Next Article in Journal
Considering Genomic and Immunological Correlates of Protection for a Dengue Intervention
Previous Article in Journal
Assessing Vaccine Hesitancy among Healthcare Workers: A Cross-Sectional Study at an Italian Paediatric Hospital and the Development of a Healthcare Worker’s Vaccination Compliance Index
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Evaluation of the Safety and Protection Efficacy of spiC and nmpC or rfaL Deletion Mutants of Salmonella Enteritidis as Live Vaccine Candidates for Poultry Non-Typhoidal Salmonellosis

1
Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agri-food Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou 225009, China
2
Jiangsu Key Lab of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
3
Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou 225009, China
*
Author to whom correspondence should be addressed.
These authors contributed equally.
Vaccines 2019, 7(4), 202; https://doi.org/10.3390/vaccines7040202
Submission received: 31 October 2019 / Revised: 25 November 2019 / Accepted: 28 November 2019 / Published: 30 November 2019
(This article belongs to the Section Veterinary Vaccines)

Abstract

:
Salmonella enterica serovar Enteritidis (S. Enteritidis) is a host-ranged pathogen that can infect both animals and humans. Poultry and poultry products are the main carriers of S. Enteritidis, which can be transmitted to humans through the food chain. To eradicate the prevalence of S. Enteritidis in poultry farms, it is necessary to develop novel vaccines against the pathogen. In this study, we constructed two vaccine candidates, CZ14-1∆spiC∆nmpC and CZ14-1∆spiC∆rfaL, and evaluated their protective efficacy. Both mutant strains were much less virulent than the parental strain, as determined by the 50% lethal dose (LD50) for three-day-old specific-pathogen free (SPF) White Leghorns and Hyline White chickens. Immunization with the mutant candidates induced highly specific humoral immune responses and expression of cytokines IFN-γ, IL-1β, and IL-6. In addition, the mutant strains were found to be persistent for almost three weeks post-infection. The survival percentages of chickens immunized with CZ14-1∆spiC∆nmpC and CZ14-1∆spiC∆rfaL reached 80% and 75%, respectively, after challenge with the parental strain. Overall, these results demonstrate that the two mutant strains can be developed as live attenuated vaccines.

1. Introduction

The Salmonella enterica serovar Enteritidis (S. Enteritidis) is one of the most common Salmonella serotypes causing human infections worldwide [1,2]. Human salmonellosis is frequently caused by consumption of Salmonella-contaminated foods. From 2015 to 2017, the proportion of human salmonellosis caused by S. Enteritidis increased from 54.7% to 61.2% in the EU (EFSA, 2018). In the USA, S. Enteritidis was the predominant serotype in reported human Salmonella infections from 2011 to 2016, and it was also the most common serotype in clinical chicken isolates [1]. In China, our surveillance results over the last 10 years showed that about 30% of Salmonella isolates from poultry meat were S. Enteritidis. In addition, epidemiological reports implicated that human S. Enteritidis cases are frequently caused by the consumption of contaminated chicken products [3].
Since poultry and poultry products, including eggs, have been identified as the major source of S. Enteritidis in human foods worldwide, there is an urgent need for efficient programs to control Salmonella infections in poultry. Strict biosecurity measures, including tight control of the housing environment, feed quality, water supply, management pattern, and complete exclusion of sources of infection (rodents, insects, wild birds, etc.), should be applied to prevent the introduction of the pathogen into poultry farms [4,5]. However, the high costs of these measures restrict their application in many developing countries. Antimicrobial therapy is another strategy that can be used to control Salmonella infection, but the excessive use of antibiotics has led to the emergence of multidrug-resistant strains [6,7]. Bacteriophage therapy is another method developed to control Salmonella infection in poultry by reducing the bacterial load in chickens, but treatment of bacteriophage can cause phage-resistant S. Enteritidis [8,9]. Therefore, vaccination is an efficient strategy for the control of Salmonella infections in poultry. To date, some S. Enteritidis vaccines have been developed and used in the poultry industry. Application of these vaccines decreased the incidence of S. Enteritidis in poultry flocks [10]. However, administration of live attenuated vaccines is not yet able to prevent the spread of S. Enteritidis in poultry flocks in China [11]. New vaccines are needed to prevent the infection and the spread of S. Enteritidis in the poultry industry.
SpiC is an SPI-2-encoded effector protein that is secreted by SPI-2 type III secretion system (T3SS) of Salmonella into macrophages [12]. Deletion of spiC has been confirmed to significantly reduce Salmonella virulence in mice and chickens [12,13,14]. Furthermore, the spiC-deleted S. Pullorum strain has been evaluated as a potential vaccine candidate in chickens [13,15]. To distinguish vaccinated animals from naturally infected ones, the so-called DIVA (differentiating infected from vaccinated animals) strategy has been successfully applied in vaccine development [16]. Vaccine candidates with the genes encoding the outer segment of bacterial lipopolysaccharide (LPS) and outer membrane proteins deleted can be developed as DIVA vaccines, which can be differentiated from natural infection by using available diagnostic serological tests [17,18]. The rfaL gene encodes a membrane enzyme involved in the synthesis of LPS, and deletion of the gene changes smooth LPS to rough LPS, which cannot react with the antibodies against the O antigen [15,19]. In addition, the rfaL-deleted mutant of S. Typhimurium was also attenuated in mice [20]. OmpD is the most abundant porin in the outer membrane of Salmonella involved in inducing the B1b cell antibody response in mice [21,22]. Deletion of the ompD gene (also named nmpC) slightly reduced the virulence of S. Typhimurium in mice. Therefore, both rfaL and nmpC can be chosen as target genes for construction of DIVA vaccine candidates based on deletion of spiC in S. Enteritidis.
In the present study, we constructed two S. Enteritidis vaccine candidates and evaluated their protective efficacy in chickens. The mutants CZ14-1∆spiC∆nmpC and CZ14-1∆spiC∆rfaL were generated by λ-Red-mediated recombination [23] and homologous recombination using suicide vector pGMB151 [24]. The feasibility of these two mutants as live attenuated vaccines was subsequently evaluated by analyzing virulence, immune response, protective efficacy, etc.

2. Materials and Methods

2.1. Bacterial Strains and Development of S. Enteritidis Mutant Strains

S. Enteritidis CZ14-1 is a clinical strain that was obtained from chicken eggs from a poultry farm in the Jiangsu Province of China in 2016 [11]. It is a wild type virulent strain with resistance to Nalidixic acid. Deletion of spiC in CZ14-1 was performed using the suicide vector pGMB151 based on homologous recombination, as previously described [24]. Either rfaL or nmpC was subsequently deleted using the one-step activation method of the λ-Red mutation system [22]. The open reading frame (ORF) of the targeted genes were completely deleted, and this was confirmed by PCR analysis and sequencing. All of the strains were cultured in Luria-Bertani (LB) agar medium, LB broth, and Brilliant green agar (Becton Dickinson) at 37 °C.

2.2. Chickens

The specific-pathogen free (SPF) White Leghorns and Hyline White chickens were purchased from Jinan Sipaffrey Poultry Co., Ltd. (Jinan, Shangdong, China). The 3-day old chickens were confirmed as free from Salmonella infection by both bacteriological examination and serum detection. Each group of chickens was housed in separate rearing isolators and supplied with commercial feed and drinking water. The food and the water for chickens were tested to be Salmonella negative. All of the animal experiments and management procedures were undertaken with the permission of the Animal Welfare and Ethics Committees of Yangzhou University (Approval No. SYXK(Su)2016-0020) and complied with the guidelines of the institutional administrative committee and the ethics committee for laboratory animals.

2.3. Virulence Assessment

To evaluate the virulence of the two mutants (CZ14-1∆spiC∆nmpC and CZ14-1∆spiC∆rfaL) in chickens, the 50% lethal doses (LD50) were determined using 50 chickens for each strain randomly assigned into five groups. Each group was inoculated intramuscularly with 10-fold dilutions of the mutants from 1 × 105 to 1 × 109 CFU. Another group of 50 was inoculated intramuscularly with the parental strain CZ14-1 from 1 × 104 to 1 × 108 CFU. In addition, ten chickens were inoculated with 200 µL of phosphate-buffered saline (PBS) via the same route. Birds that died were recorded daily for three weeks post-infection, and the LD50 was calculated using the Reed–Muench method [25].

2.4. Biochemical Test and Growth Ability In Vitro

To investigate the effect of deleted genes on the biological properties of S. Enteritidis mutants, a biochemical test was performed using an API20E plate (BioMérieux, France), and the results compared to those for the parental strain CZ14-1. In addition, the growth ability was determined by measuring the optical density (OD600) of each strain cultured in 15 mL of LB broth at 37 °C with shaking at 180 rpm. The OD600 was monitored every hour for 18 h as previously described [26].

2.5. Bacterial Vaccination and Changes in Body Weight in Chickens

A total of 60 7-day old Hyline White chickens were randomly divided into three groups (n = 20 for each group): CZ14-1∆spiC∆nmpC-vaccinated group, CZ14-1∆spiC∆rfaL-vaccinated group, and non-vaccinated control group. The vaccinated groups were immunized intramuscularly with 1 × 105 CFU of one of the mutant strains in 100 µL PBS, while the control group was only inoculated with PBS. At 14 days old, the chickens were again immunized via the same route and the same dose of bacteria. The body weights of these chickens were then measured at 7, 14, and 21 days after the second vaccination.

2.6. Persistence and Clearance of Bacteria in Chicken Tissues and Organs

A total of 90 3-day old Hyline White chickens were randomly assigned into three groups (n = 30 for each group). Each group was immunized intramuscularly with 1 × 105 CFU of CZ14-1, CZ14-1∆spiC∆nmpC, or CZ14-1∆spiC∆rfaL. Liver, spleen, ileum, and cecum samples from five chickens from each group were aseptically collected at 1, 3, 7, 14, and 21 days post immunization. The samples were weighed and suspended in 1 mL of PBS for homogenization. The homogenates were diluted serially and subsequently inoculated on Brilliant blue agar plates for bacterial recovery at 37 °C. After overnight cultivation, the bacterial number was counted and reported as log10 CFU/g.

2.7. Immune Response

The humoral immune response was evaluated through measurement of IgG titers specifically against S. Enteritidis by indirect enzyme-linked immunosorbent assay (ELISA) using the heat-killed CZ14-1 strain as the coating antigen. Serum samples were collected at 7, 14, and 21 days post immunization and then serially diluted to be considered as the primary antibody. Horseradish peroxidase (HRP)-conjugated goat anti-chicken IgG (1:10,000 dilution, Sigma-Aldrich) was used as the secondary antibody. The HRP activity was determined using 3,3′,5,5′-tetramethylbenzidine (TMB, Sigma-Aldrich), and the OD450 value was determined with an ELISA reader (BioTek, USA).
To evaluate the T-cell immune response and inflammatory response induced by the vaccine strains, the expression of cytokines IFN-γ, IL-1β, IL-6, and CXCLi1 was measured by qRT-PCR analysis with TaqmanTM probes as previously described [27]. Total RNA was extracted from spleen samples collected at 7, 14, and 21 days post-vaccination using an RNA plus Mini kit (Qiagen, Germany) according to the manufacturer’s instructions. The RNA was reverse transcribed into cDNA and subjected to qRT-PCR analysis, which was performed in triplicate and repeated three times.

2.8. Immune Protection Assessment

To evaluate the protective efficacy of the two vaccine candidates, 60 7-day old Hyline White chickens were randomly divided into three groups of 20 chickens (groups A, B, and C). Groups A and B were immunized intramuscularly with 1 × 105 CFU of the mutants CZ14-1∆spiC∆nmpC and CZ14-1∆spiC∆rfaL, respectively. Group C received 100 µL of PBS as the negative control. One week post-infection, groups A and B were immunized again with the same mutants using the same route and dose. After one week, all of the groups were challenged with 2 × 109 CFU of CZ14-1. The number of surviving chickens was recorded, and clinical symptoms were observed daily for three weeks. In addition, to examine the persistence and the clearance of the wild-type strain in the vaccinated chickens, 90 7-day old chickens were divided into three groups of 30 chickens and subjected to the same immunization and challenge process as described above. After challenging, five chickens from each group were killed at 3, 7, 14, 21, 28, and 35 days post-challenge, liver, spleen, ileum, and cecum samples were collected, and the bacterial number was counted on Brilliant green agar plates, as mentioned above.

2.9. Statistical Analysis

All data are described as mean ± standard error of the mean (SEM) unless otherwise specified and were analyzed using GraphPad Prism version 7.0. A p value of less than 0.05 was considered statistically significant.

3. Results

3.1. Virulence of the Two Vaccine Candidates

The virulence of the parental and the two mutant strains was evaluated in three-day-old SPF White Leghorns and Hyline White chickens. As shown in Table 1, the LD50 of the CZ14-1∆spiC∆nmpC (1.58 × 107 CFU) and CZ14-1∆spiC∆rfaL (7.76 × 106 CFU) strains were 710-fold and 346-fold higher than that of parental strain CZ14-1 (2.24 × 104 CFU) in White Leghorn chickens; in Hyline White chickens, the LD50 of the CZ14-1∆spiC∆nmpC (2.57 × 107 CFU) and CZ14-1∆spiC∆rfaL (4.07 × 106 CFU) strains were 2030-fold and 407-fold higher than that of the CZ14-1 strain (1.26×104 CFU) Table 2. The results indicate that both mutant strains were attenuated and that CZ14-1∆spiC∆nmpC was highly attenuated compared to CZ14-1∆spiC∆rfaL. In addition, the Hyline White chickens were more sensitive to S. Enteritidis than the SPF chickens.

3.2. Biochemical Property and Growth Ability of Mutant Strains

The biochemical test performed using the API20E plate reflected that the mutated genes had no significant effect on bacterial metabolism (unpublished). Mutation of rfaL in CZ14-1∆spiC∆rfaL caused a deficiency in S. Enteritidis in utilizing mannitol. Moreover, the bacterial growth data showed that mutation of rfaL decreased growth under laboratory conditions. However, the growth curve of CZ14-1∆spiC∆nmpC was similar to that of the parental strain, which showed that deletion of nmpC did not perturb the bacterial growth in vitro Figure 1a.

3.3. Changes in Body Weight Following Immunization

The mean body weights of the chickens in the two vaccinated groups and the PBS group at 7, 14, and 21 days post-vaccination are shown in Figure 1b. Chickens in both vaccinated groups showed increased body weight compared to that of the PBS group. Furthermore, the bodyweight of chickens in the CZ14-1∆spiC∆rfaL group was significantly higher than that of the PBS group. No significant difference was detected between the CZ14-1∆spiC∆nmpC-vaccinated group and the PBS group at 7 and 21 days post-vaccination.

3.4. Bacterial Colonization and Persistence in Chicken Tissues and Organs

All of the tissues and organs collected from chickens inoculated with PBS were negative for bacterial recovery. However, although the virulence of the two mutant strains was significantly decreased compared to the parental strain, the colonization ability of both mutant strains in all four tissue and organ types tested was significantly stronger than that of the wild-type strain at 3 days post-immunization (Figure 2). Bacterial recovery was not detected in tissue and organ types tested from the wild-type-infected group at 1 and 3 days post-immunization. While ≥4/5 (four out of five chickens) of CZ14-1∆spiC∆nmpC and all five of the CZ14-1∆spiC∆rfaL-immunized chickens were found to be positive for Salmonella in the liver at 1 and 3 days post-immunization (Figure 2a), the CZ14-1∆spiC∆rfaL exhibited faster invasion into the spleen than the parental strain and CZ14-1∆spiC∆nmpC (Figure 2b). At seven and 14 days post-immunization, the bacterial counts in the spleen, the liver, and the ileum were similar among the three groups (Figure 2a–c), but there was a significant difference between cecum from the wild-type-infected group and that from chickens vaccinated with CZ14-1∆spiC∆rfaL (Figure 2d). At 21 days post-immunization, bacteria were almost eliminated in the liver and the ileum in all three groups. Only one chicken vaccinated with CZ14-1∆spiC∆nmpC was found to be Salmonella-positive in the spleen (Figure 2c). However, in the cecum, a low level of bacterial recovery could still be detected in all three groups (Figure 2d).

3.5. Immune Response

To evaluate the humoral immune response induced by the two vaccine candidates, serum IgG levels were determined using an indirect ELISA assay. In both of the vaccinated groups, the serum IgG against S. Enteritidis was detected at 7 days post immunization and increased dramatically at 14 and 21 days post immunization then remained stable from 21 to 35 days post immunization (Figure 3a, 3b). At 28 days post immunization, the serum IgG level was significantly higher in the CZ14-1∆spiC∆rfaL-immunized group than that in the CZ14-1∆spiC∆nmpC-immunized chickens (Figure 3b).
qRT-PCR analysis of cytokine expression levels in the spleen showed that both vaccinated groups displayed higher IFN-γ levels than the control group at 7, 14, and 21 days post-immunization, and no difference was detected between the two vaccinated groups (Figure 3c). The expression of IL-1β was higher in the two vaccinated groups than the PBS group at 7 and 14 days post immunization (Figure 3e), while IL-6 showed higher expression at 14 and 21 days post-immunization in the vaccinated groups than that in the PBS group (Figure 3d). Immunization with the two mutant strains had no effect on the expression of CXCLi1, which was similar in the vaccinated groups and the PBS group (Figure 3f).

3.6. Immune Protection

The survival percentages for the chickens vaccinated intramuscularly with CZ14-1∆spiC∆rfaL and CZ14-1∆spiC∆nmpC followed by challenge with the wild-type strain CZ14-1 are shown in Table 3. Four and five chickens died in groups A and B, respectively, following the challenge with the parental strain. However, 19 out of 20 non-vaccinated chickens died in group C. The clinical symptoms in the vaccinated group were slight and temporary after the challenge compared with the severe diarrhea seen in the non-vaccinated chickens.
To detect the bacteria eradication ability of the vaccinated groups, liver, spleen, ileum, and cecum were collected for bacterial recovery and counting. As shown in Figure 4, the bacterial number was high at 3 and 7 days post-challenge but decreased dramatically at 14 days post-challenge. The bacteria were eradicated at 21, 28, and 35 days post-challenge in the ileum, the liver, and the spleen, respectively (Figure 4b–d). In the cecum, only one chicken in the CZ14-1∆spiC∆nmpC-vaccinated group had almost 10 CFU/g of Salmonella at 35 days post-challenge (Figure 4e). Since most of the chickens in the non-vaccinated group were dead at 7 days post-challenge, no data were collected for the following days in this group.

4. Discussion

Until now, three major types of vaccines, namely, killed bacteria, subunit vaccines, and live attenuated vaccines, have been developed to control S. Enteritidis in the poultry industry [28,29,30,31,32]. The killed bacteria and subunit vaccines can induce strong antibodies to eradicate extracellular bacteria, but it is difficult for these antibodies to kill the intracellular S. Enteritidis, which could be achieved with the live attenuated strains of Salmonella [28,32,33]. A live attenuated Salmonella vaccine should be attenuated, immunogenic, protective, and allow differentiation of vaccinated chickens from naturally infected flocks [34,35]. In this study, we constructed two mutants with virulence factor (SPI-2 effector) spiC mutated combined with a gene involved in the synthesis of LPS (rfaL) or out-membrane protein (nmpC). Both mutants are highly attenuated, strongly immunogenic, and provide excellent protective efficacy against parental strain attack. In addition, ELISA or plate agglutination test (PAT) can be used to differentiate vaccinated from naturally infected chickens.
A live attenuated vaccine should be avirulent. SpiC is a SPI2-encoded effector involved in the survival of Salmonella in macrophages [36,37]. Deletion of spiC in S. Typhimurium and S. Pullorum caused significantly decreased virulence in mice and chickens, respectively [13,37]. Although deletion of spiC has been achieved in S. Enteritidis C50041, which was confirmed to be less virulent than the parental strain, a mouse-derived isolate [38], in this study, we constructed an spiC-deleted strain combined with rfaL or nmpC gene using chicken-borne isolate CZ14-1. Both double-gene mutants showed greatly decreased virulence compared to that of the parental strain. Furthermore, immunization with the mutants did not affect the chickens’ body weights, leading to better growth than in the non-vaccinated group. No indications of negative effects on the health of the immunized birds were observed.
To eradicate intracellular Salmonella, it is critical to stimulate cellular immune responses in the immunized animals [39,40]. At the acute infection stage, the Salmonella-infected host can produce both Th1 and Th2 immune responses to diminish the growth of the invading bacteria, but in the second stage of persistent infection, the reduced Th1 response disrupts the balance between Th1 and Th2, and a new equilibrium between the host and persistent Salmonella infection is developed and continues [41]. Both mutants in this study can induce increased IFN-γ and IL-6 secretion from 7 to 21 days post-immunization, indicating that a prolonged Th1 immune response can be induced by these mutants. Furthermore, the antibody against S. Enteritidis was produced at 7 days post-immunization, after which production increased and then remained at a stable concentration to kill the extracellular bacteria. However, bacterial shedding is another key point for live attenuated vaccines [42]. In this study, both mutants were almost eliminated in the liver, the spleen, and the ileum at 21 days post-immunization, and the bacteria were not detected at 28 days post-immunization. These results demonstrate that both mutants can be eliminated from the chickens but induce strong cellular immune and humoral responses.

5. Conclusions

Our data demonstrated that the two mutants could significantly protect the chickens from virulent parental strain challenge. The lower virulence and the higher protective efficacy showed that CZ14-1∆spiC∆nmpC was better than CZ14-1∆spiC∆rfaL. However, for the eradication of the colonized parental strain in tissues, the group immunized with CZ14-1∆spiC∆rfaL was better than that immunized with CZ14-1∆spiC∆nmpC. Since the spiC deletion mutant of Salmonella has proven to be an appropriate vaccine candidate against S. Pullorum/S. Gallinarum infection [13,14], the present two double-mutants show potential for use as vaccines against S. Enteritidis infection in poultry.

Author Contributions

Conceptualization, Q.L. and X.J.; methodology, J.R. and Y.Z.; software, J.R. and C.Y.; validation, Z.Q., and H.X.; formal analysis, Y.Y.; investigation, Y.Z.; resources, J.R.; data curation, Y.Z.; writing—original draft preparation, Y.Z. and J.R.; writing—review and editing, Q.L.; visualization, Q.L. and S.G.; supervision, X.J.; project administration, Q.L.; funding acquisition, X.J.”.

Funding

This research was funded by the National Key Research and Development Program of China, grant no. 2017YFD0500705, 2017YFD0500100; National Natural Science Foundation of China, grant no. 31730094; Jiangsu province agricultural science and technology independent innovation funds, grant no. CX(16)1028; and The Priority Academic Program Development of Jiangsu Higher Education Institutions (PAPD).

Acknowledgments

We thank Hongying Chen, Rui Wang, and Ran Wang for their help with animal test.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Centers for Disease Control and Prevention. National Enteric Disease Surveillance: Salmonella Annual Report, 2016; Centers for Disease Control and Prevention: Atlanta, GA, USA, 2016.
  2. EFSA. The European Union summary report on trends and sources of zoonoses, zoonotic agents and food-borne outbreaks in 2017. EFSA J. 2018, 16, 5500. [Google Scholar]
  3. M’ikanatha, N.M.; Sandt, C.H.; Localio, A.R.; Tewari, D.; Rankin, S.C.; Whichard, J.M.; Altekruse, S.F.; Lautenbach, E.; Folster, J.P.; Russo, A. Multidrug-resistant Salmonella isolates from retail chicken meat compared with human clinical isolates. Foodborne Pathog. Dis. 2017, 7, 929–934. [Google Scholar] [CrossRef]
  4. Davies, R.H.; Breslin, M. Persistence of Salmonella Enteritidis phage type 4 in the environment and arthropod vectors on an empty free-range chicken farm. Environ. Microbiol. 2003, 5, 79–84. [Google Scholar] [CrossRef]
  5. Gast, R. Microbiology of shell egg production in the United State. In Producing Sage Eggs—Micorbial Ecology of Salmonella; Ricke, C., Richard, K., Eds.; Academic Press: London, UK, 2016; pp. 25–44. [Google Scholar]
  6. Tollefson, L.; Miller, M.A. Antibiotic use in food animals: Controlling the human health impact. J. AOAC Int. 2000, 83, 245–254. [Google Scholar] [PubMed]
  7. Djordjevic, S.P.; Cain, A.K.; Evershed, N.J.; Falconer, L.; Levings, R.S.; Lightfoot, D.; Hall, R.M. Emergence and evolution of multiply antibiotic-resistant Salmonella enterica serovar Paratyphi B D-tartrate-utilizing strains containing SGI1. Antimicrob. Agents Chemother. 2009, 53, 2319–2326. [Google Scholar] [CrossRef] [PubMed]
  8. Gonçalves, G.A.; Donato, T.C.; Baptista, A.A.; Corrêa, I.M.; Garcia, K.C.; Andreatti Filho, R.L. Bacteriophage-induced reduction in Salmonella Enteritidis counts in the crop of broiler chickens undergoing preslaughter feed withdrawal. Poult. Sci. 2014, 93, 216–220. [Google Scholar] [CrossRef] [PubMed]
  9. Hong, Y.; Schmidt, K.; Marks, D.; Hatter, S.; Marshall, A.; Albino, L.; Ebner, P. Treatment of Salmonella-contaminated eggs and pork with a broad-spectrum, single bacteriophage: Assessment of efficacy and resistance development. Foodborne Pathog. Dis. 2016, 13, 679–688. [Google Scholar] [CrossRef] [PubMed]
  10. Wigley, P.; Barrow, P.; Villareal-Ramos, B. Immunity to Salmonella in domestic (food animal) species. Vaccines against S. enterica infections. Vaccines for chickens. In Salmonella Infections. Clinical, Immunological and Molecular Aspects; Mastroeni, P., Maskell, D., Eds.; Cambridge University Press: Cambridge, UK, 2006; pp. 308–309. [Google Scholar]
  11. Fei, X.; Yin, K.; Yin, C.; Hu, Y.; Li, J.; Zhou, Z.; Tian, Y.; Geng, S.; Chen, X.; Pan, Z.; et al. Analyses of prevalence and molecular typing reveal the spread of antimicrobial-resistant Salmonella infection across two breeder chicken farms. Poult. Sci. 2018, 97, 4374–4383. [Google Scholar] [CrossRef] [PubMed]
  12. Wang, Y.; Cai, Y.; Zhang, J.; Liu, D.; Gong, X.; Pan, Z.; Geng, S.; Jiao, X. Controversy surrounding the function of spiC protein in Salmonella: An overview. Front. Microbiol. 2019, 10, 1784. [Google Scholar] [CrossRef]
  13. Geng, S.; Jiao, X.; Barrow, P.; Pan, Z.; Chen, X. Virulence determinants of Salmonella Gallinarum biovar Pullorum identified by PCR signature-tagged mutagenesis and the spiC mutant as a candidate live attenuated vaccine. Vet. Microbiol. 2014, 168, 388–394. [Google Scholar] [CrossRef]
  14. Cheng, Z.; Yin, J.; Kang, X.; Geng, S.; Hu, M.; Pan, Z.; Jiao, X. Safety and protective efficacy of a spiC and crp deletion mutant of Salmonella Gallinarum as a live attenuated vaccine for fowl typhoid. Res. Vet. Sci. 2016, 107, 50–54. [Google Scholar] [CrossRef] [PubMed]
  15. Guo, R.; Jiao, Y.; Li, Z.; Zhu, S.; Fei, X.; Geng, S.; Pan, Z.; Chen, X.; Li, Q.; Jiao, X. Safety, protective immunity, and diva capability of a rough mutant Salmonella Pullorum vaccine candidate in broilers. Front. Microbiol. 2017, 8, 547. [Google Scholar] [CrossRef] [PubMed]
  16. Van Oirschot, J.T.; Bruschke, C.J.; van Rijn, P.A. Vaccination of cattle against bovine viral diarrhorea. Vet. Microbiol. 1999, 64, 169–183. [Google Scholar] [CrossRef]
  17. Kong, Q.; Yang, J.; Liu, Q.; Alamuri, P.; Roland, K.L.; Curtiss, R., 3rd. Effect of deletion of genes involved in lipopolysaccharide core and o-antigen synthesis on virulence and immunogenicity of Salmonella enterica serovar Typhimurium. Infect. Immun. 2011, 79, 4227–4239. [Google Scholar] [CrossRef] [PubMed]
  18. Selke, M.; Meens, J.; Springer, S.; Frank, R.; Gerlach, G.F. Immunization of pigs to prevent disease in humans: Construction and protective efficacy of a Salmonella enterica serovar Typhimurium live negative-marker vaccine. Infect. Immun. 2007, 75, 2476–2483. [Google Scholar] [CrossRef] [PubMed]
  19. Pérez, J.M.; McGarry, M.A.; Marolda, C.L.; Valvano, M.A. Functional analysis of the large periplasmic loop of the Escherichia coli K-12 Waal O-antigen ligase. Mol. Microbiol. 2008, 70, 1424–1440. [Google Scholar] [CrossRef] [PubMed]
  20. Nagy, G.; Palkovics, T.; Otto, A.; Kusch, H.; Kocsis, B.; Dobrindt, U.; Engelmann, S.; Hecker, M.; Emody, L.; Pal, T.; et al. “Gently rough”: The vaccine potential of a Salmonella enterica regulatory lipopolysaccharide mutant. J. Infect. Dis. 2008, 198, 1699–1706. [Google Scholar] [CrossRef]
  21. Gil-Cruz, C.; Bobat, S.; Marshall, J.L.; Kingsley, R.A.; Ross, E.A.; Henderson, I.R.; Leyton, D.L.; Coughlan, R.E.; Khan, M.; Jensen, K.T.; et al. The porin OmpD from nontyphoidal Salmonella is a key target for a protective B1b cell antibody response. Proc. Natl. Acad. Sci. USA 2009, 106, 9803–9808. [Google Scholar] [CrossRef]
  22. Zhang, Y.; Dominguez-Medina, C.; Cumley, N.J.; Heath, J.N.; Essex, S.J.; Bobat, S.; Schager, A.; Goodall, M.; Kracker, S.; Buckley, C.D.; et al. IgG1 is required for optimal protection after immunization with the purified porin OmpD from Salmonella Typhimurium. J. Immunol. 2017, 199, 4103–4109. [Google Scholar] [CrossRef]
  23. Datsenko, K.A.; Wanner, B.L. One-step inactivation of chromosomal genes in Escherichia coli K-12 using PCR products. Proc. Natl. Acad. Sci. USA 2000, 97, 6640–6645. [Google Scholar] [CrossRef]
  24. Li, Q.; Hu, Y.; Xu, Y.; Chen, J.; Fang, L.; Liu, Z.; Jiao, X. A gene knock-in method used to purify plasmid pSPI12 from Salmonella enterica serovar Pullorum and characterization of IpaJ. J. Microbiol. Methods 2014, 98, 128–133. [Google Scholar] [CrossRef] [PubMed]
  25. Reed, L.J.; Muench, H. A simple method of estimating fifty percent endpoints. Am. J. Hyg. 1938, 27, 493–497. [Google Scholar]
  26. Li, Q.; Yuan, Y.; Wang, X.; Chen, J.; Wu, Y.; Wang, X.; Xu, L.; Yin, K.; Liu, Z.; Yin, C.; et al. Comparative study of Salmonella enterica serovar Enteritidis genes expressed within avian and murine macrophages via selective capture of transcribed sequences (SCOTS). Appl. Microbiol. Biotechnol. 2018, 102, 6567–6579. [Google Scholar] [CrossRef] [PubMed]
  27. Yin, C.; Xu, L.; Li, Y.; Liu, Z.; Gu, D.; Li, Q.; Jiao, X. Construction of pSPI12-cured Salmonella enterica serovar Pullorum and identification of IpaJ as an immune response modulator. Avian Pathol. 2018, 47, 410–417. [Google Scholar] [CrossRef] [PubMed]
  28. Woodward, M.J.; Gettinby, G.; Breslin, M.F.; Corkish, J.D.; Houghton, S. The efficacy of salenvac, a Salmonella enterica subsp. enterica serotype Enteritidis iron-restricted bacterin vaccine, in laying chickens. Avian Pathol. 2002, 31, 383–392. [Google Scholar] [CrossRef]
  29. Babu, U.; Dalloul, R.A.; Okamura, M.; Lillehoj, H.S.; Xie, H.; Raybourne, R.B.; Gaines, D.; Heckert, R.A. Salmonella Enteritidis clearance and immune responses in chickens following Salmonella vaccination and challenge. Vet. Immunol. Immunopathol. 2004, 101, 251–257. [Google Scholar] [CrossRef]
  30. Singh, B.R. Salmonella vaccines for animals and birds and their future perspective. Open Vaccine J. 2009, 2, 100–112. [Google Scholar] [CrossRef]
  31. Tran, T.Q.; Quessy, S.; Letellier, A.; Desrosiers, A.; Boulianne, M. Immune response following vaccination against Salmonella Enteritidis using 2 commercial bacterins in laying hens. Can. J. Vet. Res. 2010, 74, 185–192. [Google Scholar]
  32. Barrow, P.A. Salmonella infections: Immune and non-immune protection with vaccines. Avian Pathol. 2007, 36, 1–13. [Google Scholar] [CrossRef]
  33. Zhang-Barber, L.; Turner, A.K.; Barrow, P.A. Vaccination for control of Salmonella in poultry. Vaccine 1999, 17, 2538–2545. [Google Scholar] [CrossRef]
  34. Adriaensen, C.; De Greve, H.; Tian, J.Q.; De Craeye, S.; Gubbels, E.; Eeckhaut, V.; Van Immerseel, F.; Ducatelle, R.; Kumar, M.; Hernalsteens, J.P. A live Salmonella enterica serovar Enteritidis vaccine allows serological differentiation between vaccinated and infected animals. Infect. Immun. 2007, 75, 2461–2468. [Google Scholar] [CrossRef] [PubMed]
  35. Methner, U.; Barrow, P.A.; Berndt, A.; Rychlik, I. Salmonella Enteritidis with double deletion in phoP fliC--a potential live Salmonella vaccine candidate with novel characteristics for use in chickens. Vaccine 2011, 29, 3248–3253. [Google Scholar] [CrossRef] [PubMed]
  36. Ochman, H.; Soncini, F.C.; Solomon, F.; Groisman, E.A. Identification of a pathogenicity island required for Salmonella survival in host cells. Proc. Natl. Acad. Sci. USA 1996, 93, 7800–7804. [Google Scholar] [CrossRef] [PubMed]
  37. Uchiya, K.; Barbieri, M.A.; Funato, K.; Shah, A.H.; Stahl, P.D.; Groisman, E.A. A Salmonella virulence protein that inhibits cellular trafficking. EMBO J. 1999, 18, 3924–3933. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Zeng, L.; Li, B.; Yan, S.; Pan, Z.; Geng, S.; Jiao, X. Construction of genetically engineered attenuated vaccines Salmonella Entertidis C50041ΔspiCΔcrp. Chin. J. Zoonoses 2015, 31, 97–101. [Google Scholar]
  39. Mastroeni, P.; Chabalgoity, J.A.; Dunstan, S.J.; Maskell, D.J.; Dougan, G. Salmonella: Immune responses and vaccines. Vet. J. 2001, 161, 132–164. [Google Scholar] [CrossRef]
  40. Chausse, A.M.; Grepinet, O.; Bottreau, E.; Robert, V.; Hennequet-Antier, C.; Lalmanach, A.C.; Lecardonnel, J.; Beaumont, C.; Velge, P. Susceptibility to Salmonella carrier-state: A possible Th2 response in susceptible chicks. Vet. Immunol. Immunopathol. 2014, 159, 16–28. [Google Scholar] [CrossRef]
  41. Gal-Mora, O. Persistent infection and long-term carriage of typhoidal and nontyphoidal Salmonellae. Clin. Microbiol. Rev. 2019, 32, e00088–e000118. [Google Scholar] [CrossRef] [Green Version]
  42. Lalsiamthara, J.; Kamble, N.M.; Lee, J.H. A live attenuated Salmonella Enteritidis secreting detoxified heat labile toxin enhances mucosal immunity and confers protection against wild-type challenge in chickens. Vet. Res. 2016, 47, 60. [Google Scholar] [CrossRef]
Figure 1. Changes in growth ability of mutants in Luria-Bertani (LB) broth (a) and body weight of chickens post immunization (b). (a) The growth curves of the wild type stain CZ14-1, the mutant strains CZ14-1∆spiC∆nmpC and CZ14-1∆spiC∆rfaL in LB broth. Deletion of rfaL in CZ14-1∆spiC caused the decreased growth ability of S. Enteritidis in LB broth. (b) Changes in mean body weights of chickens after immunization. The vaccinated groups were intramuscularly inoculated with 1 × 105 CFU mutants in seven-day-old chickens, and control group received 100 μL phosphate-buffered saline (PBS). ** p ≤ 0.01; *** p ≤ 0.001.
Figure 1. Changes in growth ability of mutants in Luria-Bertani (LB) broth (a) and body weight of chickens post immunization (b). (a) The growth curves of the wild type stain CZ14-1, the mutant strains CZ14-1∆spiC∆nmpC and CZ14-1∆spiC∆rfaL in LB broth. Deletion of rfaL in CZ14-1∆spiC caused the decreased growth ability of S. Enteritidis in LB broth. (b) Changes in mean body weights of chickens after immunization. The vaccinated groups were intramuscularly inoculated with 1 × 105 CFU mutants in seven-day-old chickens, and control group received 100 μL phosphate-buffered saline (PBS). ** p ≤ 0.01; *** p ≤ 0.001.
Vaccines 07 00202 g001
Figure 2. Bacterial colonization in tissues and organs of immunized chickens. Chickens were immunized by parental strain CZ14-1, vaccine candidates CZ14-1∆spiC∆nmpC, and CZ14-1∆spiC∆rfaL, respectively (a). Bacterial colonization in liver (b), spleen (c), ileum (d), and cecum (e) of the immunized chickens. The number of bacteria was determined and expressed as log10CFU/g. Data are expressed as the mean ± SEM, * p ≤ 0.05; *** p ≤ 0.001.
Figure 2. Bacterial colonization in tissues and organs of immunized chickens. Chickens were immunized by parental strain CZ14-1, vaccine candidates CZ14-1∆spiC∆nmpC, and CZ14-1∆spiC∆rfaL, respectively (a). Bacterial colonization in liver (b), spleen (c), ileum (d), and cecum (e) of the immunized chickens. The number of bacteria was determined and expressed as log10CFU/g. Data are expressed as the mean ± SEM, * p ≤ 0.05; *** p ≤ 0.001.
Vaccines 07 00202 g002
Figure 3. Immune response in spleens from chickens immunized with vaccine candidates. The 7-day-old Hyline White chickens were immunized with 1 × 105 CFU of mutants (a), and the antisera were subjected to test IgG titers at 1, 7, 14, 21, 28, and 35 days post immunization (b). The total RNA was extracted from spleens of chickens vaccinated by mutants at 7, 14, and 21 days post immunization and subsequently subjected to the qRT-PCR analysis for detection the expression levels of IFN-γ (c), IL-6 (d), IL-1β (e), and CXCLi1 (f), respectively. ** p ≤ 0.01; *** p ≤ 0.001.
Figure 3. Immune response in spleens from chickens immunized with vaccine candidates. The 7-day-old Hyline White chickens were immunized with 1 × 105 CFU of mutants (a), and the antisera were subjected to test IgG titers at 1, 7, 14, 21, 28, and 35 days post immunization (b). The total RNA was extracted from spleens of chickens vaccinated by mutants at 7, 14, and 21 days post immunization and subsequently subjected to the qRT-PCR analysis for detection the expression levels of IFN-γ (c), IL-6 (d), IL-1β (e), and CXCLi1 (f), respectively. ** p ≤ 0.01; *** p ≤ 0.001.
Vaccines 07 00202 g003
Figure 4. Bacterial colonization in challenged chickens. The chickens were immunized with vaccine candidates CZ14-1∆spiC∆nmpC and CZ14-1∆spiC∆rfaL, respectively, and then challenged with 2 ×109 CFU of CZ14-1 (a). Bacterial colonization in liver (b), spleen (c), ileum (d), and cecum (e) of vaccinated chickens after challenge with parental strain CZ14-1. The number of bacteria was determined and is represented as log10 CFU/g. Data are expressed as the mean ± SEM, * p ≤ 0.05; ** p ≤ 0.01.
Figure 4. Bacterial colonization in challenged chickens. The chickens were immunized with vaccine candidates CZ14-1∆spiC∆nmpC and CZ14-1∆spiC∆rfaL, respectively, and then challenged with 2 ×109 CFU of CZ14-1 (a). Bacterial colonization in liver (b), spleen (c), ileum (d), and cecum (e) of vaccinated chickens after challenge with parental strain CZ14-1. The number of bacteria was determined and is represented as log10 CFU/g. Data are expressed as the mean ± SEM, * p ≤ 0.05; ** p ≤ 0.01.
Vaccines 07 00202 g004
Table 1. The LD50 of S. Enteritidis CZ14-1, CZ14-1∆spiC∆nmpC and CZ14-1∆spiC∆rfaL in three-day-old White Leghorn chickens after intramuscular immunization.
Table 1. The LD50 of S. Enteritidis CZ14-1, CZ14-1∆spiC∆nmpC and CZ14-1∆spiC∆rfaL in three-day-old White Leghorn chickens after intramuscular immunization.
StrainsChallenge Dose (CFU)No. of Deaths/Total No. of ChickensLD50 (CFU)
CZ14-11 × 10810/102.24 × 104
1 × 1079/10
1 × 1067/10
1 × 1056/10
1 × 1044/10
CZ14-1∆spiC∆nmpC1 × 101010/101.58 × 107
1 × 10910/10
1 × 10810/10
1 × 1075/10
1 × 1062/10
CZ14-1∆spiC∆rfaL1 × 101010/107.76 × 106
1 × 10910/10
1 × 10810/10
1 × 1077/10
1 × 1063/10
Table 2. The LD50 of S. Enteritidis CZ14-1, CZ14-1∆spiC∆nmpC and CZ14-1∆spiC∆rfaL in three-day-old Hyline White chickens after intramuscular immunization.
Table 2. The LD50 of S. Enteritidis CZ14-1, CZ14-1∆spiC∆nmpC and CZ14-1∆spiC∆rfaL in three-day-old Hyline White chickens after intramuscular immunization.
StrainsChallenge Dose (CFU)No. of Deaths/Total No. of ChickensLD50 (CFU)
CZ14-11 × 10810/101.26 × 104
1 × 10710/10
1 × 1069/10
1 × 1056/10
1 × 1045/10
CZ14-1∆spiC∆nmpC1 × 10910/102.57 × 107
1 × 10810/10
1 × 1075/10
1 × 1062/10
1 × 1050/10
CZ14-1∆spiC∆rfaL1 × 10910/104.07 × 106
1 × 10810/10
1 × 1076/10
1 × 1063/10
1 × 1050/10
Table 3. Protective efficiency of two vaccine candidates in Hyline White chickens.
Table 3. Protective efficiency of two vaccine candidates in Hyline White chickens.
GroupVaccinationNo.ChallengeSurvivors/Total (Survival Rate)
StrainDose (CFU)StrainRouteDose
ACZ14-1∆spiC∆nmpC1 × 10520CZ14-1i.m.a2 × 10916/20 (80%)
BCZ14-1∆spiC∆rfaL1 × 10520CZ14-1i.m.2 × 10915/20 (75%)
CPBS-20CZ14-1i.m.2 × 1091/20 (5%)
a represents the intramuscularly inoculation.

Share and Cite

MDPI and ACS Style

Li, Q.; Zhu, Y.; Ren, J.; Qiao, Z.; Yin, C.; Xian, H.; Yuan, Y.; Geng, S.; Jiao, X. Evaluation of the Safety and Protection Efficacy of spiC and nmpC or rfaL Deletion Mutants of Salmonella Enteritidis as Live Vaccine Candidates for Poultry Non-Typhoidal Salmonellosis. Vaccines 2019, 7, 202. https://doi.org/10.3390/vaccines7040202

AMA Style

Li Q, Zhu Y, Ren J, Qiao Z, Yin C, Xian H, Yuan Y, Geng S, Jiao X. Evaluation of the Safety and Protection Efficacy of spiC and nmpC or rfaL Deletion Mutants of Salmonella Enteritidis as Live Vaccine Candidates for Poultry Non-Typhoidal Salmonellosis. Vaccines. 2019; 7(4):202. https://doi.org/10.3390/vaccines7040202

Chicago/Turabian Style

Li, Qiuchun, Yue Zhu, Jingwei Ren, Zhuang Qiao, Chao Yin, Honghong Xian, Yu Yuan, Shizhong Geng, and Xinan Jiao. 2019. "Evaluation of the Safety and Protection Efficacy of spiC and nmpC or rfaL Deletion Mutants of Salmonella Enteritidis as Live Vaccine Candidates for Poultry Non-Typhoidal Salmonellosis" Vaccines 7, no. 4: 202. https://doi.org/10.3390/vaccines7040202

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop