Next Article in Journal
Electronic Package Leaflets for Vaccines: What Are People’s Perceptions in Italy?
Next Article in Special Issue
The National Strategies for and Challenges in Infection Prevention and Control of the Healthcare System in the Kingdom of Saudi Arabia (Review Study)
Previous Article in Journal
COVID-19 Vaccination Drive in a Low-Volume Primary Care Clinic: Challenges & Lessons Learned in Using Homegrown Self-Scheduling Web-Based Mobile Platforms
Previous Article in Special Issue
Mosquirix™ RTS, S/AS01 Vaccine Development, Immunogenicity, and Efficacy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Oral Administration with Recombinant Attenuated Regulated Delayed Lysis Salmonella Vaccines Protecting against Staphylococcus aureus Kidney Abscess Formation

State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, Hohhot 010070, China
*
Author to whom correspondence should be addressed.
Vaccines 2022, 10(7), 1073; https://doi.org/10.3390/vaccines10071073
Submission received: 10 May 2022 / Revised: 24 June 2022 / Accepted: 24 June 2022 / Published: 4 July 2022
(This article belongs to the Special Issue Infectious Disease Immunotherapy Research)

Abstract

:
Abscess formation is one of the main symptoms of Staphylococcus aureus infection. It is very important to inhibit abscess formation for preventing S. aureus persistent infection. To find a feasible solution, the live oral vaccines delivering S. aureus antigens, rEsxAB and rHlam, were constructed, which were based on the attenuated regulated delayed lysis Salmonella enterica subspecies Serovar Typhimurium strain χ11802, and the inhibiting effect on abscess formation was evaluated in mice kidneys. As the results showed, after oral administration, humoral immunity was induced via the mucosal route as the antigen-specific IgG in the serum and IgA in the intestinal mucus both showed significant increases. Meanwhile, the production of IFN-γ and IL-17 in the kidney tissue suggested that Th1/Th17-biased cellular immunity played a role in varying degrees. After challenged intravenously (i.v.) with S. aureus USA300, the χ11802(pYA3681−esxAB)-vaccinated group showed obvious inhibition in kidney abscess formation among the vaccinated group, as the kidney abscess incidence rate and the staphylococcal load significantly reduced, and the kidney pathological injury was improved significantly. In conclusion, this study provided experimental data and showed great potential for live oral vaccine development with the attenuated regulated delayed lysis Salmonella Typhimurium strains against S. aureus infection.

1. Introduction

Staphylococcus aureus, a commensal pathogen, is acquired both in the community and in hospitals [1]. S. aureus causes a wild range of diseases, from mild to life-threatening infections [1,2]. Abscess formation makes S. aureus infections persistent [3]. Additionally, it seems that kidney abscess is an important symptom of S. aureus infection [4,5,6]. With the abuse of antibiotics, the multidrug-resistant S. aureus strains continue to emerge and make it much more difficult to clinically treat infection, especially methicillin-resistant S. aureus (MRSA) and vancomycin-resistant S. aureus (VRSA) [7,8]. Therefore, the protective vaccine is a promising choice.
In recent years, virulent factors have shown potential as subunit vaccines against S. aureus infection, which present effective protection [9,10]. Among them, EsxA, EsxB, and Hla are promising candidate vaccines for inhibiting abscess formation [6,11]. EsxA and EsxB are secreted via the S. aureus ESAT-6 secretion system and play important roles in S. aureus murine abscess formation [12]. Their sequences remain highly conserved in the genome among different S. aureus strains [13]. What is more, when used as subunit vaccines and used to vaccinate mice by intraperitoneal (i.p.) injection, EsxA and EsxB both enable to induce Th1- and Th17-biased cellular immunity, which plays a major part in inhibiting S. aureus infection [14]. As Charalampia et al. reported, mice were challenged with esxA and esxB mutant S. aureus strains and showed a defect in abscess formation [12]. Hla (α-hemolysin) is a pore-forming cytotoxin binding to most eukaryotic cells and is possibly relevant to the pathogenesis of sepsis, pneumonia, and severe skin infection [15]. HlaH35L is an attenuated Hla mutant and was used to protect against S. aureus infections in recent studies [11,16]. As Adam et al. reported, HlaH35L showed its ability to moderate the severity of S. aureus skin infections and inhibited the abscess formation [11]. Furthermore, administration with 4C-Staph/alum including EsxAB (EsxA and EsxB fused for expression) and HlaH35L exhibits protective efficacy in four murine models, including kidney abscess, peritonitis, skin, and pneumonia models [6]. In our previous study, another Hla mutation (Hlam) was chosen, with the histidine mutated to leucine at the 285th aa, as a subunit vaccine. It also showed high protection in S. aureus lethal infection (data not shown). In this research, EsxAB and Hlam were chosen as delivered S. aureus antigens.
Live attenuated Salmonella enterica subspecies serovar Typhimurium strains (hereafter Salmonella Typhimurium) have been developed as delivery vectors, carrying heterologous antigens and used for immunization via the mucosal route [17,18,19]. There are several good traits with the recombinant attenuated Salmonella Typhimurium strains. Firstly, oral administration with live recombinant attenuated Salmonella Typhimurium strains provides needle-free, low-cost protection against disease, and the strains will survive a period under the stress of GI trace [19,20]. At the same time, the strains will elicit long-lasting protection with mucosal and cell-mediated immune responses, against a range of pathogens [21,22]. Secondly, they can also invade and colonize the deep effector lymphoid tissues, which is essential for inducing memory T cells [23]. Thirdly, the balanced-lethal system design of the live attenuated Salmonella Typhimurium strains eliminates the drug-resistance markers, and the plasmid vector can be stably maintained with heterologous antigen expression [24]. Fourthly, Salmonella Typhimurium outer membrane proteins, flagellin, and its ghosts have been developed as adjuvants enhancing antigen-specific immune response [25,26,27]. Moreover, the regulated delayed lysis system offered another means for attenuation and added safety features to confer biological containment. This system allows attenuated Salmonella Typhimurium strains to colonize in the tissue only if the araC PBAD promoter is activated with an exogenous source of arabinose supplied [28]. After the cells of attenuated Salmonella Typhimurium strains replicate for 6–10 rounds, the arabinose accumulated within the bacterial cells will be exhausted [29]. At the same time, the C2-regulated promoter, P22 PR will work and antisense mRNA synthesis will block the residual mRNA of asdA and murA, which encode the essential enzymes for the bacterial cell wall [28]. These concerted activities eventually result in cell lysis. The details of this system have been described elsewhere [28,30].
In this study, attenuated regulated delayed lysis Salmonella Typhimurium strain χ11802 was utilized as an antigen delivery vector, expressing S. aureus EsxAB (rEsxAB) and Hlam (rHlam), respectively. This strategy emphasized the T-cell-mediated immunity of the oral vaccines. After BALB/c mice were orally administrated with these recombinant attenuated Salmonella Typhimurium strains, and then challenged with S. aureus USA300 by i.v., the inhibition efficacy of kidney abscess formation was evaluated. As a result, among all the vaccinated groups, the χ11802(pYA3681−esxAB)-vaccinated group showed obvious inhibition in kidney abscess formation. These results showed the great potential for the live oral vaccine development with the attenuated regulated delayed lysis Salmonella Typhimurium strains against S. aureus infection.

2. Materials and Methods

2.1. Mice and Ethics Statement

The 6–8-week-old specific-pathogen-free (SPF) female BALB/c mice were purchased from Beijing Vital River Laboratory Animal Technology Co., Ltd. (Beijing, China), and fed under SPF conditions. All animal-related experimental protocols applied in this study were conducted under the standards of the Ethics Committee of Inner Mongolia Medical University (SCXK2016-0001).

2.2. Bacterial Strains and Growth Conditions

Attenuated regulated delayed lysis Salmonella Typhimurium strain χ11802 and Escherichia coli strain χ6212 were kindly provided by the research team of Dr. Roy Curtiss III, together with the pYA3681 plasmid. All the strains and plasmids are listed in Table 1. χ11802 and χ6212 were cultured in Luria–Bertani (LB) broth or on an agar plate under aerobic condition at 37 °C, both needed supplementing with 50 μg/mL Diaminopimelic acid (DAP), and 0.02% w/v arabinose was additionally needed for χ11802 growth. S. aureus USA300 was reserved in our laboratory and cultured in LB broth or agar plate at 37 °C under aerobic conditions overnight.

2.3. Construction of rHlam and rEsxAB Delivery Vector Based on Attenuated Regulated Delayed Lysis Salmonella Typhimurium Strain χ11802

The ESAT-6 secretory proteins, EsxA and EsxB, were merged with a flexible linker, (GGGGS)3. Then, S. aureus antigen genes, the esxAB, and the hlam, were optimized and synthesized into cloning vector pUC57, respectively, by GenScript Biotech (Nanjing, China), according to the codon bias of Salmonella Typhimurium. Then, the blaSS (β-lactamase type II signal sequence) together with the HA-tag fragment was fused to esxAB and hlam for heterologous protein secretion [31]. Antigen gene fragments in pUC57 and linearized pYA3681 plasmid vector were harvested by being digested with restriction enzymes, NcoI and SmaI. The antigen gene fragments were fused into pYA3681 with T4 DNA ligase. Then, the recombinant plasmids were introduced into competent χ6212 by the heat-shock method, and the bacteria cells were coated on the LB agar plate and cultured overnight at 37 °C under aerobic conditions. The colonies were inoculated into the LB broth for recombinant plasmid harvest. After the plasmids were double-enzyme digested (NcoI and SmaI) and sequenced for verification, they were finally introduced into χ11802 competent cells, and the recombinant strains of χ11802 were grown on the LB agar plate containing 0.2% w/v arabinose under the same condition as described above. Positive clones were recognized by double-enzyme digestion and sequencing of the recombinant plasmids in χ11802. All recombinant strains were then stored at −80 °C for later experiments.

2.4. SDS-PAGE and Western Blot

SDS-PAGE and Western blot were conducted following standard methods. After recombinant strains of χ11802 were induced by Isopropyl-β-D-thiogalactopyranoside (IPTG, 1 mg/mL), pellets were harvested by centrifugation at 5000× g, then resuspended with phosphate-buffered saline (PBS) solution. The secreted proteins in the medium were 20-fold concentrated by Trichloroacetic Acid (TCA) precipitation and washed 3 times with PBS solution. Bacterial pellets were ultrasonic crashed at a low temperature. All samples were mixed with pre-stained protein-loading buffer, boiled for 10 min, and analyzed by 12% SDS-PAGE. Protein samples in the polyacrylamide gel were transferred to the 0.22 μm PVDF membrane and blocked for 1 h at room temperature with 5% w/v skimmed milk/TBST solution (tris buffered saline solution containing 0.2% v/v Tween-20). PVDF membranes were incubated at 4 °C overnight with mouse anti-HA tag monoclonal antibody (Proteintech; Rosemont, IL, USA, used at 1:10,000). After washing 3 times with TBST solution, polyvinylidene difluoride (PVDF) membranes were incubated with HRP-conjugated goat anti-mouse IgG secondary antibody (Proteintech; Rosemont, IL, USA, used at 1:5000) for 1 h at room temperature. Then, the membranes were washed 3 times with TBST as before. The protein bands were visualized with ECL Western blotting substrate (Thermo Fisher Scientifics, Waltham, MA, USA) and the signal was analyzed by the ChemiScope chemiluminescence imaging system (CLINX, Shanghai, China).

2.5. The Dependence of Recombinant Attenuated Regulated Delayed Lysis Salmonella Typhimurium Strains on Arabinose In Vitro and In Vivo

Recombinant attenuated regulated delayed lysis Salmonella Typhimurium strains, χ11802(pYA3681−esxAB) and χ11802(pYA3681-hlam), were streaked on the LB agar plate with or without 0.02% w/v arabinose, incubated at 37 °C overnight. Then, the single bacterial colony of recombinant strains on the LB agar plate (with arabinose) was inoculated into LB broth (without arabinose), as the first generation. Then, the first generation was subcultured with a series of gradient concentrations, 1:10, 1:100, and 1:1000 v/v, as the second generation. If there was no obvious growth inhibition, bacteria from 1:10 subcultured group was subcultured again with a series of gradient concentrations, 1:10, 1:100, and 1:1000 v/v, as the third generation. This step would be repeated until obvious bacterial growth inhibition appeared. At the same time, the recombinant strains of χ11802 from the LB broth (without arabinose) or 1:10 subcultured group were also subcultured in LB broth with 0.02% w/v arabinose as the positive control.
After deprivation of food and water for 5 h, the BALB/c mice were each orally administrated with ~3 × 108 CFU recombinant strains of χ11802, which expressed EsxAB or Hlam and were resuspended in 100 μL PBS solution. These strains were cultured in LB broth with 0.02% w/v arabinose, centrifuged, and resuspended with PBS solution. Then, 2 h later, food and water were returned to the mice. On days 1, 7, 14, and 28 after vaccination, the intestine, stomach, mesenteric lymph nodes (MLN), spleen, and Peyer’s Patches were collected from every 3 mice for the bacterial loading evaluation. All tissues were homogenized, and the numerical difference in bacterial colonies on a selenite cystine agar plate with or without 0.02% w/v arabinose was regarded as the number of arabinose-dependent strains.

2.6. BALB/c Mice Orally Administrated with Recombinant Attenuated Regulated Delayed Lysis Salmonella Typhimurium Strains

BALB/c mice were randomly divided into 4 groups, initially. There were 19 mice in each group and orally administrated at the same time with the PBS solution, χ11802(pYA3681), χ11802(pYA3681−esxAB), or χ11802(pYA3681−hlam). Each of the strains were ~3 × 108 CFU resuspended in 100 μL PBS solution. Two weeks after primary immunization, all groups were booster immunized. Nine in each group were used for evaluating the antigen-specific IgA in the intestinal mucosa, the Interferon-γ (IFN-γ) and interleukin 17 (IL-17) in the tissue of the kidneys and spleens. The other mice, whose serum-specific IgG was measured by enzyme-linked immunosorbent assay (ELISA), were utilized for S. aureus USA300 challenge experiments. The food and water were withdrawn and returned as previously described.

2.7. rEsxAB- and rHlam-Specific IgG Level in the Serum Measured by ELISA

Sera were collected on day 14 after primary and booster immunization, respectively. Blood samples were incubated at 37 °C for 2 h and centrifuged for serum. The sera were stored at −20 °C for later detection. The 96-well MICROLON ELISA plates were coated with 100 μL protein diluent (5 μg/mL rEsxAB or rHlam protein, extracted in the previous study), and placed at 4 °C overnight. Then, the plates were washed 3 times with PBST (PBS with 0.05% v/v Tween-20) solution, blocked with 5% BSA at 37 °C for 2 h, and further washed 3 times with PBST solution. Corresponding to the coated protein, 100 μL/well diluted sera (1:100 dilution with PBS solution) were added, and the plates were incubated at 37 °C for 2 h. After washing with PBST solution 5 times, HRP-conjugated goat anti-mouse IgG (Proteintech) solution (1:2000 dilution with 1% BSA) was added to the wells, and the plates were incubated at 37 °C for 2 h. Finally, after washing another 5 times with PBST solution, 100 μL/well TMB single-component substrate (Solarbio, Beijing, China) solution was added, with the incubation at 37 °C for 2 h. Then, 50 μL/well ELISA stop solution (Solarbio, Beijing, China) was added, and absorbance was measured at OD450.

2.8. rEsxAB- and rHlam-Specific IgA in the Mouse Intestinal Mucosa Measured by ELISA

Intestinal mucus was collected on day 7 after booster immunization. Each sample (each ~0.05 g) was suspended with 500 μL PBS buffer. After centrifuging, the supernatant was collected and then stored at −20 °C. The methods of rEsxAB- and rHlam-specific IgA detection were mostly the same as previously described in Section 2.7, while the secondary antibody was HRP-conjugated goat anti-mouse IgA (Proteintech).

2.9. IFN-γ and IL-17 Levels in the Kidney and Spleen Tissue

IFN-γ and IL-17 levels in the tissue of BALB/c mouse kidneys and spleens were measured by ELISA. Then, 1 mL PBS solution was added to 0.1 g tissue. After homogenization and centrifugation, the supernatant was used for IFN-γ and IL-17 detection by double antibody sandwich method with the ELISA kit (R&D Systems, Minneapolis, MN, USA). Most practices were the same as antibody detection.

2.10. BALB/c Mice Challenged with S. aureus USA300

On day 14 after the booster immunization, all BALB/c mice were intravenous injected (i.v.) with S. aureus USA300 of 5 × 107 CFU in 50 μL PBS solution. Additionally, they were finally sacrificed after 14-day continuous observation. Then, the abscess incidence, bacterial loading, and histopathological damage in the kidneys were evaluated.

2.11. Statistical Analysis

Data were analyzed with GraphPad Prism software and the R. Shapiro–Wilk test was used for testing normality. IgG and IgA levels were presented as the mean ± SD and analyzed by t-test. IFN-γ and IL-17 levels were presented as the mean ± SD and analyzed by one-way ANOVA and Tukey’s multiple-comparison test. Kruskal–Wallis test and Dunn’s multiple-comparison test were utilized for bacterial loading analysis and the proportion of abscess area analysis as part of the data did not meet the normal distribution.

3. Results

3.1. rEsxAB and rHlam Synthesis in Recombinant Attenuated Regulated Delayed Lysis Salmonella Typhimurium Strain χ11802 and Secretion into the Medium

rEsxAB and rHlam expression strains were constructed with regulated delayed lysis attenuated Salmonella Typhimurium strain χ11802. The schemes of recombinant plasmids are shown in the Figure 1A,B. Extracted from recombinant strains of χ11802, the plasmids were verified by double-enzyme digestion with NcoI and SmaI, and the esxAB (Figure 1C, Lane 2) and hlam (Figure 1D, Lane 2) gene fragments were consistent with their expected length. The sequencing results further confirmed these (data not shown). The β-lactamase secretion signal peptide (coded by blaSS) was added to the N-terminal of the antigen proteins, which aims to transfer the proteins to the periplasmic space via the type II secretion system and eventually released to the outside [32]. Western blot showed that rEsxAB and rHlam were synthesized in χ11802, and the recombinant proteins were released into the medium (Figure 1E,F).

3.2. The Survival of Recombinant Attenuated Regulated Delayed Lysis Salmonella Typhimurium Strains Strictly Dependent on Arabinose In Vitro and In Vivo

Recombinant attenuated regulated delayed lysis Salmonella Typhimurium strains required an exogenous source of arabinose for colonization and survival [28]. However, it was unclear whether this would be different or add side effects to the recombinant strains after S. aureus antigen genes were inserted into the pYA3681 plasmid. These were verified in three steps. Firstly, recombinant strains, χ11802(pYA3681−esxAB) and χ11802(pYA3681−hlam), were streaked on LB agar plate with or without 0.02% w/v arabinose. As a result, the LB agar plate with arabinose enabled the recombinant strains to grow well, while it showed growth inhibition on the arabinose-free LB agar plate (Figure 2A). This indicated that arabinose must be provided for recombinant strain growth. Secondly, considering that the recombinant strains would colonize in mice, we inoculated the recombinant strains of χ11802 into LB broth to determine growth restrictions in an arabinose-free liquid environment. The results showed that the recombinant strains could only be subcultured to the second generation in arabinose-free LB broth, compared with their growth status in LB broth with arabinose (Figure 2B). This provided a basis for the growth restriction of recombinant strains in the host. Thirdly, the BALB/c mice were orally administrated with recombinant strains to determine how long the recombinant strains would survive in vivo. On days 1, 7, 14, and 28 after oral administration, bacterial loading of the recombinant strains of χ11802 was evaluated. Although it showed that there were individual differences in mice, and recombinant strains migrated into MLN (mesenteric lymph nodes), spleen, and Peyer’s patch to a certain degree, the recombinant strains of χ11802 could be cleaned up on day 28 after oral administration. In the meantime, all vaccinated mice stayed in a good state.

3.3. Recombinant Attenuated Regulated Delayed Lysis Salmonella Typhimurium Strains Eliciting Serum IgG- and Intestinal Mucus IgA-Mediated Immune Responses

To determine the humoral and mucosal immunity elicited by recombinant attenuated regulated delayed lysis Salmonella Typhimurium strains, the serum IgG and intestinal mucus IgA were evaluated by ELISA. On day 14 after primary and booster immunization, mouse serum was collected and rEsxAB- and rHlam-specific IgG were measured. The χ11802(pYA3681−esxAB)-vaccinated group and χ11802(pYA3681−hlam)-vaccinated group both showed that IgG level was significantly higher than the χ11802(pYA3681)-vaccinated group (Figure 3A–D). This indicated that orally administrated recombinant attenuated Salmonella Typhimurium strains successfully elicited humoral immunity. These vaccinated BALB/c mice (n = 10) were then challenged with S. aureus USA300.
On day 7 after the booster immunization, the remaining vaccinated BALB/c mice (n = 9) in each group were evaluated using mucus IgA. It showed that the IgA level was also significantly higher than that in the χ11802(pYA3681)-vaccinated group in the intestinal mucus of the χ11802(pYA3681−esxAB)-vaccinated group and χ11802(pYA3681−hlam)-vaccinated group, which suggested that mucosal immunity was elicited (Figure 3E,F). These kidneys and spleens of mice were then used for measuring IL-17 and IFN-γ levels.

3.4. IL-17 and IFN-γ Production Elicited by Recombinant Attenuated Regulated Delayed Lysis Salmonella Typhimurium Strains

In previous research, Th1- and Th17-mediated immunity were indicated to be very important for protection against S. aureus infection [14,33]. Therefore, the IL-17 and IFN-γ levels in the kidney and the spleen were measured. After booster immunization, the IL-17 and IFN-γ levels of the χ11802(pYA3681−esxAB)-vaccinated group were significantly higher than those of the χ11802(pYA3681)-vaccinated group in the kidney (Figure 4A,B). The IFN-γ level of the χ11802(pYA3681−hlam)-vaccinated group was also significantly higher than the χ11802(pYA3681)-vaccinated group. Regarding the production of IL-17 and IFN-γ in mouse spleen, they showed no significant increase (Figure S1A,B). These results indicated that Th1- and Th17-biased cellular responses were elicited after oral administration with χ11802(pYA3681−esxAB). Oral administration with χ11802(pYA3681− hlam) could elicit a Th1 cellular response while showing a weak effect for the Th17 cellular response.

3.5. Mouse Kidney Abscess Formation after Challenged with S. aureus USA 300

After challenge with S. aureus USA300 by i.v., all BALB/c mouse kidneys were collected on day 14 (Figure 5A,B). There were 10 mice in each vaccinated group initially, except 1 died in the χ11802(pYA3681−esxAB)-vaccinated group before challenged. The kidney abscess formation was evaluated, and the degree of abscesses was summarized into four patterns (Figure 5C). The abscess area proportion of the whole kidney surface was approximately estimated. As the results showed, the χ11802(pYA3681−esxAB)-vaccinated group presented a relatively low incidence of kidney abscess formation with a significant difference against the χ11802(pYA3681)-vaccinated group (Figure 5D), and the staphylococcal load was also significantly decreased in contrast with the χ11802(pYA3681)-vaccinated group (Figure 5E). However, the χ11802(pYA3681−hlam)-vaccinated group did not present an obvious inhibiting effect on abscess formation (Figure 5D,E).

3.6. Kidney Tissue Lesion after BALB/c Mice Challenged with S. aureus USA 300

The degree of kidney tissue lesion was further evaluated by kidney pathology slides, which were stained with hematoxylin and eosin (H&E) (Figure 6). The kidney pathological sections of the mice without vaccination and challenge were used as the blank control (Control Check). All BALB/c mice showed a varying degree of kidney injury after challenge with S. aureus USA300. Among them, the kidney injury degree of the χ11802(pYA3681−esxAB)-vaccinated group was the least. The PBS group, the χ11802(pYA3681)-vaccinated group, and the χ11802(pYA3681−hlam)-vaccinated group all presented atrophy of kidney tubule epithelial cells, interstitial edema, and neutrophil infiltration. However, the χ11802(pYA3681−esxAB)-vaccinated group showed improvement.

4. Discussion

As an opportunistic pathogen, S. aureus causes a wide range of diseases. In most cases, antibiotics are utilized for the treatment of S. aureus infection. However, the multiple resistant strains reduce the effectiveness of the antibiotic. What is worse, the abscess formation will make it more difficult to cure [3]. Previous studies have mentioned that kidney abscess is an important symptom of S. aureus infection [4,5,6]. In this study, we attempted to develop a kind of protective vaccine to inhibit abscess formation and eventually prevent S. aureus infection. It has been confirmed that intracellular persistence makes S. aureus escape from professional phagocytes and extracellular antibiotics [34]. Therefore, we aimed to emphasize the cooperation of humoral immunity and cellular immunity in this study.
The S. aureus antigens selected in the research were experimentally proved to be able to inhibit or moderate the abscess formation [11,12]. The Hla mutation (Hlam) was used as delivered antigen according to its immune protective effect in our previous experimental data. S. aureus EsxA and EsxB as subunit vaccines, utilized for immunization alone or together, were able to promote the induction of Th1- and Th17-biased immune responses [14]. However, the disadvantage of subunit vaccines is obvious: the proteins need to be extracted at a high cost. A low-cost and easy-to-use method is desired for immunization, which synthesizes and delivers antigens at the same time with no compromised immune protective effect. Live attenuated Salmonella Typhimurium seems to be a choice.
Live attenuated Salmonella Typhimurium has been developed as a mature delivery vector. As live oral vaccines, there has been meaningful progress in antibacterial [29,35], antiviral [36], antiparasitic [37,38], and antitumor [39] research. Though with good features as delivery vaccines, live attenuated Salmonella Typhimurium strains remain potential side effects. Meanwhile, there is no clear evidence showing how recombinant proteins will affect bacterial properties. Therefore, the dependence of the recombinant strains on arabinose was tested in vitro and in vivo. The results showed that arabinose was necessarily externally provided for the growth of recombinant strains (Figure 2A,B). This provided a basis that, after oral administration, the recombinant strains would be gradually cleaned up in the host. Further research showed that the recombinant strains migrated into MLN (mesenteric lymph nodes), spleen, and Peyer’s patch to a certain degree; however, eventually cleared up within 28 days after oral administration (Figure 2C). During observation, all the vaccinated BALB/c mice stayed in a good state. These indicated that oral administration with χ11802(pYA3681−esxAB) and χ11802(pYA3681−hlam) would not cause obvious damage to the host.
Humoral immunity and cellular immunity are important against pathogen invasions [40]. After oral administration with the recombinant strains, the humoral immune response was elicited at varying levels via the mucosal route in the host. rEsxAB- and rHlam-specific IgG in the serum, as well as IgA in the intestinal mucosa, were detected. In the early stage of S. aureus infection, specific antibodies and complement components in the serum play critical roles in the initiation of phagocytosis [41]. When S. aureus invades the extracellular, rEsxAB- and rHlam-specific IgG could be crucial for binding and neutralizing. However, in previous studies, S. aureus EsxA and EsxB as subunit vaccines, utilized for immunization individually or together elicited high titers of specific antigens, but they did not prevent infection, which other immune mechanisms must compensate for protection [14]. As research showed, Th1- and Th17-biased immune responses worked in S. aureus infection [42], and live attenuated Salmonella Typhimurium could elicit specific cellular immunity [28].
As previous studies have mentioned, S. aureus can invade host cells, and the intracellular S. aureus persists and replicates, some of which can enter the cytoplasm [34,43,44]. The intracellular S. aureus will release after phagocyte lysis and serves as a potential route for dissemination of infection [45]. Once S. aureus survives in phagocytes, to eliminate the intracellular pathogen, T helper cells, cytotoxic T cells, natural killer cells, and cytokine IFN-γ are required [46]. IFN-γ is the key cytokine to developing Th1-biased cellular immune responses [47]. Th17 cells could mediate serotype-independent protection, including recruiting neutrophils and macrophages to mucosal sites and activating B cell antibody responses [48]. Moreover, in the mucosal tissues, long-life Th17 effector memory cells had potential against pathogens [49]. In this study, IFN-γ and IL-17 in the kidney and spleen of BALB/c mice were evaluated after booster immunization. The IFN-γ level of kidney tissue in the χ11802(pYA3681−esxAB)-vaccinated group and the χ11802(pYA3681−hlam)-vaccinated group were significantly higher than that in the χ11802(pYA3681)-vaccinated group, and the IL-17 level of kidney tissue in the χ11802(pYA3681−esxAB)-vaccinated group was also significantly higher than that in the χ11802(pYA3681)-vaccinated group (Figure 4A,C). The previous study has reported that live attenuated Salmonella typhimurium vaccines delivering S. aureus antigens could elicit Th1- and Th17-biased cellular immune responses [50]. It could be inferred that, in this study, Th1/Th17 cellular immune responses could play essential roles in S. aureus infection. However, findings on IFN-γ and IL-17 levels in mouse spleen remained inconclusive in the kidneys, which was worth further exploration (Figure S1A,B).
After BALB/c mice were challenged with S. aureus USA300 by i.v., all of them were evaluated for kidney abscess formation on day 14 (Figure 5A,B). Among all the vaccinated groups, the χ11802(pYA3681−esxAB)-vaccinated group showed obvious inhibition in abscess formation. This is consistent with the subunit vaccine efficacy of EsxA and EsxB for inhibiting kidney abscess formation [12,14]. However, the χ11802(pYA3681−hlam)-vaccinated group did not show distinctive inhibition efficacy on abscess formation. As the protective effects of Hla were presented in murine pneumonia, χ11802(pYA3681−hlam) potential deserves further development.
This study showed that oral administration with χ11802(pYA3681−esxAB) and χ11802(pYA3681−hlam) could elicit humoral immunity and varying degrees of Th1/Th17 cellular immune response in the host. However, only χ11802(pYA3681−esxAB) could effectively inhibit abscess formation in the kidney. There are still several limitations that should be mentioned in this study. Firstly, only the S. aureus USA300 strain was used for the challenge experiment, and other serotypes of S. aureus should also be tested to obtain more common conclusions. Secondly, this study only showed the efficacy of recombinant attenuated regulated delayed lysis Salmonella Typhimurium strains as oral vaccines in inhibiting kidney abscess formation and some basic effects on the host immune system; the immune mechanism remains further studied.
In summary, our research showed the great potential of recombinant attenuated regulated delayed lysis Salmonella Typhimurium strains as oral vaccines inhibiting S. aureus abscess formation.

5. Conclusions

Oral administration with recombinant regulated delayed lysis attenuated Salmonella Typhimurium strain χ11802 delivering S. aureus EsxAB can elicit humoral, Th1- and Th17-mediated cellular immunity, and shows the significant inhibition of kidney abscess formation. This study provided experimental data and showed the great potential for the live oral vaccine development with regulated lysis attenuated Salmonella Typhimurium strains against S. aureus infection.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/vaccines10071073/s1, Figure S1: IFN-γ and IL-17 production in mouse spleen tissue elicited by oral administration with the recombinant strains of χ11802.

Author Contributions

Investigation, H.L., X.B. and S.S.; methodology, Y.L. (Yanchen Liang), Y.L. (Yang Liu) and H.Z.; software, N.P.; writing—review and editing, X.W. and Y.L. (Yanchen Liang); project administration, X.W. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Science and Technology Major Project of the Inner Mongolia Autonomous Region of China (grant numbers 2021ZD0013), Key Scientific and Technological Research Program of Inner Mongolia Autonomous Region (grant numbers 2021GG0156), and the National Natural Science Foundation of China (grant numbers 32060800).

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki. All animal-related experimental protocols applied in this study were conducted under the standards of the Ethics Committee of Inner Mongolia Medical University (SCXK2016-0001).

Informed Consent Statement

Not applicable.

Data Availability Statement

All data that this study is based upon are available from the corresponding author upon request.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Lowy, F.D. Staphylococcus aureus Infections. N. Engl. J. Med. 1998, 339, 520–532. [Google Scholar] [CrossRef] [PubMed]
  2. Sheets, A. Emergence of community-acquired methicillin-resistant Staphylococcus aureus USA 300 clone as the predominant cause of skin and soft-tissue infections. J. Emerg. Med. 2006, 31, 235. [Google Scholar] [CrossRef]
  3. Cheng, A.G.; Kim, H.K.; Burts, M.L.; Krausz, T.; Schneewind, O.; Missiakas, D.M. Genetic requirements for Staphylococcus aureus abscess formation and persistence in host tissues. FASEB J. 2009, 23, 3393–3404. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Rauch, S.; Dedent, A.C.; Kim, H.K.; Wardenburg, J.B.; Missiakas, D.M.; Schneewind, O. Abscess Formation and Alpha-Hemolysin Induced Toxicity in a Mouse Model of Staphylococcus aureus Peritoneal Infection. Infect. Immun. 2012, 80, 3721–3732. [Google Scholar] [CrossRef] [Green Version]
  5. Kim, H.K.; Dedent, A.; Cheng, A.G.; Mcadow, M.; Bagnoli, F.; Missiakas, D.M.; Schneewind, O. IsdA and IsdB antibodies protect mice against Staphylococcus aureus abscess formation and lethal challenge. Vaccine 2010, 28, 6382–6392. [Google Scholar] [CrossRef] [Green Version]
  6. Bagnoli, F.; Fontana, M.R.; Soldaini, E.; Mishra, R.P.N.; Fiaschi, L.; Cartocci, E.; Nardi-Dei, V.; Ruggiero, P.; Nosari, S.; De Falco, M.G.; et al. Vaccine composition formulated with a novel TLR7-dependent adjuvant induces high and broad protection against Staphylococcus aureus. Proc. Natl. Acad. Sci. USA 2015, 112, 3680–3685. [Google Scholar] [CrossRef] [Green Version]
  7. Turner, N.A.; Sharma-Kuinkel, B.K.; Maskarinec, S.A.; Eichenberger, E.M.; Shah, P.P.; Carugati, M.; Holland, T.L.; Fowler, V.G. Methicillin-resistant Staphylococcus aureus: An overview of basic and clinical research. Nat. Rev. Microbiol. 2019, 17, 203–218. [Google Scholar] [CrossRef]
  8. Cong, Y.; Yang, S.; Rao, X. Vancomycin resistant Staphylococcus aureus infections: A review of case updating and clinical features. J. Adv. Res. 2020, 21, 169–176. [Google Scholar] [CrossRef]
  9. Salgado-Pabón, W.; Schlievert, P.M. Models matter: The search for an effective Staphylococcus aureus vaccine. Nat. Rev. Microbiol. 2014, 12, 585–591. [Google Scholar] [CrossRef]
  10. Spaulding, A.R.; Salgado-Pabón, W.; Merriman, J.A.; Stach, C.S.; Ji, Y.; Gillman, A.N.; Peterson, M.L.; Schlievert, P.M. Vaccination against Staphylococcus aureus pneumonia. J. Infect. Dis. 2014, 209, 1955–1962. [Google Scholar] [CrossRef] [Green Version]
  11. Kennedy, A.D.; Wardenburg, J.B.; Gardner, D.J.; Long, D.; Whitney, A.R.; Braughton, K.R.; Schneewind, O.; Deleo, F.R. Targeting of Alpha-Hemolysin by Active or Passive Immunization Decreases Severity of USA300 Skin Infection in a Mouse Model. J. Infect. Dis. 2010, 202, 1050–1058. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Burts, M.L.; Williams, W.A.; Debord, K.; Missiakas, D.M. EsxA and EsxB are secreted by an ESAT-6-like system that is required for the pathogenesis of Staphylococcus aureus infections. Proc. Natl. Acad. Sci. USA 2005, 102, 1169–1174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Korea, C.G.; Balsamo, G.; Pezzicoli, A.; Merakou, C.; Tavarini, S.; Bagnoli, F.; Serruto, D.; Unnikrishnan, M. Staphylococcal Esx Proteins Modulate Apoptosis and Release of Intracellular Staphylococcus aureus during Infection in Epithelial Cells. Infect. Immun. 2014, 82, 4144–4153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Zhang, B.Z.; Hua, Y.H.; Yu, B.; Lau, C.C.Y.; Cai, J.P.; Zheng, S.Y.; Yam, W.C.; Kao, R.Y.T.; Sze, K.H.; Zheng, B.J.; et al. Recombinant ESAT-6-Like Proteins Provoke Protective Immune Responses against Invasive Staphylococcus aureus Disease in a Murine Model. Infect. Immun. 2015, 83, 339–345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Wilke, G.A.; Bubeck Wardenburg, J. Role of a disintegrin and metalloprotease 10 in Staphylococcus aureus alpha-hemolysin-mediated cellular injury. Proc. Natl. Acad. Sci. USA 2010, 107, 13473–13478. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Wardenburg, J.B.; Schneewind, O. Vaccine protection against Staphylococcus aureus pneumonia. J. Exp. Med. 2008, 205, 287–294. [Google Scholar] [CrossRef] [PubMed]
  17. Curtiss, R., 3rd; Nakayama, K.; Kelly, S.M. Recombinant avirulent Salmonella vaccine strains with stable maintenance and high level expression of cloned genes in vivo. Immunol. Investig. 1989, 18, 583–596. [Google Scholar] [CrossRef]
  18. Cárdenas, L.; Clements, J.D. Oral immunization using live attenuated Salmonella spp. as carriers of foreign antigens. Clin. Microbiol. Rev. 1992, 5, 328–342. [Google Scholar] [CrossRef]
  19. Kong, W.; Clark-Curtiss, J.; Curtiss Iii, R. Utilizing Salmonella for antigen delivery: The aims and benefits of bacterial delivered vaccination. Expert Rev. Vaccines 2013, 12, 345–347. [Google Scholar] [CrossRef] [Green Version]
  20. Wang, S.; Kong, Q.; Curtiss, R. New technologies in developing recombinant attenuated Salmonella vaccine vectors. Microb. Pathog. 2013, 58, 17–28. [Google Scholar] [CrossRef] [Green Version]
  21. Shams, H.; Poblete, F.; Rüssmann, H.; Galán, J.E.; Donis, R.O. Induction of specific CD8+ memory T cells and long lasting protection following immunization with Salmonella typhimurium expressing a lymphocytic choriomeningitis MHC class I-restricted epitope. Vaccine 2001, 20, 577–585. [Google Scholar] [CrossRef]
  22. Clements, J.D.; Kohler James, J.; Pathangey, L.; Hasona, A.; Progulske-Fox, A.; Brown Thomas, A. Long-Term Immunological Memory Induced by Recombinant Oral Salmonella Vaccine Vectors. Infect. Immun. 2000, 68, 4370–4373. [Google Scholar]
  23. Broz, P.; Ohlson, M.B.; Monack, D.M. Innate immune response to Salmonella typhimurium, a model enteric pathogen. Gut Microbes 2012, 3, 62–70. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Nakayama, K.; Kelly, S.M.; Curtiss, R. Construction of an ASD+ Expression-Cloning Vector: Stable Maintenance and High Level Expression of Cloned Genes in a Salmonella Vaccine Strain. Nat. Biotechnol. 1988, 6, 693–697. [Google Scholar] [CrossRef]
  25. Jawale Chetan, V.; Lee John, H.; Waters, W.R. Salmonella enterica Serovar Enteritidis Ghosts Carrying the Escherichia coli Heat-Labile Enterotoxin B Subunit Are Capable of Inducing Enhanced Protective Immune Responses. Clin. Vaccine Immunol. 2014, 21, 799–807. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Qian, F.; Guo, A.; Li, M.; Liu, W.; Pan, Z.; Jiang, L.; Wu, X.; Xu, H. Salmonella flagellin is a potent carrier-adjuvant for peptide conjugate to induce peptide-specific antibody response in mice. Vaccine 2015, 33, 2038–2044. [Google Scholar] [CrossRef]
  27. Pérez-Toledo, M.; Valero-Pacheco, N.; Pastelin-Palacios, R.; Gil-Cruz, C.; Perez-Shibayama, C.; Moreno-Eutimio, M.A.; Becker, I.; Pérez-Tapia, S.M.; Arriaga-Pizano, L.; Cunningham, A.F.; et al. Salmonella Typhi Porins OmpC and OmpF Are Potent Adjuvants for T-Dependent and T-Independent Antigens. Front. Immunol. 2017, 8, 230. [Google Scholar] [CrossRef]
  28. Wei, K.; Soo-Young, W.; Xin, Z.; Wendy, B.; Tinge, S.A.; Roland, K.L.; Roy, C. Regulated programmed lysis of recombinant Salmonella in host tissues to release protective antigens and confer biological containment. Proc. Natl. Acad. Sci. USA 2008, 105, 9361–9366. [Google Scholar]
  29. Jiang, Y.L.; Mo, H.; Willingham, C.; Wang, S.F.; Park, J.Y.; Kong, W.; Roland, K.L.; Curtiss, R. Protection against Necrotic Enteritis in Broiler Chickens by Regulated Delayed Lysis Salmonella Vaccines. Avian Dis. 2015, 59, 475–485. [Google Scholar] [CrossRef]
  30. Ameiss, K.; Ashraf, S.; Kong, W.; Pekosz, A.; Wu, W.H.; Milich, D.; Billaud, J.N.; Curtiss, R. Delivery of woodchuck hepatitis virus-like particle presented influenza M2e by recombinant attenuated Salmonella displaying a delayed lysis phenotype. Vaccine 2010, 28, 6704–6713. [Google Scholar] [CrossRef] [Green Version]
  31. Kulkarni, R.R.; Parreira, V.R.; Jiang, Y.F.; Prescott, J.F. A Live Oral Recombinant Salmonella enterica Serovar Typhimurium Vaccine Expressing Clostridium perfringens Antigens Confers Protection against Necrotic Enteritis in Broiler Chickens. Clin. Vaccine Immunol. 2010, 17, 205–214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Xin, W.; Wanda, S.Y.; Li, Y.; Wang, S.; Mo, H.; Curtiss, R., 3rd. Analysis of type II secretion of recombinant pneumococcal PspA and PspC in a Salmonella enterica serovar Typhimurium vaccine with regulated delayed antigen synthesis. Infect. Immun. 2008, 76, 3241–3254. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Lin, L.; Ibrahim, A.S.; Xu, X.; Farber, J.M.; Avanesian, V.; Baquir, B.; Fu, Y.; French, S.W.; Edwards, J.E., Jr.; Spellberg, B. Th1-Th17 cells mediate protective adaptive immunity against Staphylococcus aureus and Candida albicans infection in mice. PLoS Pathog. 2009, 5, e1000703. [Google Scholar] [CrossRef] [PubMed]
  34. Garzoni, C.; Kelley, W.L. Staphylococcus aureus: New evidence for intracellular persistence. Trends Microbiol. 2009, 17, 59–65. [Google Scholar] [CrossRef]
  35. Liu, Q.; Su, H.; Bian, X.; Wang, S.; Kong, Q. Live attenuated Salmonella Typhimurium with monophosphoryl lipid A retains ability to induce T-cell and humoral immune responses against heterologous polysaccharide of Shigella flexneri 2a. Int. J. Med. Microbiol. 2020, 310, 151427. [Google Scholar] [CrossRef]
  36. Woo, P.; Wong, L.P.; Zheng, B.J.; Yuen, K.Y. Unique immunogenicity of hepatitis B virus DNA vaccine presented by live-attenuated Salmonella typhimurium. Vaccine 2001, 19, 2945–2954. [Google Scholar] [CrossRef]
  37. Xu, D.; Mcsorley, S.J.; Chatfield, S.N.; Dougan, G.; Liew, F.Y. Protection against Leishmania major infection in genetically susceptible BALB/c mice by gp63 delivered orally in attenuated Salmonella typhimurium (AroA-AroD-). Immunology 1995, 85, 1–7. [Google Scholar]
  38. Zhao, N.; Lv, J.; Lu, Y.; Jiang, Y.; Zhao, X. Prolonging and enhancing the protective efficacy of the EtMIC3-C-MAR against eimeria tenella through delivered by attenuated Salmonella typhimurium. Vet. Parasitol. 2020, 279, 109061. [Google Scholar] [CrossRef]
  39. Hernández-Luna, M.; Luria-Pérez, R. Cancer Immunotherapy: Priming the Host Immune Response with Live Attenuated Salmonella enterica. J. Immunol. Res. 2018, 2018, 2984247. [Google Scholar] [CrossRef] [Green Version]
  40. Zhang, F.; Ledue, O.; Jun, M.; Goulart, C.; Malley, R.; Lu, Y.J. Protection against Staphylococcus aureus Colonization and Infection by B- and T-Cell-Mediated Mechanisms. MBio 2018, 9, e01949. [Google Scholar] [CrossRef] [Green Version]
  41. Van Kessel, K.P.M.; Bestebroer, J.; Van Strijp, J.A.G. Neutrophil-Mediated Phagocytosis of Staphylococcus aureus. Front. Immunol. 2014, 5, 467. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Kim, M.R.; Hong, S.W.; Choi, E.B.; Lee, W.H.; Kim, Y.S.; Jeon, S.G.; Jang, M.H.; Gho, Y.S.; Kim, Y.K. Staphylococcus aureus-derived extracellular vesicles induce neutrophilic pulmonary inflammation via both Th1 and Th17 cell responses. Allergy 2012, 67, 1271–1281. [Google Scholar] [CrossRef] [PubMed]
  43. Lowy, F.D. Is Staphylococcus aureus an intracellular pathogen? Trends Microbiol. 2000, 8, 341–343. [Google Scholar] [CrossRef]
  44. Zautner, A.E.; Merit, K.; Gerhard, S.; Silva, H.; Hagen, F.; Claudia, M.; Bernd, K.; Wilhelm, P.H.; Andreas, P.; Stefan, B. Intracellular Persisting Staphylococcus aureus Is the Major Pathogen in Recurrent Tonsillitis. PLoS ONE 2010, 5, e9452. [Google Scholar] [CrossRef] [Green Version]
  45. Kubica, M.; Guzik, K.; Koziel, J.; Zarebski, M.; Richter, W.; Gajkowska, B.; Golda, A.; Maciag-Gudowska, A.; Brix, K.; Shaw, L. A potential new pathway for Staphylococcus aureus dissemination: The silent survival of S. aureus phagocytosed by human monocyte-derived macrophages. PLoS ONE 2008, 3, e1409. [Google Scholar] [CrossRef] [Green Version]
  46. Barbara, B.; Daniel, M.; Vincent, P. The T Cell Response to Staphylococcus aureus. Pathogens 2016, 5, 31. [Google Scholar]
  47. Reiner, S.L.; Locksley, R.M. The regulation of immunity to Leishmania major. Annu. Rev. Immunol. 1995, 13, 151–177. [Google Scholar] [CrossRef]
  48. Chen, K.; Jeremy, L.Y.; David, Z.M.; Casey, J.J. Th17 Cells Mediate Clade-Specific, Serotype-Independent Mucosal Immunity. Immunity 2011, 35, 997–1009. [Google Scholar] [CrossRef] [Green Version]
  49. Islander, U.; Andersson, A.; Lindberg, E.; Adlerberth, I.; Rudin, A. Superantigenic Staphylococcus aureus Stimulates Production of Interleukin-17 from Memory but Not Naive T Cells. Infect. Immun. 2010, 78, 381–386. [Google Scholar] [CrossRef] [Green Version]
  50. Xu, C.; Zhang, B.Z.; Lin, Q.B.; Deng, J.; Yu, B.; Arya, S.; Yuen, K.Y.; Huang, J.D. Live attenuated Salmonella typhimurium vaccines delivering SaEsxA and SaEsxB via type III secretion system confer protection against Staphylococcus aureus infection. BMC Infect. Dis. 2018, 18, 195. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Construction of pYA3681−esxAB and pYA3681−hlam plasmids and protein expression in recombinant strains of χ11802. (A) Construction scheme of pYA3681−esxAB. (B) Construction scheme of pYA3681−hlam. (C) The recombinant pYA3681−esxAB plasmid was identified by restriction endonuclease digestion. 8K DNA marker (Lane M1), pYA3681−esxAB plasmid (Lane 1), pYA3681−esxAB plasmid double-enzyme digested with NcoI and SmaI (Lane 2), and 2K DNA marker (Lane M2). (D) The recombinant pYA3681−hlam plasmid was identified by restriction endonucleases. 8K DNA marker (Lane M1), pYA3681−hlam plasmid (Lane 1), pYA3681−hlam plasmid double-enzyme digested with NcoI and SmaI (Lane 2), and 2K DNA marker (Lane M2). (E) rEsxAB and rHlam synthesis in recombinant strains of χ11802 and the recombinant proteins released in the supernatant culture medium were analyzed by Western blot. (F) Intensity ratio of EsxAB and Hlam expression in recombinant strains of χ11802.
Figure 1. Construction of pYA3681−esxAB and pYA3681−hlam plasmids and protein expression in recombinant strains of χ11802. (A) Construction scheme of pYA3681−esxAB. (B) Construction scheme of pYA3681−hlam. (C) The recombinant pYA3681−esxAB plasmid was identified by restriction endonuclease digestion. 8K DNA marker (Lane M1), pYA3681−esxAB plasmid (Lane 1), pYA3681−esxAB plasmid double-enzyme digested with NcoI and SmaI (Lane 2), and 2K DNA marker (Lane M2). (D) The recombinant pYA3681−hlam plasmid was identified by restriction endonucleases. 8K DNA marker (Lane M1), pYA3681−hlam plasmid (Lane 1), pYA3681−hlam plasmid double-enzyme digested with NcoI and SmaI (Lane 2), and 2K DNA marker (Lane M2). (E) rEsxAB and rHlam synthesis in recombinant strains of χ11802 and the recombinant proteins released in the supernatant culture medium were analyzed by Western blot. (F) Intensity ratio of EsxAB and Hlam expression in recombinant strains of χ11802.
Vaccines 10 01073 g001
Figure 2. Recombinant strains of χ11802 survival status in vitro and in vivo in the absence of arabinose. (A) χ11802(pYA3681−esxAB) and χ11802(pYA3681−hlam) survival status on LB plate with or without 0.02% w/v arabinose. (B) χ11802(pYA3681−esxAB) and χ11802(pYA3681−hlam) survival status in LB broth with or without 0.02% w/v arabinose. (C) Bacterial loading of χ11802(pYA3681−esxAB) and χ11802(pYA3681−hlam) in mouse intestines, stomachs, MLNs, spleens, and Peyer’s patches over time, after oral administration, with the Mean value of the bacterial loading presented as red dash.
Figure 2. Recombinant strains of χ11802 survival status in vitro and in vivo in the absence of arabinose. (A) χ11802(pYA3681−esxAB) and χ11802(pYA3681−hlam) survival status on LB plate with or without 0.02% w/v arabinose. (B) χ11802(pYA3681−esxAB) and χ11802(pYA3681−hlam) survival status in LB broth with or without 0.02% w/v arabinose. (C) Bacterial loading of χ11802(pYA3681−esxAB) and χ11802(pYA3681−hlam) in mouse intestines, stomachs, MLNs, spleens, and Peyer’s patches over time, after oral administration, with the Mean value of the bacterial loading presented as red dash.
Vaccines 10 01073 g002
Figure 3. Antigen-specific IgG and IgA immune responses after BALB/c mice were orally administrated with recombinant strains of χ11802. In (AE), mice were orally administrated with χ11802(pYA3681), χ11802(pYA3681−esxAB), or χ11802(pYA3681−hlam). (A,B) Serum anti-rEsxAB specific IgG level on day 14 after primary and booster immunization. (C,D) Serum anti-rHlam specific IgG level on day 14 after primary and booster immunization. (E,F) Intestinal mucus anti-rEsxAB and anti-rHlam specific IgA level on day 7 after booster immunization. *** p < 0.001.
Figure 3. Antigen-specific IgG and IgA immune responses after BALB/c mice were orally administrated with recombinant strains of χ11802. In (AE), mice were orally administrated with χ11802(pYA3681), χ11802(pYA3681−esxAB), or χ11802(pYA3681−hlam). (A,B) Serum anti-rEsxAB specific IgG level on day 14 after primary and booster immunization. (C,D) Serum anti-rHlam specific IgG level on day 14 after primary and booster immunization. (E,F) Intestinal mucus anti-rEsxAB and anti-rHlam specific IgA level on day 7 after booster immunization. *** p < 0.001.
Vaccines 10 01073 g003
Figure 4. IFN-γ and IL-17 production in mouse kidney tissue elicited by oral administration with the recombinant strains of χ11802. In (A,B), mice were orally administrated with PBS solution, χ11802(pYA3681), χ11802(pYA3681−esxAB), or χ11802(pYA3681−hlam). (A) IFN-γ level in mouse kidney tissue. (B) IL-17 level in mouse kidney tissue. * p < 0.05; ** p < 0.01; ns, no significance.
Figure 4. IFN-γ and IL-17 production in mouse kidney tissue elicited by oral administration with the recombinant strains of χ11802. In (A,B), mice were orally administrated with PBS solution, χ11802(pYA3681), χ11802(pYA3681−esxAB), or χ11802(pYA3681−hlam). (A) IFN-γ level in mouse kidney tissue. (B) IL-17 level in mouse kidney tissue. * p < 0.05; ** p < 0.01; ns, no significance.
Vaccines 10 01073 g004
Figure 5. Abscess formation after BALB/c mice were challenged with S. aureus USA300 by i.v. (A) Immunization and challenge strategy. (B) Abscess formation of BALB/c mouse kidneys. (C) Four different degrees of kidney abscess symptoms. (D) Abscess area proportion of total kidney surface area. (E) Colonization of S. aureus USA300 in the kidneys after BALB/c mice challenge. * p < 0.05; ** p < 0.01; ns, no significance.
Figure 5. Abscess formation after BALB/c mice were challenged with S. aureus USA300 by i.v. (A) Immunization and challenge strategy. (B) Abscess formation of BALB/c mouse kidneys. (C) Four different degrees of kidney abscess symptoms. (D) Abscess area proportion of total kidney surface area. (E) Colonization of S. aureus USA300 in the kidneys after BALB/c mice challenge. * p < 0.05; ** p < 0.01; ns, no significance.
Vaccines 10 01073 g005
Figure 6. Histopathological evaluation of kidney after BALB/c mice were challenged with S. aureus USA300. Pathological section stained with H&E. The 100× magnification with a 50 μm bar. The 200× magnification with a 100 μm bar.
Figure 6. Histopathological evaluation of kidney after BALB/c mice were challenged with S. aureus USA300. Pathological section stained with H&E. The 100× magnification with a 50 μm bar. The 200× magnification with a 100 μm bar.
Vaccines 10 01073 g006
Table 1. Bacterial strains and plasmids used in this study.
Table 1. Bacterial strains and plasmids used in this study.
Strain or PlasmidDescriptionSource or Reference
Strain
χ6212F λ φ80 Δ(lacZYA-argF) endA1 recA1 hsdR17 deoR thi-1 glnV44 gyrA96 relA1 ΔasdA4Genotype referenced Xin et al. [32]
χ11802ΔPmurA25::TT araC PBAD murA DasdA27::TT araC PBADc2 Δ(wza-wcaM)-8 Δpmi-2426 ΔrelA198::araC PBAD lacI TT ΔrecF126Genotype referenced Jiang et al. [29]
χ6212(pYA3681−esxAB)χ6212 carrying pYA3681−esxABThis study
χ6212(pYA3681−hlam)χ6212 carrying pYA3681−hlamThis study
χ11802(pYA3681)χ11802 carrying pYA3681This study
χ11802 (pYA3681−esxAB)χ11802 carrying pYA3681−esxABThis study
χ11802 (pYA3681−hlam)χ11802 carrying pYA3681−hlamThis study
S. aureus USA300 American Type Culture Collection (Manassas, VA, USA)
Plasmid
pUC57−esxABpUC57::blaSS::HA-tag:: esxABGenScript Bio Co.
pUC57−hlampUC57::blaSS::HA-tag:: hlamGenScript Bio Co.
pYA3681Ptrc pBR ori araC* PBAD SD-GTG-asdA SD-GTG-murA P22 PR antisense mRNAGenotype referenced Jiang et al. [29]
pYA3681−esxABpYA3681::blaSS::HA-tag:: esxABThis study
pYA3681−hlampYA3681::blaSS::HA-tag:: hlamThis study
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Liang, Y.; Zhang, H.; Pan, N.; Liu, Y.; Sheng, S.; Li, H.; Bao, X.; Wang, X. Oral Administration with Recombinant Attenuated Regulated Delayed Lysis Salmonella Vaccines Protecting against Staphylococcus aureus Kidney Abscess Formation. Vaccines 2022, 10, 1073. https://doi.org/10.3390/vaccines10071073

AMA Style

Liang Y, Zhang H, Pan N, Liu Y, Sheng S, Li H, Bao X, Wang X. Oral Administration with Recombinant Attenuated Regulated Delayed Lysis Salmonella Vaccines Protecting against Staphylococcus aureus Kidney Abscess Formation. Vaccines. 2022; 10(7):1073. https://doi.org/10.3390/vaccines10071073

Chicago/Turabian Style

Liang, Yanchen, Haochi Zhang, Na Pan, Yang Liu, Shouxin Sheng, Haotian Li, Xuemei Bao, and Xiao Wang. 2022. "Oral Administration with Recombinant Attenuated Regulated Delayed Lysis Salmonella Vaccines Protecting against Staphylococcus aureus Kidney Abscess Formation" Vaccines 10, no. 7: 1073. https://doi.org/10.3390/vaccines10071073

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