% [54]
Non-living DBA/2J Phenol-inactivated S. Typhimurium (3 × 107 cfu) IP 6 7 days IP S. Typhimurium C5 <10 101 – 107 cfu 0% – 10% [54]
Non-living BALB/c Porin (100 μg) SC 2 10 days IV S. Typhimurium C5 1 × 105 10 × LD50 25% [76]
Non-living BALB/c LPS (50 μg) SC 2 10 days IV S. Typhimurium C5 1 × 105 10 × LD50 35.7% [76]
Non-living BALB/c Porin (100 μg) + LPS (50 μg) SC 2 10 days IV S. Typhimurium C5 1 × 105 10 × LD50 100% [76]
Non-living BALB/c Outer-membrane Protein (50 μg) SC 2 10 days IV S. Typhimurium C5 1 × 105 50 × LD50 100% [75]
a

((Mortality in controls–Mortality in vaccine group) / Mortality in controls) × 100

b

Not reported

c

The reported LD50 is increased > 10,000 fold as compared to unimmunized mice, it is assumed then that 100% mortality in controls, and 0% mortality in vaccinated animals at 10,000 × LD50 challenge

d

The reported LD50 is increased > 1,000 fold as compared to unimmunized mice, it is assumed then that 100% mortality in controls, and 0% mortality in vaccinated animals at 1,000 × LD50 challenge

e

Mortality not reported, however of the remaining mice not sacrificed for analysis by day 15, none had died and a significant decrease in NTS tissue burden was reported

f

Mortality not reported, however, of the mice not sacrificed for analysis by day 15, several had died, and the NTS tissue burden reported was comparable to unvaccinated controls

g

Mortality not reported, the lack of protection from mortality however was stated in the results.

Table 2.

Vaccine efficacy mediated by living and non-living NTS vaccines in resistant mice

Vaccine type Mouse strain Vaccine (dose) Route of vaccination Number of doses Duration till challenge Route of challenge Challenge strain Reported LD50 Challenge Inoculum size Vaccine Efficacya Reference
Live C3H/HeNCrlBR Live attenuated aroA S. Typhimurium SL3235 (7 × 104 cfu) IP 1 21 days IP S. Typhimurium W118-2 1 × 103 1,000 × LD50 100% [47]
Live CF1 Live attenuated rough mutant S. Typhimurium SL1004 (6 × 106 cfu) IP 1 14 days IP S. Typhimurium LT2 5 × 102 1,000 × LD50 100% [77]
Live ddY Live attenuated rough mutant S. Typhimurium SL1004 (6 × 106 cfu) IP 1 14 days IP S. Typhimurium LT2 1 × 103 1,000 × LD50 100% [77]
Non-living A/J Phenol-inactivated S. Typhimurium (3 × 107 cfu) IP 6 7 days IP S. Typhimurium C5 ~ 104 107 cfu 90% [54]
Non-living C3H/HeNCrlBR Acetone-killed S. Typhimurium W118-2 (60 μg) IP 2 21 days IP S. Typhimurium W118-2 1.2 × 103 19 × LD50 100% [46]
Non-living C3H/HeNCrlBR Crude LPS (100 μg) IP 2 21 days IP S. Typhimurium W118-2 1.2 × 103 19 × LD50 100% [46]
Non-living C3H/HeNCrlBR Purified LPS (100 μg) IP 2 21 days IP S. Typhimurium W118-2 1.2 × 103 19 × LD50 89% [46]
Non-living CBA x C57Bl/6 Porin (10 μg) IP 2 10 days IP S. Typhimurium SH6739 1 × 105 27 × LD50 86% [78]
Non-living CD-1 Acetone-inactivated S. Typhimurium W118-2 (108 cfu) IP 1 21 days IP S. Typhimurium W118-2 1 × 104 1,000 × LD50 100%b [43]
Non-living CD-1 LPS (25 μg) IP 1 21 days IP S. Typhimurium W118-2 1 × 104 500 × LD50 100%b [43]
Non-living CF1 LPS (100 μg) IP 1 14 days IP S. Typhimurium LT2 5 × 102 1,000 × LD50 0% [77]
Non-living CF1 Formalin-inactivated S. Typhimurium P173C (109 cfu) IP 4 21 days IP S. Typhimurium P173C < 102 1,000 × LD50 100% [79]
Non-living CF1 Formalin-inactivated S. Enteritidis Br1203bM (109 cfu) IP 4 21 days IP S. Enteritidis 203 102 > 1,000 × LD50 100% [79]
Non-living ddY LPS (100 μg) IP 1 14 days IP S. Typhimurium LT2 1 × 103 1,000 × LD50 0% [77]
Non-living NMRI OPS-Porin Conjugate (5 μg) IP 4 6 days IP S. Typhimurium SH 2201 4.4 × 104 200 × LD50 90% [80]
Non-living NMRI Porin (5ug) IP 4 6 days IP S. Typhimurium SH 2201 4.4 × 104 200 × LD50 70% [80]
Non-living White Swiss Heat-inactivated S. Typhimurium Suc EE (108 cfu) IP 1 10 days IP S. Typhimurium Suc LL 5 × 104 100 × LD50 100% [81]
Non-living White Swiss Heat-inactivated S. Typhimurium Suc EE (250 μg) IP 3 NRc IV S. Typhimurium Suc LL 1 × 106 100 × LD50 100% [82]
Non-living CD-1 OPS-Tetanus Toxoid conjugate (2.5 μg) SC 3 1 week IP S. Typhimurium W118 1.9 × 104 100 × LD50 100% [39]
Non-living White Swiss Porin (50 μg) SC 2 10 days IP S. Typhimurium LT2-71 1.9 × 103 100 × LD50 100% [83]
Non-living White Swiss LPS (20 μg) SC 2 10 days IP S. Typhimurium LT2-71 1.9 × 103 100 × LD50 50% [83]
Non-living White Swiss Ethyl-inactivated S. Enteritidis 5694 (3 × 106 cfu) IP, IV, SC 3 1 week IP, IV, SC S. Enteritidis 5694 3.5 × 102 (IV) 100 – 1,000 × LD50 11% – 60% [27, 28]
a

((Mortality in controls – Mortality in vaccine group) / Mortality in controls) × 100

b

Mortality in controls not reported, 100% mortality in unvaccinated controls is assumed however at the high challenge dose reported

c

Not reported

Table 3.

Reported comparative natural resistance to individual NTS isolates in commonly used mouse strainsa

Reported Mouse NTS susceptibility phenotype Mouse Strain Reference(Salmonella strain used)
Hormaeche, 1979 [50] (S. Typhimurium C5) Plant and Glynn, 1976 [53] (S. Typhimurium C5) Collins, 1972 [52] (S. Enteritidis 5694)
IV LD50 (LOG 10) SC LD50 (LOG 10) SC LD50 IV LD50 Oral LD50
Susceptible BALB/c 1.27 < 1 < 10
Susceptible DBA/1 < 10
Susceptible C57Bl 2 × 101 5 – 10 5 × 103
Susceptible B10 1.26 < 1
Susceptible B10.D2 < 1 < 1 5 × 101
Susceptible B10.A 1.5 < 1
Susceptible B10.Br < 1 < 1
Resistant DBA/2 1.6 NDb 2×105
Resistant C3H 2.1 ND 1 × 106
Resistant CBA 3.0 6.9 1 × 107
Resistant A/J 4.26 ND 4 × 102 5 × 105
Resistant A/JAX 2 × 106
Resistant F1 (BALB/C x CBA) 3 × 106
Resistant F1 x CBA 9 × 106
Resistant CD-1 3 × 103 2 × 106
Resistant CF-1 5 × 103 6 × 106
a

Reproduced from indicated reports

b

Not done

The use of susceptible mice in NTS vaccine development offers several advantages, as well as disadvantages. Of notable importance, the significantly lowered LD50 with virulent NTS in these mice allows a wider range of challenge inocula to be tested. This is particularly pertinent to the oral challenge model, which necessitates a much larger inoculum size that can be on the order of 1,000-fold greater than that required to achieve similar lethality with parental challenge (Table 3). Moreover, in resistant mice it may not be possible to achieve a lethal dose following oral challenge if a weakly virulent NTS strain is given. On the other hand, a sub-lethal infection with highly virulent strains of S. Typhimurium and S. Enteritidis administered parenterally cannot be established in susceptible mice, as the LD50 for parenteral challenge is frequently less than 20 colony forming units (cfu) [46, 49, 50, 5254]. A final point to consider is that inbred susceptible mice do not model accurately the genetic heterogeneity of human populations.

5.2 Genetic basis of mouse susceptibility to Salmonella

Mutations in several important genetic loci in mice have identified susceptibility factors for infection with Salmonella, and are associated with significant lowering of the LD50 [23, 4951, 53]. Of these, NTS susceptibility loci originally referred to as Immunity to Typhimurium (Ity) and resistance to Lipopolysaccharide (Lps), involve genes associated with innate immunity. The Ity locus contains the Natural resistance associated macrophage protein 1 gene (Nramp1) which encodes an intracellular endosome-associated pH-controlled ion transport protein, present within resident peritoneal and splenic macrophages. A mutation in this gene found in susceptible mice leads to defects in phagocyte endolysosome function that impairs control of intracellular infections and increases susceptibility to lethal Salmonella infection [55, 56]. Mutations in the Lps locus were originally identified in the C3H/HeJ mouse strain, a spontaneous mutant derived from the C3H line, that is insensitive to the toxic effects of bacterial lipopolysaccharide (LPS) [57]. The inability to respond to LPS, an important Salmonella pathogen associated molecular pattern, causes these mice to become highly susceptible to Salmonella infection, as they are unable to mount an adequate innate immune response to control the early stages of infection. The mouse Lps locus is associated with a mutation in the gene for Toll-like Receptor 4 (TLR4), the innate immune receptor for LPS, that renders it nonfunctional for signaling [5860]. The broad importance of LPS recognition in natural Salmonella immunity has been further confirmed, as defects in the endotoxin recognition system including LPS Binding Protein (LBP), and CD-14 have also been associated with Salmonella susceptibility [23, 61, 62].

6. Immunization of Susceptible Mouse Strains with Live Attenuated NTS Vaccines and Protection against NTS Challenge

6.1 Oral immunization and oral challenge

As summarized in Table 1, every report of susceptible mice (e.g., BALB/c, C57Bl/6) immunized orally with S. Typhimurium attenuated mutants has demonstrated a high level of protection when the mice were subsequently challenged orally with wild type S. Typhimurium [3033, 63, 64]. This is true for an array of live vaccines carrying different attenuating mutations. Moreover, in most studies the oral challenge was rigorous and contained > 10,000 × LD50 of virulent S. Typhimurium [30, 32, 33].

6.2 Oral immunization and parenteral challenge

The literature search was unable to identify reports describing susceptible mice immunized orally with attenuated S. Typhimurium or S. Enteritidis vaccines that were then challenged parenterally with wild type organisms.

6.3. Parenteral immunization and oral challenge

There are multiple reports of experiments with susceptible mice immunized with a variety of attenuated S. Typhimurium or S. Enteritidis vaccines by parenteral routes (IP, IV or SC) and were then challenged orally [31, 57, 63, 6567]. In all instances a high level of vaccine efficacy was observed (Table 1).

6.4. Parenteral immunization and parenteral challenge

There are also several reports of susceptible strains of mice that were immunized parenterally (IP or IV) with attenuated S. Typhimurium or S. Enteritidis vaccines and were then shown to be highly protected when subsequently challenged with wild type NTS administered IP (Table 1) [47, 57, 68].

7. Immunization of Susceptible Mouse Strains with Non-Living NTS Vaccines and Protection against Challenge with Virulent NTS

7.1. Oral immunization followed by oral or parenteral challenge

Our search uncovered no reports of susceptible mice immunized orally with non-living S. Typhimurium or S. Enteritidis vaccines who were then challenged with the homologous wild type serovar given either orally or parenterally.

7.2. Parenteral immunization followed by oral challenge

Several reports describe the parenteral immunization of susceptible strains of mice (e.g., BALB/c, C57Bl, C57Bl/6) with chemical or heat-inactivated whole cell S. Typhimurium or S. Enteritidis vaccines or with a subunit vaccine (flagellin) followed by oral challenge with low doses of virulent organisms (Table 1). The only vaccine or regimen that achieved a high level of protection against oral challenge was when inactivated S. Enteritidis were administered with Complete Freunds Adjuvant (CFA) and the challenge dose was low and near the LD50 [69, 70]. The only other hint of protection in the face of oral challenge was observed following SC immunization of C57Bl/6 mice with S. Typhimurium flagellin protein FliC; weak protection was recorded against oral challenge with a low dose of S. Typhimurium [64].

7.3. Parenteral immunization followed by parenteral challenge

As summarized in Table 1, non-living S. Typhimurium vaccines have provided generally disappointing and inconsistent results in instances where both the vaccine and the challenge were administered parenterally to susceptible strains of mice (Table 1). With only one exception [46], parenteral immunization with inactivated whole cell vaccines have failed to protect susceptible mice against parenteral challenge with virulent NTS. Those few inactivated parenteral vaccines that did confer measurable protective efficacy against wild type challenge involved immunization with subunit vaccines, followed by challenge with weakly virulent Salmonella strains (parenteral LD50 > 10,000 cfu) or low inocula of virulent strains [46, 54, 7176].

8. Immunization of Resistant Mouse Strains with Live Attenuated NTS Vaccines and Protection against Challenge

8.1. Oral immunization followed by oral or parenteral challenge

We did not find any reports of resistant strains of mice immunized orally with attenuated NTS vaccines and then challenged, either orally or parenterally.

8.2 Parenteral immunization followed by parenteral challenge

Our search revealed that resistant mice immunized by the IP route with live attenuated aroA and a rough LPS mutant of S. Typhimurium, were protected against subsequent IP challenge with 1,000 × LD50 of virulent S. Typhimurium (Table 2) [47, 77].

9. Immunization of Resistant Mouse Strains with Non-Living NTS Vaccines and Protection against Challenge

9.1 Oral immunization followed by either oral or parenteral challenge

Our search failed to reveal any reports of oral immunization of resistant mice with non-living S. Typhimurium or S. Enteritidis vaccines followed by either oral or parenteral challenge with wild type organisms where mortality was reported. In the one study, CD-1 mice immunized orally or IM with heat-killed S. Enteritidis showed a slight decrease in tissue cfu burden following oral challenge with highly virulent S. Enteritidis. However, this would be unlikely to translate to a decrease in mortality [26].

9.2. Parenteral immunization and parenteral challenge

Resistant mice have proven to be a robust model for testing the efficacy of a panoply of non-living S. Typhimurium and S. Enteritidis vaccines, including inactivated whole cell, subunit and polysaccharide-protein conjugates, in preventing mortality following challenge with NTS (Table 2). Several publications describe parenteral (IP or IV) immunization of resistant mice (e.g., C3H/HeNCrIBR, CD-1, CF1, ddy, White Swiss), followed by parenteral (primarily IP) challenge with virulent S. Typhimurium or S. Enteritidis (Table 2) [27, 39, 43, 46, 54, 7783]. In all reports a high level of protection was observed, even with challenge inocula containing wild type organisms equivalent to 1,000 × LD50 (Table 2). These reports contrast with the highly variable protection reported when susceptible mice were challenged after being immunized with non-living vaccines.

10. Role of Mouse Salmonella Susceptibility Gene Alleles and Vaccine Efficacy

The variability in protective efficacy seen with non-living vaccines in different strains of mice led to considerable confusion in the field, for a period of time. The situation was clarified when studies to address this conundrum established, unequivocally, that the background murine genetic Salmonella susceptibility locus exerts a powerful influence on the observed efficacy of non-living vaccines [46, 54]. In one key study conducted by Robson and Vas, inbred mouse strains that vary in innate susceptibility to infection with NTS were vaccinated IP with phenol-inactivated S. Typhimurium and then challenged by the IP route with graded inocula of S. Typhimurium ranging from 101 to 107 cfu [54]. Strikingly, highly susceptible (C57Bl/6J, BALB/cJ, C3H/HeJ) and moderately susceptible (DBA/2/J) mice immunized with whole-cell killed-vaccine were not significantly protected against fatal S. Typhimurium infection at any challenge dose tested. In contrast, resistant (A/J) mice were highly protected against lethal Salmonella challenge at all levels tested. Notably, both naïve and vaccinated highly susceptible mice succumbed to infection with less than 10 S. Typhimurium cfu, whereas infection in naïve resistant A/J mice was consistently fatal only at 105 cfu, and immunized mice were fully protected at 107 cfu.

Eisenstein et al. immunized mice of the C3H lineage that vary in inherent susceptibility to Salmonella by the IP route with either whole-cell killed vaccine or purified LPS and then challenged the mice by IP inoculation with ~ 20 × LD50 of virulent S. Typhimurium [46]. Naturally resistant C3H/HeNCrlBR mice were fully protected, whereas highly susceptible C3H/HeJ mice (that are non-responsive to bacterial LPS) showed no significant protection against challenge. C3HeB/FeJ mice that are innately responsive to LPS but nonetheless more highly susceptible to S. Typhimurium, as compared to traditionally resistant mice, displayed an intermediate protection phenotype; partial protection was observed compared to resistant C3H/HeNCrlBR mice. This study was the first to demonstrate that the differential performance of non-living vaccines in different mouse strains that vary in natural susceptibility to wild-type Salmonella infection is consistent across different types of vaccine preparations.

11. Mechanisms of protective immunity to S. Typhimurium and S. Enteritidis in mice

11.1 Humoral immunity

The extensive work conducted towards understanding the mechanistic basis of the protective adaptive immune response to Salmonella has been reviewed elsewhere [23, 34, 84] and will be discussed here only briefly, with an emphasis on mechanisms of immunity in susceptible versus resistant strains of mice. Immunization with non-living vaccines can stimulate the production of humoral immunity, including antibody against Salmonella outer membrane antigens and flagellin that can provide protection against extracellular bacteria. Non-living vaccines, in contrast, stimulate comparatively negligible cytotoxic T-cell mediated cellular immunity [84]. Mucosal IgA plays an important role against oral infection with Salmonella as a first line of defense at the mucosal epithelial surface, through inhibition of bacterial penetration into the Peyer’s patches [85, 86]. Protection by circulating serum antibody can potentially play a role in inhibiting cell-to-cell transmission of Salmonella following exit from infected cells, as well as a reduction in overall systemic bacterial cell burden [68, 8789].

The systemic immune responses mediated by antibody to Salmonella include direct complement mediated cytocidal activity by IgM and IgG, as well as opsonophagocytic uptake into professional phagocytes such as macrophages, neutrophils, and dendritic cells [88]. Of these, antibody-mediated opsonophagocytic uptake, which is associated with secretion of pro-inflammatory cytokines and intracellular oxidative burst, is likely more important in vivo as natural resistance to serum and complement-mediated bactericidal activity may be common among invasive non-typhoidal Salmonella [90]. Killing by antibody-mediated opsonophagocytosis is effective against complement-resistant strains, and opsonophagocytosis is important for presentation of antigenic peptides and activation of CD4+ T-helper cells [79, 90]. Surface bound antibody on B-cells can also mediate internalization and processing of bacterial cells, for subsequent presentation of Salmonella-specific peptide epitopes on MHC-II molecules to T-cells [91]. Presentation of Salmonella antigens by antibody mediated opsonophagocytosis can aid in the development of cell-mediated immunity, that may not progress normally otherwise, as Salmonella outer membrane polysaccharides are known to interfere with non-specific opsonophagocytic uptake [92, 93]. The importance of antibody-mediated protection against Salmonella has been demonstrated in genetic models of B-cell deficiency. In these studies, transgenic derivatives of the susceptible C57Bl/6 mouse strain, defective for the generation of B-cells, were unable to mount a protective immune response against virulent Salmonella Typhimurium following vaccination with a live attenuated aroA mutant. Furthermore, greatly diminished T-cell activation was observed in these B-cell deficient mice, and full protection was not restored by passive immunization with anti-Typhimurium sera, further underscoring the important role that antibody and B-cells play in the generation of cellular immunity [67, 94].

11.2. Cellular immunity

Immunization with live attenuated non-typhoidal Salmonella vaccine strains elicits robust both humoral immunity and cellular immunity that is important for the control of intracellular infection [84]. Cellular immunity against Salmonella involves the coordinated action of CD4+ T helper and CD8+ Cytotoxic T lymphocytes (CTL) against Salmonella proteins [84, 95, 96]. Cellular responses against Salmonella proteins have been demonstrated for several Salmonella protein antigens including, for example, flagellin FliC [34, 64, 9799]. T-helper type I immunity, mediated by tumor necrosis factor-α, (TNF-α), interleukin-12 (IL-12) and interferon-γ (IFN-γ), that includes the action of activated macrophages, subsets of CD8+ and CD4+ T-cells, and opsonophagocytic antibodies, is strongly associated with the protective immune response to Salmonella [95, 100103]. Vaccination of mice with live attenuated strains of Salmonella has been demonstrated to elicit potent cellular immunity as characterized by the production of high levels of IFN-γ upon ex-vivo restimulation of immune T-cells with Salmonella antigens [64, 98, 104]. Indeed, observations from both mouse and human studies have highlighted the overt susceptibility associated with perturbation of the IL-12/IFN-γ axis towards infection with Salmonella [61, 105108]. Allelic specificity of the Major Histocompatibility Complex-II (MHC-II) has also been implicated as important for clearance of Salmonella infection in both mice and humans [34, 109, 110].

11.3. Importance of antibody and T-cells to immunity against NTS

Elucidation of the comparative contribution of humoral and cellular immunity in functional protection against Salmonella infection in highly susceptible mice has been demonstrated in several published reports. Adoptive transfer models of immunity with immune serum and T-cells from mice immunized with live attenuated aroA Salmonella Typhimurium, into susceptible naïve BALB/c mice, has provided evidence that both humoral and cellular immunity are necessary for immunity to oral challenge with virulent Salmonella Typhimurium. In this model, naïve mice receiving either immune serum or T-cells separately, succumbed to oral infection, whereas mice receiving both were protected [66]. Further studies with this model have highlighted the importance of O-specific antibody against bacterial polysaccharide as a functional correlate of protection [65]. Separate studies with susceptible BALB/c mice genetically ablated for B-cell responses, has further confirmed the dual requirement for both antibody and T-cells in the IP model of Salmonella Typhimurium challenge. In these experiments, partial protection against challenge was seen following passive transfer of anti-Salmonella Typhimurium immune serum into B-cell deficient BALB/c mice that had been previously immunized with live attenuated aroA Salmonella Typhimurium. Protection however was not seen following transfer of immune serum into naïve mice, or in aroA Salmonella Typhimurium vaccinated B-cell deficient mice [68].

Evidence for antibody-mediated protection against parenteral challenge with highly virulent NTS in resistant mice is provided in several published reports. Protection from parenteral challenge mediated by immunization with killed whole cell Salmonella Typhimurium in resistant White Swiss mice is mirrored by the rise in titer of functional bactericidal antibody [81, 82]. In this model, the substantial protection observed following challenge is similarly associated with a large reduction of Salmonella CFU in the liver and spleen as compared to unvaccinated mice and can be seen as early as six hours after challenge. Resistant CD-1 and NMRI mice immunized with O-polysaccharide based conjugate vaccines were also shown to produce high levels of functional opsonophagocytic antibody that could transfer protection by passive immunization into naïve hosts [39, 80, 111113]. As further evidence for the protective role of antibody in resistant mice, passively transferred monoclonal IgG and IgM specific for Salmonella Typhimurium OPS mediated significant protection against IP challenge with LD100 levels of Salmonella Typhimurium in resistant C3H/HeN mice [114]. By comparison, the failure of passive transfer of these same monoclonal IgG and IgM anti-OPS antibodies to provide protection against IP challenge with LD100 levels of S. Typhimurium in susceptible C3H/HeJ mice [114], underscores the limitations of antibody-mediated protection alone in susceptible mice.

Based on results from these mechanistic and functional immunological studies in susceptible and resistant mice immunized with living as compared to non-living NTS vaccines, several important points emerge. In susceptible mice challenged by either the parenteral or oral route with highly virulent NTS strains, the coordinated action of both humoral as well as cellular immunity is required for effective protection from Salmonella Typhimurium challenge. This is likely a result of the failure to adequately control intracellular infection for example in Nramp1 deficient mice [23, 56]. The contribution of humoral immunity alone to protection by vaccines against NTS in mice is generally only seen in host-pathogen interactions that can result in a sub-lethal infection, such as can be produced in either resistant mice challenged with either highly or weakly virulent NTS strains, or susceptible mice challenged with weakly virulent NTS strains. Under these conditions, functional bactericidal and opsonophagocytic antibody directed against NTS likely causes an immediate reduction in Salmonella challenge inoculum present extracellularly, that functions to lower a fatal Salmonella dose to a level that is sub-lethal [81, 82]. This sub-lethal infecting population becomes disseminated to secondary sites of infection, and may also potentially generate cell-mediated immunity akin to that elicited during a natural sub-lethal infection in naïve animals [26, 43, 69, 81, 82, 97]. As highly susceptible mice are impaired for control of Salmonella infection, and succumb to very low parenteral challenge doses, they are virtually unable to be infected at sub-lethal levels with fully virulent strains, and thus protection by antibody is likely not significant [54].

12. Conclusions

The mouse model is expected to play an invaluable role in accelerating the development of safe and effective vaccination strategies against recently emerging NTS that are capable of causing invasive infections in humans. As emphasized in this review, choice of a mouse strain is of fundamental importance among the variables that affect the experimental outcome of investigational NTS vaccines.

The choice of mouse strain used for testing NTS vaccines should be tailored to the expected mechanism of protective immunity generated and the background virulence of the proposed NTS challenge strain. For vaccines designed to elicit both humoral and cellular immunity, such as live attenuated strains, both resistant as well as susceptible mice can be used to test for protection against highly virulent (as well as weakly virulent) NTS challenge strains. Efficacy against highly virulent NTS strains by vaccines for which the correlate of protection is expected to be antibody may be better studied in resistant mice. Thus, parenteral NTS conjugate vaccines, for example, should be tested for efficacy in resistant strains of mice.

Based on the findings revealed in our review, another consideration in selecting the appropriate mouse model for testing the efficacy of NTS vaccines is the selection of the route of challenge. Oral challenge, which resembles the natural route of infection in humans, can be accomplished in either susceptible or resistant mice, in testing live oral vaccines, with the caveat that the LD50 in resistant mice challenged with weakly virulent NTS strains may be so high as to pose a practical obstacle to achieving lethality in control animals.

Our review uncovered no examples where parenteral non-living NTS vaccines found to protect resistant mice against mortality when challenged parenterally were also tested for their ability to protect against oral challenge. We identify this as an important knowledge gap to be filled. If parenteral NTS vaccines under development, such as conjugates, porins and common proteins, can be shown to protect resistant mice against oral challenge, this will be an important breakthrough for generating relevant pre-clinical data for regulatory agencies.

Two vaccine strategies that have proved useful for developing successful human vaccines against S. Typhi are attenuated strains that serve as live oral vaccines [35, 115] and parenteral conjugates consisting of capsular Vi polysaccharide linked to a carrier protein [116118]. Not surprisingly, these same strategies are being followed to develop safe and effective vaccines against invasive S. Typhimurium and S. Enteritidis disease. Carefully selected mouse models will be particularly critical for non-living NTS vaccine development in order to generate relevant pre-clinical data for regulatory agencies that includes evidence of vaccine efficacy.

Acknowledgments

Funding: RS was supported by NIH T32 AI07524 Fellowship Training Program in Vaccinology (MML, PI). MML, RS, SMT and JEG also received support from Middle Atlantic RCE Program, NIAID/NIH 2 U54 AI057168.

Abbreviations

NTS

Non-Typhoidal Salmonella

LPS

Lipopolysaccharide

IP

intraperitoneal

IV

intravenous

SC

subcutaneous

IM

intramuscular

Footnotes

Conflict of interest: The authors declare no conflict of interest with regard to this manuscript.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Ivanoff B, Levine MM, Lambert PH. Vaccination against typhoid fever: present status. Bull World Health Organ. 1994;72(6):957–71. [PMC free article] [PubMed] [Google Scholar]
  • 2.Preliminary FoodNet data on the incidence of infection with pathogens transmitted commonly through food - 10 states, 2009. MMWR Morb Mortal Wkly Rep. 2010 Apr 16;59(14):418–22. [PubMed] [Google Scholar]
  • 3.Vugia DJ, Samuel M, Farley MM, Marcus R, Shiferaw B, Shallow S, et al. Invasive Salmonella infections in the United States, FoodNet, 1996–1999: incidence, serotype distribution, and outcome. Clin Infect Dis. 2004 Apr 15;38( Suppl 3):S149–56. doi: 10.1086/381581. [DOI] [PubMed] [Google Scholar]
  • 4.Berkley JA, Lowe BS, Mwangi I, Williams T, Bauni E, Mwarumba S, et al. Bacteremia among children admitted to a rural hospital in Kenya. N Engl J Med. 2005 Jan 6;352(1):39–47. doi: 10.1056/NEJMoa040275. [DOI] [PubMed] [Google Scholar]
  • 5.Mandomando I, Macete E, Sigauque B, Morais L, Quinto L, Sacarlal J, et al. Invasive non-typhoidal Salmonella in Mozambican children. Trop Med Int Health. 2009 Dec;14(12):1467–74. doi: 10.1111/j.1365-3156.2009.02399.x. [DOI] [PubMed] [Google Scholar]
  • 6.Brent AJ, Oundo JO, Mwangi I, Ochola L, Lowe B, Berkley JA. Salmonella bacteremia in Kenyan children. Pediatr Infect Dis J. 2006 Mar;25(3):230–6. doi: 10.1097/01.inf.0000202066.02212.ff. [DOI] [PubMed] [Google Scholar]
  • 7.Graham SM, Walsh AL, Molyneux EM, Phiri AJ, Molyneux ME. Clinical presentation of non-typhoidal Salmonella bacteraemia in Malawian children. Trans R Soc Trop Med Hyg. 2000 May–Jun;94(3):310–4. doi: 10.1016/s0035-9203(00)90337-7. [DOI] [PubMed] [Google Scholar]
  • 8.Kariuki S, Revathi G, Kariuki N, Kiiru J, Mwituria J, Hart CA. Characterisation of community acquired non-typhoidal Salmonella from bacteraemia and diarrhoeal infections in children admitted to hospital in Nairobi, Kenya. BMC Microbiol. 2006;6:101. doi: 10.1186/1471-2180-6-101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Ikumapayi UN, Antonio M, Sonne-Hansen J, Biney E, Enwere G, Okoko B, et al. Molecular epidemiology of community-acquired invasive non-typhoidal Salmonella among children aged 2 29 months in rural Gambia and discovery of a new serovar, Salmonella enterica Dingiri. J Med Microbiol. 2007 Nov;56(Pt 11):1479–84. doi: 10.1099/jmm.0.47416-0. [DOI] [PubMed] [Google Scholar]
  • 10.Lepage P, Bogaerts J, Van Goethem C, Ntahorutaba M, Nsengumuremyi F, Hitimana DG, et al. Community-acquired bacteraemia in African children. Lancet. 1987 Jun 27;1(8548):1458–61. doi: 10.1016/s0140-6736(87)92207-0. [DOI] [PubMed] [Google Scholar]
  • 11.O’Dempsey TJ, McArdle TF, Lloyd-Evans N, Baldeh I, Laurence BE, Secka O, et al. Importance of enteric bacteria as a cause of pneumonia, meningitis and septicemia among children in a rural community in The Gambia, West Africa. Pediatr Infect Dis J. 1994 Feb;13(2):122–8. doi: 10.1097/00006454-199402000-00009. [DOI] [PubMed] [Google Scholar]
  • 12.Walsh AL, Phiri AJ, Graham SM, Molyneux EM, Molyneux ME. Bacteremia in febrile Malawian children: clinical and microbiologic features. Pediatr Infect Dis J. 2000 Apr;19(4):312–8. doi: 10.1097/00006454-200004000-00010. [DOI] [PubMed] [Google Scholar]
  • 13.Sigauque B, Roca A, Mandomando I, Morais L, Quinto L, Sacarlal J, et al. Community-acquired bacteremia among children admitted to a rural hospital in Mozambique. Pediatr Infect Dis J. 2009 Feb;28(2):108–13. doi: 10.1097/INF.0b013e318187a87d. [DOI] [PubMed] [Google Scholar]
  • 14.Tennant SM, Diallo S, Levy H, Livio S, Sow SO, Tapia M, et al. Identification by PCR of non-typhoidal Salmonella enterica serovars associated with invasive infections among febrile patients in Mali. PLoS Negl Trop Dis. 2010;4(3):e621. doi: 10.1371/journal.pntd.0000621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Enwere G, Biney E, Cheung YB, Zaman SM, Okoko B, Oluwalana C, et al. Epidemiologic and clinical characteristics of community-acquired invasive bacterial infections in children aged 2–29 months in The Gambia. Pediatr Infect Dis J. 2006 Aug;25(8):700–5. doi: 10.1097/01.inf.0000226839.30925.a5. [DOI] [PubMed] [Google Scholar]
  • 16.Kingsley RA, Msefula CL, Thomson NR, Kariuki S, Holt KE, Gordon MA, et al. Epidemic multiple drug resistant Salmonella Typhimurium causing invasive disease in sub-Saharan Africa have a distinct genotype. Genome Res. 2009 Dec;19(12):2279–87. doi: 10.1101/gr.091017.109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Kariuki S, Revathi G, Gakuya F, Yamo V, Muyodi J, Hart CA. Lack of clonal relationship between non-Typhi Salmonella strain types from humans and those isolated from animals living in close contact. FEMS Immunol Med Microbiol. 2002 Jul 12;33(3):165–71. doi: 10.1111/j.1574-695X.2002.tb00587.x. [DOI] [PubMed] [Google Scholar]
  • 18.Carter PB, Collins FM. The route of enteric infection in normal mice. J Exp Med. 1974 May 1;139(5):1189–203. doi: 10.1084/jem.139.5.1189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Haraga A, Ohlson MB, Miller SI. Salmonellae interplay with host cells. Nat Rev Microbiol. 2008 Jan;6(1):53–66. doi: 10.1038/nrmicro1788. [DOI] [PubMed] [Google Scholar]
  • 20.Collins FM. Recall of immunity in mice vaccinated with Salmonella Enteritidis or Salmonella Typhimurium. J Bacteriol. 1968 Jun;95(6):2014–21. doi: 10.1128/jb.95.6.2014-2021.1968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Jarvelainen HA, Galmiche A, Zychlinsky A. Caspase-1 activation by Salmonella. Trends Cell Biol. 2003 Apr;13(4):204–9. doi: 10.1016/s0962-8924(03)00032-1. [DOI] [PubMed] [Google Scholar]
  • 22.Watson KG, Holden DW. Dynamics of growth and dissemination of Salmonella in vivo. Cell Microbiol. 2010 Oct;12(10):1389–97. doi: 10.1111/j.1462-5822.2010.01511.x. [DOI] [PubMed] [Google Scholar]
  • 23.Valdez Y, Ferreira RB, Finlay BB. Molecular mechanisms of Salmonella virulence and host resistance. Curr Top Microbiol Immunol. 2009;337:93–127. doi: 10.1007/978-3-642-01846-6_4. [DOI] [PubMed] [Google Scholar]
  • 24.Jones BD, Ghori N, Falkow S. Salmonella Typhimurium initiates murine infection by penetrating and destroying the specialized epithelial M cells of the Peyer’s patches. J Exp Med. 1994 Jul 1;180(1):15–23. doi: 10.1084/jem.180.1.15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Vazquez-Torres A, Jones-Carson J, Baumler AJ, Falkow S, Valdivia R, Brown W, et al. Extraintestinal dissemination of Salmonella by CD18-expressing phagocytes. Nature. 1999 Oct 21;401(6755):804–8. doi: 10.1038/44593. [DOI] [PubMed] [Google Scholar]
  • 26.Collins FM, Carter PB. Comparative immunogenicity of heat-killed and living oral Salmonella vaccines. Infect Immun. 1972 Oct;6(4):451–8. doi: 10.1128/iai.6.4.451-458.1972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Collins FM. Effect of specific immune mouse serum on the growth of Salmonella Enteritidis in mice preimmunized with living or ethyl alcohol-killed vaccines. J Bacteriol. 1969 Feb;97(2):676–83. doi: 10.1128/jb.97.2.676-683.1969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Collins FM. Effect of specific immune mouse serum on the growth of Salmonella Enteritidis in nonvaccinated mice challenged by various routes. J Bacteriol. 1969 Feb;97(2):667–75. doi: 10.1128/jb.97.2.667-675.1969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Chatfield SN, Strugnell RA, Dougan G. Live Salmonella as vaccines and carriers of foreign antigenic determinants. Vaccine. 1989 Dec;7(6):495–8. doi: 10.1016/0264-410x(89)90271-5. [DOI] [PubMed] [Google Scholar]
  • 30.Zhang X, Kelly SM, Bollen WS, Curtiss R., 3rd Characterization and immunogenicity of Salmonella Typhimurium SL1344 and UK-1 Δcrp and Δcdt deletion mutants. Infect Immun. 1997 Dec;65(12):5381–7. doi: 10.1128/iai.65.12.5381-5387.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Curtiss R, 3rd, Wanda SY, Gunn BM, Zhang X, Tinge SA, Ananthnarayan V, et al. Salmonella enterica serovar Typhimurium strains with regulated delayed attenuation in vivo. Infect Immun. 2009 Mar;77(3):1071–82. doi: 10.1128/IAI.00693-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Heithoff DM, Sinsheimer RL, Low DA, Mahan MJ. An essential role for DNA adenine methylation in bacterial virulence. Science. 1999 May 7;284(5416):967–70. doi: 10.1126/science.284.5416.967. [DOI] [PubMed] [Google Scholar]
  • 33.Heithoff DM, Enioutina EY, Daynes RA, Sinsheimer RL, Low DA, Mahan MJ. Salmonella DNA adenine methylase mutants confer cross-protective immunity. Infect Immun. 2001 Nov;69(11):6725–30. doi: 10.1128/IAI.69.11.6725-6730.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Pasetti MF, Levine MM, Sztein MB. Animal models paving the way for clinical trials of attenuated Salmonella enterica serovar Typhi live oral vaccines and live vectors. Vaccine. 2003 Jan 17;21(5–6):401–18. doi: 10.1016/s0264-410x(02)00472-3. [DOI] [PubMed] [Google Scholar]
  • 35.Galen JE, Pasetti MF, Tennant S, Ruiz-Olvera P, Sztein MB, Levine MM. Salmonella enterica serovar Typhi live vector vaccines finally come of age. Immunol Cell Biol. 2009 Jul;87(5):400–12. doi: 10.1038/icb.2009.31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Matsui H, Suzuki M, Isshiki Y, Kodama C, Eguchi M, Kikuchi Y, et al. Oral immunization with ATP-dependent protease-deficient mutants protects mice against subsequent oral challenge with virulent Salmonella enterica serovar Typhimurium. Infect Immun. 2003 Jan;71(1):30–9. doi: 10.1128/IAI.71.1.30-39.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Miller SI, Loomis WP, Alpuche-Aranda C, Behlau I, Hohmann E. The PhoP virulence regulon and live oral Salmonella vaccines. Vaccine. 1993;11(2):122–5. doi: 10.1016/0264-410x(93)90006-j. [DOI] [PubMed] [Google Scholar]
  • 38.McFarland WC, Stocker BA. Effect of different purine auxotrophic mutations on mouse-virulence of a Vi-positive strain of Salmonella dublin and of two strains of Salmonella Typhimurium. Microb Pathog. 1987 Aug;3(2):129–41. doi: 10.1016/0882-4010(87)90071-4. [DOI] [PubMed] [Google Scholar]
  • 39.Watson DC, Robbins JB, Szu SC. Protection of mice against Salmonella Typhimurium with an O-specific polysaccharide-protein conjugate vaccine. Infect Immun. 1992 Nov;60(11):4679–86. doi: 10.1128/iai.60.11.4679-4686.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Carter PB, Collins FM. Assessment of typhoid vaccines by using the intraperitoneal route of challenge. Infect Immun. 1977 Sep;17(3):555–60. doi: 10.1128/iai.17.3.555-560.1977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Srinivasan A, McSorley SJ. Activation of Salmonella-specific immune responses in the intestinal mucosa. Arch Immunol Ther Exp (Warsz) 2006 Jan–Feb;54(1):25–31. doi: 10.1007/s00005-006-0003-5. [DOI] [PubMed] [Google Scholar]
  • 42.Moon JJ, McSorley SJ. Tracking the dynamics of Salmonella specific T cell responses. Curr Top Microbiol Immunol. 2009;334:179–98. doi: 10.1007/978-3-540-93864-4_8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Angerman CR, Eisenstein TK. Comparative efficacy and toxicity of a ribosomal vaccine, acetone-killed cells, lipopolysaccharide, and a live cell vaccine prepared from Salmonella Typhimurium. Infect Immun. 1978 Feb;19(2):575–82. doi: 10.1128/iai.19.2.575-582.1978. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Eisenstein TK, Angerman CR. Immunity to experimental Salmonella infection: studies on the protective capacity and immunogenicity of lipopolysaccharide, acetone-killed cells, and ribosome-rich extracts of Salmonella Typhimurium in C3H/HeJ and CD-1 mice. J Immunol. 1978 Sep;121(3):1010–4. [PubMed] [Google Scholar]
  • 45.Eisenstein TK, Deakins LW, Killar L, Saluk PH, Sultzer BM. Dissociation of innate susceptibility to Salmonella infection and endotoxin responsiveness in C3HeB/FeJ mice and other strains in the C3H lineage. Infect Immun. 1982 May;36(2):696–703. doi: 10.1128/iai.36.2.696-703.1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Eisenstein TK, Killar LM, Sultzer BM. Immunity to infection with Salmonella Typhimurium: mouse-strain differences in vaccine- and serum-mediated protection. J Infect Dis. 1984 Sep;150(3):425–35. doi: 10.1093/infdis/150.3.425. [DOI] [PubMed] [Google Scholar]
  • 47.Killar LM, Eisenstein TK. Immunity to Salmonella Typhimurium infection in C3H/HeJ and C3H/HeNCrlBR mice: studies with an aromatic-dependent live S. Typhimurium strain as a vaccine. Infect Immun. 1985 Mar;47(3):605–12. doi: 10.1128/iai.47.3.605-612.1985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Collins FM. Immunity to enteric infection in mice. Infect Immun. 1970 Mar;1(3):243–50. doi: 10.1128/iai.1.3.243-250.1970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Hormaeche CE. Genetics of natural resistance to Salmonellae in mice. Immunology. 1979 Jun;37(2):319–27. [PMC free article] [PubMed] [Google Scholar]
  • 50.Hormaeche CE. Natural resistance to Salmonella Typhimurium in different inbred mouse strains. Immunology. 1979 Jun;37(2):311–8. [PMC free article] [PubMed] [Google Scholar]
  • 51.Hormaeche CE, Maskell DJ, Harrington K, Joysey H, Brock J. Mechanisms of natural resistance to mouse typhoid. Bull Eur Physiopathol Respir. 1983 Mar–Apr;19(2):137–42. [PubMed] [Google Scholar]
  • 52.Collins FM. Salmonellosis in orally infected specific pathogen-free C57B1 mice. Infect Immun. 1972 Feb;5(2):191–8. doi: 10.1128/iai.5.2.191-198.1972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Plant J, Glynn AA. Genetics of resistance to infection with Salmonella Typhimurium in mice. J Infect Dis. 1976 Jan;133(1):72–8. doi: 10.1093/infdis/133.1.72. [DOI] [PubMed] [Google Scholar]
  • 54.Robson HG, Vas SI. Resistance of inbred mice to Salmonella Typhimurium. J Infect Dis. 1972 Oct;126(4):378–86. doi: 10.1093/infdis/126.4.378. [DOI] [PubMed] [Google Scholar]
  • 55.Vidal SM, Malo D, Vogan K, Skamene E, Gros P. Natural resistance to infection with intracellular parasites: isolation of a candidate for Bcg. Cell. 1993 May 7;73(3):469–85. doi: 10.1016/0092-8674(93)90135-d. [DOI] [PubMed] [Google Scholar]
  • 56.Cellier MF, Courville P, Campion C. Nramp1 phagocyte intracellular metal withdrawal defense. Microbes Infect. 2007 Nov–Dec;9(14–15):1662–70. doi: 10.1016/j.micinf.2007.09.006. [DOI] [PubMed] [Google Scholar]
  • 57.Eisenstein TK, Killar LM, Stocker BA, Sultzer BM. Cellular immunity induced by avirulent Salmonella in LPS-defective C3H/HeJ mice. J Immunol. 1984 Aug;133(2):958–61. [PubMed] [Google Scholar]
  • 58.Poltorak A, He X, Smirnova I, Liu MY, Van Huffel C, Du X, et al. Defective LPS signaling in C3H/HeJ and C57BL/10ScCr mice: mutations in Tlr4 gene. Science. 1998 Dec 11;282(5396):2085–8. doi: 10.1126/science.282.5396.2085. [DOI] [PubMed] [Google Scholar]
  • 59.Qureshi ST, Lariviere L, Leveque G, Clermont S, Moore KJ, Gros P, et al. Endotoxin-tolerant mice have mutations in Toll-like receptor 4 (Tlr4) J Exp Med. 1999 Feb 15;189(4):615–25. doi: 10.1084/jem.189.4.615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Talbot S, Totemeyer S, Yamamoto M, Akira S, Hughes K, Gray D, et al. Toll-like receptor 4 signalling through MyD88 is essential to control Salmonella enterica serovar Typhimurium infection, but not for the initiation of bacterial clearance. Immunology. 2009 Dec;128(4):472–83. doi: 10.1111/j.1365-2567.2009.03146.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Wick MJ. Living in the danger zone: innate immunity to Salmonella. Curr Opin Microbiol. 2004 Feb;7(1):51–7. doi: 10.1016/j.mib.2003.12.008. [DOI] [PubMed] [Google Scholar]
  • 62.Fierer J, Swancutt MA, Heumann D, Golenbock D. The role of lipopolysaccharide binding protein in resistance to Salmonella infections in mice. J Immunol. 2002 Jun 15;168(12):6396–403. doi: 10.4049/jimmunol.168.12.6396. [DOI] [PubMed] [Google Scholar]
  • 63.Harrison JA, Villarreal-Ramos B, Mastroeni P, Demarco de Hormaeche R, Hormaeche CE. Correlates of protection induced by live Aro- Salmonella Typhimurium vaccines in the murine typhoid model. Immunology. 1997 Apr;90(4):618–25. doi: 10.1046/j.1365-2567.1997.00158.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.McSorley SJ, Cookson BT, Jenkins MK. Characterization of CD4+ T cell responses during natural infection with Salmonella Typhimurium. J Immunol. 2000 Jan 15;164(2):986–93. doi: 10.4049/jimmunol.164.2.986. [DOI] [PubMed] [Google Scholar]
  • 65.Hormaeche CE, Mastroeni P, Harrison JA, Demarco de Hormaeche R, Svenson S, Stocker BA. Protection against oral challenge three months after i.v. immunization of BALB/c mice with live Aro Salmonella Typhimurium and Salmonella Enteritidis vaccines is serotype (species)-dependent and only partially determined by the main LPS O antigen. Vaccine. 1996 Mar;14(4):251–9. doi: 10.1016/0264-410x(95)00249-z. [DOI] [PubMed] [Google Scholar]
  • 66.Mastroeni P, Villarreal-Ramos B, Hormaeche CE. Adoptive transfer of immunity to oral challenge with virulent Salmonellae in innately susceptible BALB/c mice requires both immune serum and T cells. Infect Immun. 1993 Sep;61(9):3981–4. doi: 10.1128/iai.61.9.3981-3984.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Mastroeni P, Simmons C, Fowler R, Hormaeche CE, Dougan G. 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. 2000 Jan;68(1):46–53. doi: 10.1128/iai.68.1.46-53.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.McSorley SJ, Jenkins MK. Antibody is required for protection against virulent but not attenuated Salmonella enterica serovar Typhimurium. Infect Immun. 2000 Jun;68(6):3344–8. doi: 10.1128/iai.68.6.3344-3348.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Collins FM. Immunogenicity of living and heat-killed Salmonella pullorum vaccines. Infect Immun. 1973 May;7(5):735–42. doi: 10.1128/iai.7.5.735-742.1973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Collins FM. Effect of adjuvant on immunogenicity of a heat-killed Salmonella vaccine. J Infect Dis. 1972 Jul;126(1):69–76. doi: 10.1093/infdis/126.1.69. [DOI] [PubMed] [Google Scholar]
  • 71.Matsui K, Arai T. Protective immunity induced by porin in experimental mouse salmonellosis. Microbiol Immunol. 1989;33(9):699–708. doi: 10.1111/j.1348-0421.1989.tb00957.x. [DOI] [PubMed] [Google Scholar]
  • 72.Matsui K, Arai T. Protective immunities induced by porins from mutant strains of Salmonella Typhimurium. Microbiol Immunol. 1990;34(11):917–27. doi: 10.1111/j.1348-0421.1990.tb01070.x. [DOI] [PubMed] [Google Scholar]
  • 73.Jazani NH, Worobec E, Shahabi S, Nejad GB. Conjugation of tetanus toxoid with Salmonella Typhimurium PTCC 1735 O-specific polysaccharide and its effects on production of opsonizing antibodies in a mouse model. Can J Microbiol. 2005 Apr;51(4):319–24. doi: 10.1139/w05-008. [DOI] [PubMed] [Google Scholar]
  • 74.Gil-Cruz C, Bobat S, Marshall JL, Kingsley RA, Ross EA, Henderson IR, et al. The porin OmpD from nontyphoidal Salmonella is a key target for a protective B1b cell antibody response. Proc Natl Acad Sci U S A. 2009 Jun 16;106(24):9803–8. doi: 10.1073/pnas.0812431106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Udhayakumar V, Muthukkaruppan VR. Protective immunity induced by outer membrane proteins of Salmonella Typhimurium in mice. Infect Immun. 1987 Mar;55(3):816–21. doi: 10.1128/iai.55.3.816-821.1987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Muthukkumar S, Muthukkaruppan VR. Mechanism of protective immunity induced by porin-lipopolysaccharide against murine salmonellosis. Infect Immun. 1993 Jul;61(7):3017–25. doi: 10.1128/iai.61.7.3017-3025.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Kita E, Nishi K, Emoto M, Katsui N, Yasui K, Kashiba S. Analysis of immunity to infection with Salmonella Typhimurium in outbred mice. I. Requirement of the antibody to non-O antigen for protection in mice that are not protected by the RNA-rich vaccine. Immunology. 1987 Aug;61(4):535–41. [PMC free article] [PubMed] [Google Scholar]
  • 78.Kuusi N, Nurminen M, Saxen H, Valtonen M, Makela PH. Immunization with major outer membrane proteins in experimental salmonellosis of mice. Infect Immun. 1979 Sep;25(3):857–62. doi: 10.1128/iai.25.3.857-862.1979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Ornellas EP, Roantree RJ, Steward JP. The specificity and importance of humoral antibody in the protection of mice against intraperitoneal challenge with complement-sensitive and complement-resistant Salmonella. J Infect Dis. 1970 Feb;121(2):113–23. doi: 10.1093/infdis/121.2.113. [DOI] [PubMed] [Google Scholar]
  • 80.Svenson SB, Nurminen M, Lindberg AA. Artificial Salmonella vaccines: O-antigenic oligosaccharide-protein conjugates induce protection against infection with Salmonella Typhimurium. Infect Immun. 1979 Sep;25(3):863–72. doi: 10.1128/iai.25.3.863-872.1979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Kenny K, Herzberg M. Early antibody response in mice to either infection or immunization with Salmonella Typhimurium. J Bacteriol. 1967 Mar;93(3):773–8. doi: 10.1128/jb.93.3.773-778.1967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Herzberg M, Nash P, Hino S. Degree of immunity induced by killed vaccines to experimental salmonellosis in mice. Infect Immun. 1972 Jan;5(1):83–90. doi: 10.1128/iai.5.1.83-90.1972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Tabaraie B, Sharma BK, Sharma PR, Sehgal R, Ganguly NK. Evaluation of Salmonella porins as a broad spectrum vaccine candidate. Microbiol Immunol. 1994;38(7):553–9. doi: 10.1111/j.1348-0421.1994.tb01822.x. [DOI] [PubMed] [Google Scholar]
  • 84.Mastroeni P, Menager N. Development of acquired immunity to Salmonella. J Med Microbiol. 2003 Jun;52(Pt 6):453–9. doi: 10.1099/jmm.0.05173-0. [DOI] [PubMed] [Google Scholar]
  • 85.Michetti P, Mahan MJ, Slauch JM, Mekalanos JJ, Neutra MR. Monoclonal secretory immunoglobulin A protects mice against oral challenge with the invasive pathogen Salmonella Typhimurium. Infect Immun. 1992 May;60(5):1786–92. doi: 10.1128/iai.60.5.1786-1792.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Michetti P, Porta N, Mahan MJ, Slauch JM, Mekalanos JJ, Blum AL, et al. Monoclonal immunoglobulin A prevents adherence and invasion of polarized epithelial cell monolayers by Salmonella Typhimurium. Gastroenterology. 1994 Oct;107(4):915–23. doi: 10.1016/0016-5085(94)90214-3. [DOI] [PubMed] [Google Scholar]
  • 87.Eisenstein TK. Intracellular pathogens: the role of antibody-mediated protection in Salmonella infection. Trends Microbiol. 1998 Apr;6(4):135–6. doi: 10.1016/s0966-842x(98)01242-6. [DOI] [PubMed] [Google Scholar]
  • 88.Casadevall A, Pirofski LA. A reappraisal of humoral immunity based on mechanisms of antibody-mediated protection against intracellular pathogens. Adv Immunol. 2006;91:1–44. doi: 10.1016/S0065-2776(06)91001-3. [DOI] [PubMed] [Google Scholar]
  • 89.Cunningham AF, Gaspal F, Serre K, Mohr E, Henderson IR, Scott-Tucker A, et al. Salmonella induces a switched antibody response without germinal centers that impedes the extracellular spread of infection. J Immunol. 2007 May 15;178(10):6200–7. doi: 10.4049/jimmunol.178.10.6200. [DOI] [PubMed] [Google Scholar]
  • 90.Roantree RJ, Rantz LA. A Study of the Relationship of the Normal Bactericidal Activity of Human Serum to Bacterial Infection. J Clin Invest. 1960 Jan;39(1):72–81. doi: 10.1172/JCI104029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Souwer Y, Griekspoor A, Jorritsma T, de Wit J, Janssen H, Neefjes J, et al. B cell receptor-mediated internalization of Salmonella: a novel pathway for autonomous B cell activation and antibody production. J Immunol. 2009 Jun 15;182(12):7473–81. doi: 10.4049/jimmunol.0802831. [DOI] [PubMed] [Google Scholar]
  • 92.Liang-Takasaki CJ, Saxen H, Makela PH, Leive L. Complement activation by polysaccharide of lipopolysaccharide: an important virulence determinant of Salmonellae. Infect Immun. 1983 Aug;41(2):563–9. doi: 10.1128/iai.41.2.563-569.1983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Jimenez-Lucho VE, Joiner KA, Foulds J, Frank MM, Leive L. C3b generation is affected by the structure of the O-antigen polysaccharide in lipopolysaccharide from Salmonellae. J Immunol. 1987 Aug 15;139(4):1253–9. [PubMed] [Google Scholar]
  • 94.Mittrucker HW, Raupach B, Kohler A, Kaufmann SH. Cutting edge: role of B lymphocytes in protective immunity against Salmonella Typhimurium infection. J Immunol. 2000 Feb 15;164(4):1648–52. doi: 10.4049/jimmunol.164.4.1648. [DOI] [PubMed] [Google Scholar]
  • 95.Mastroeni P, Villarreal-Ramos B, Hormaeche CE. Role of T cells, TNF alpha and IFN gamma in recall of immunity to oral challenge with virulent Salmonellae in mice vaccinated with live attenuated aro- Salmonella vaccines. Microb Pathog. 1992 Dec;13(6):477–91. doi: 10.1016/0882-4010(92)90014-f. [DOI] [PubMed] [Google Scholar]
  • 96.Lo WF, Ong H, Metcalf ES, Soloski MJ. T cell responses to Gram-negative intracellular bacterial pathogens: a role for CD8+ T cells in immunity to Salmonella infection and the involvement of MHC class Ib molecules. J Immunol. 1999 May 1;162(9):5398–406. [PubMed] [Google Scholar]
  • 97.Collins FM. Vaccines and cell-mediated immunity. Bacteriol Rev. 1974 Dec;38(4):371–402. doi: 10.1128/br.38.4.371-402.1974. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Bergman MA, Cummings LA, Alaniz RC, Mayeda L, Fellnerova I, Cookson BT. CD4+-T-cell responses generated during murine Salmonella enterica serovar Typhimurium infection are directed towards multiple epitopes within the natural antigen FliC. Infect Immun. 2005 Nov;73(11):7226–35. doi: 10.1128/IAI.73.11.7226-7235.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Bergman MA, Cummings LA, Barrett SL, Smith KD, Lara JC, Aderem A, et al. CD4+ T cells and toll-like receptors recognize Salmonella antigens expressed in bacterial surface organelles. Infect Immun. 2005 Mar;73(3):1350–6. doi: 10.1128/IAI.73.3.1350-1356.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Mastroeni P, Arena A, Costa GB, Liberto MC, Bonina L, Hormaeche CE. Serum TNF alpha in mouse typhoid and enhancement of a Salmonella infection by anti-TNF alpha antibodies. Microb Pathog. 1991 Jul;11(1):33–8. doi: 10.1016/0882-4010(91)90091-n. [DOI] [PubMed] [Google Scholar]
  • 101.Mittrucker HW, Kohler A, Mak TW, Kaufmann SH. Critical role of CD28 in protective immunity against Salmonella Typhimurium. J Immunol. 1999 Dec 15;163(12):6769–76. [PubMed] [Google Scholar]
  • 102.Ravindran R, Foley J, Stoklasek T, Glimcher LH, McSorley SJ. Expression of T-bet by CD4 T cells is essential for resistance to Salmonella infection. J Immunol. 2005 Oct 1;175(7):4603–10. doi: 10.4049/jimmunol.175.7.4603. [DOI] [PubMed] [Google Scholar]
  • 103.Monack DM, Bouley DM, Falkow S. Salmonella Typhimurium persists within macrophages in the mesenteric lymph nodes of chronically infected Nramp1+/+ mice and can be reactivated by IFNgamma neutralization. J Exp Med. 2004 Jan 19;199(2):231–41. doi: 10.1084/jem.20031319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Alaniz RC, Cummings LA, Bergman MA, Rassoulian-Barrett SL, Cookson BT. Salmonella Typhimurium coordinately regulates FliC location and reduces dendritic cell activation and antigen presentation to CD4+ T cells. J Immunol. 2006 Sep 15;177(6):3983–93. doi: 10.4049/jimmunol.177.6.3983. [DOI] [PubMed] [Google Scholar]
  • 105.Lalmanach AC, Lantier F. Host cytokine response and resistance to Salmonella infection. Microbes Infect. 1999 Jul;1(9):719–26. doi: 10.1016/s1286-4579(99)80073-2. [DOI] [PubMed] [Google Scholar]
  • 106.Eckmann L, Kagnoff MF. Cytokines in host defense against Salmonella. Microbes Infect. 2001 Nov–Dec;3(14–15):1191–200. doi: 10.1016/s1286-4579(01)01479-4. [DOI] [PubMed] [Google Scholar]
  • 107.van de Vosse E, Hoeve MA, Ottenhoff TH. Human genetics of intracellular infectious diseases: molecular and cellular immunity against mycobacteria and Salmonellae. Lancet Infect Dis. 2004 Dec;4(12):739–49. doi: 10.1016/S1473-3099(04)01203-4. [DOI] [PubMed] [Google Scholar]
  • 108.van de Vosse E, van Dissel JT, Ottenhoff TH. Genetic deficiencies of innate immune signalling in human infectious disease. Lancet Infect Dis. 2009 Nov;9(11):688–98. doi: 10.1016/S1473-3099(09)70255-5. [DOI] [PubMed] [Google Scholar]
  • 109.Dunstan SJ, Stephens HA, Blackwell JM, Duc CM, Lanh MN, Dudbridge F, et al. Genes of the class II and class III major histocompatibility complex are associated with typhoid fever in Vietnam. J Infect Dis. 2001 Jan 15;183(2):261–8. doi: 10.1086/317940. [DOI] [PubMed] [Google Scholar]
  • 110.Hormaeche CE, Harrington KA, Joysey HS. Natural resistance to Salmonellae in mice: control by genes within the major histocompatibility complex. J Infect Dis. 1985 Nov;152(5):1050–6. doi: 10.1093/infdis/152.5.1050. [DOI] [PubMed] [Google Scholar]
  • 111.Svenson SB, Lindberg AA. Protection against mouse typhoid by artificial Salmonella vaccines. Scand J Infect Dis Suppl. 1980;(Suppl 24):210–5. [PubMed] [Google Scholar]
  • 112.Jorbeck HJ, Svenson SB, Lindberg AA. Artificial Salmonella vaccines: Salmonella Typhimurium O-antigen-specific oligosaccharide-protein conjugates elicit opsonizing antibodies that enhance phagocytosis. Infect Immun. 1981 May;32(2):497–502. doi: 10.1128/iai.32.2.497-502.1981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Svenson SB, Lindberg AA. Artificial Salmonella vaccines: Salmonella Typhimurium O-antigen-specific oligosaccharide-protein conjugates elicit protective antibodies in rabbits and mice. Infect Immun. 1981 May;32(2):490–6. doi: 10.1128/iai.32.2.490-496.1981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Colwell DE, Michalek SM, Briles DE, Jirillo E, McGhee JR. Monoclonal antibodies to Salmonella lipopolysaccharide: anti-O-polysaccharide antibodies protect C3H mice against challenge with virulent Salmonella Typhimurium. J Immunol. 1984 Aug;133(2):950–7. [PubMed] [Google Scholar]
  • 115.Levine MM, Taylor DN, Ferreccio C. Typhoid vaccines come of age. Pediatr Infect Dis J. 1989 Jun;8(6):374–81. doi: 10.1097/00006454-198906000-00010. [DOI] [PubMed] [Google Scholar]
  • 116.Lin FY, Ho VA, Khiem HB, Trach DD, Bay PV, Thanh TC, et al. The efficacy of a Salmonella Typhi Vi conjugate vaccine in two-to-five-year-old children. N Engl J Med. 2001 Apr 26;344(17):1263–9. doi: 10.1056/NEJM200104263441701. [DOI] [PubMed] [Google Scholar]
  • 117.Mai NL, Phan VB, Vo AH, Tran CT, Lin FY, Bryla DA, et al. Persistent efficacy of Vi conjugate vaccine against typhoid fever in young children. N Engl J Med. 2003 Oct 2;349(14):1390–1. doi: 10.1056/NEJM200310023491423. [DOI] [PubMed] [Google Scholar]
  • 118.Szu SC, Taylor DN, Trofa AC, Clements JD, Shiloach J, Sadoff JC, et al. Laboratory and preliminary clinical characterization of Vi capsular polysaccharide-protein conjugate vaccines. Infect Immun. 1994 Oct;62(10):4440–4. doi: 10.1128/iai.62.10.4440-4444.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES