Skip to main content

Recombinant-attenuated Salmonella enterica serovar Choleraesuis vector expressing the PlpE protein of Pasteurella multocida protects mice from lethal challenge

Abstract

Background

Bacterial surface proteins play key roles in pathogenicity and often contribute to microbial adhesion and invasion. Pasteurella lipoprotein E (PlpE), a Pasteurella multocida (P. multocida) surface protein, has recently been identified as a potential vaccine candidate. Live attenuated Salmonella strains have a number of potential advantages as vaccine vectors, including immunization with live vector can mimic natural infections by organisms, lead to the induction of mucosal, humoral, and cellular immune responses. In this study, a previously constructed recombinant attenuated Salmonella Choleraesuis (S. Choleraesuis) vector rSC0016 was used to synthesize and secrete the surface protein PlpE of P. multocida to form the vaccine candidate rSC0016(pS-PlpE). Subsequently, the immunogenicity of S. Choleraesuis rSC0016(pS-PlpE) as an oral vaccine to induce protective immunity against P. multocida in mice was evaluated.

Results

After immunization, the recombinant attenuated S. Choleraesuis vector can efficiently delivered P. multocida PlpE protein in vivo and induced a specific immune response against this heterologous antigen in mice. In addition, compared with the inactivated vaccine, empty vector (rSC0016(pYA3493)) and PBS immunized groups, the rSC0016(pS-PlpE) vaccine candidate group induced higher antigen-specific mucosal, humoral and mixed Th1/Th2 cellular immune responses. After intraperitoneal challenge, the rSC0016(pS-PlpE) immunized group had a markedly enhanced survival rate (80%), a better protection efficiency than 60% of the inactivated vaccine group, and significantly reduced tissue damage.

Conclusions

In conclusion, our study found that the rSC0016(pS-PlpE) vaccine candidate provided good protection against challenge with wild-type P. multocida serotype A in a mouse infection model, and may potentially be considered for use as a universal vaccine against multiple serotypes of P. multocida in livestock, including pigs.

Peer Review reports

Background

Pasteurella multocida (P. multocida) is an important opportunistic bacteria that causes a wide range of infections including fowl cholera, bovine haemorrhagic septicaemia, rabbit pasteurellosis, swine atrophic rhinitis [1]. At the same time, it is also an important zoonotic pathogen that can cause serious abscess formation, meningitis, and leading to significantly harmful impacts on human health [1, 2]. Based on its capsular composition, the P. multocida is divided into A, B, D, E and F capsular serogroups and further classified into 16 serotypes (1–16) based on distinct lipopolysaccharide structures [3, 4]. Progressive atrophic rhinitis (PAR) and pneumonic pasteurellosis (PN) in pigs are the main diseases caused by toxigenic and nontoxigenic strains of types A and D P. multocida [1, 5]. As an important pathogen of pigs, P. multocida can coinfect with other pathogens including porcine reproductive and respiratory syndrome, porcine circovirus and Mycoplasma hyopneumoniae [6, 7]. This coinfection may cause immunosuppression to reduce the host’s immunity and resistance to other diseases.

Antibiotics therapy is usually used for the treatment of P. multocida infection [1, 8]. However, under intensive feeding conditions, the extensive use and misuse of antibiotics has led to the increasing resistance of P. multocida, which eventually has led to treatment failure [8]. Previous outcome studies also indicate that antibiotic resistant bacteria may also increases the potential risk of gene transfer to non-resistant bacteria and therefore may pose a risk to human health [9]. Vaccination is currently the most effective measure to prevent of P. multocida infection and disease [10]. Conventional inactivated P. multocida vaccine has been widely used and has played a crucial role in control and prevention of diseases caused by P. multocida globally [11]. However, traditional inactivated vaccines have many shortcomings, including thermal instability, short duration of immunization, weakly immunogenic, poor cross-protection, and inflammation at the injection site [10, 11]. The application of live attenuated vaccines, although showed a better protective efficacy, carries the risk of systemic infections after immunization as a result of potential reversion to virulence [10]. At present, inactivated vaccine (C44-1 strain) and live attenuated vaccine (EO630 strain) are two traditional vaccines commonly used in China. Immune responses and the protection in a mouse model were evaluated and found that both live attenuated vaccines (71%) and inactivated vaccines (51%) could not provide complete protection against homologous serotypes and did not protect against heterologous serotypes [11]. To address the above research questions, researchers have evaluated new types of vaccine such as subunit vaccines and DNA vaccines [12,13,14,15]. However, the correct folding, post-translational modification of proteins and harsh storage conditions still limit the development of related vaccines.

P. multocida exhibits its pathogenicity via numerous virulence factors, including adhesins, toxins, and outer membrane proteins [2, 4]. At the same time, these proteins are an ideal target antigen for a P. multocida protective vaccine. The PlpE protein is a surface protein and is considered as one of the major antigens among P. multocida for vaccine development [16]. After immunization, recombinant protein PlpE provided 80–100% protection in mice and induced cross-serotype protective immunity [13, 14, 17]. It also protected chickens against fowl cholera challenge [17]. Importantly, this protein was present in all P. multocida serotypes with the sequence identity over 90% [17]. These findings suggested that PlpE is an ideal target antigen for the development of anti-P. multocida vaccines. Of note, the recombinant protein-based vaccines often require different types of adjuvants to elicit a high levels of protective immune [18]. Live attenuated Salmonella, as an immune modulator and vaccine adjuvant, induces strong mucosal antibody and cell-mediated immune responses, has been successfully developed and used as a vaccine vector, and is an attractive platform for delivering various heterologous antigens to the immune system [19]. Over the years, achieving a balance between attenuation and immunogenicity has proved remarkably difficult: some live attenuated Salmonella vaccine vectors have been insufficiently attenuated, whereas others were over-attenuated but insufficiently immunogenic [20]. In order to address these questions, Roy Curtiss and colleagues have described a variety of novel strategies for the construction of attenuated vector, including regulated delayed attenuation, regulated delayed antigen synthesis, and regulated delayed lysis (for balance between safety and immunogenicity) [21,22,23]. At the same time, by constructing a balanced lethal system using complementation of the asd (aspartate semialdehyde dehydrogenase) gene in trans, to ensure the stability of the plasmid and the synthesis of heterologous antigens [22]. The Salmonella vector modified by the above system has been verified to deliver antigen proteins of different pathogens and to strengthen the antigen-specific antibody and T-cell immune responses, which can provide the host with more effective and durable immune protection [24,25,26,27]. These studies fully demonstrate that the attenuated Salmonella vector vaccine has great potential as a novel vaccine for protection against heterologous pathogens.

In previous research, our laboratory constructed a recombinant attenuated S. Choleraesuis vector containing the regulated delayed attenuation system and regulated delayed exogenous synthesis system, which we have named rSC0016 [27]. At the same time, its sopB gene was also knocked out to reduce the host intestinal inflammatory response induced by the Salmonella vector itself [26]. In this study, we constructed a recombinant plasmid based on the recombinant attenuated S. Choleraesuis vector expressing heterologous antigen PlpE, and assessed its immunogenicity and protective effect in mice. The results serve as a solid foundation for further development of an efficient and inexpensive universal vaccine against P. multocida disease for livestock, including pigs.

Results

Identification of P. multocida serotype A and LD50 results

Amplification with species-specific PCR successfully confirmed the isolate as P. multocida by amplification of DNA fragments (KMT; ~ 460 bp) (Fig. 1A). P. multocida capsular typing proven by the presence of the hyaD-hyaC gene of serotypes-A specific and amplified product, showed a fragment size of ~ 1044 bp (Fig. 1B). These results demonstrated that the isolate from diseased pigs was confirmed as P. multocida serotype A. Both primers failed to amplify the expected size bands in Salmonella and E. coli, indicating the specificity of the primers. To determine whether the P. multocida field strain was lethal to BALB/c mice and to measure the LD50 of the strain, six-week-old mice were intraperitoneally (i.p.) injected with serial dilutions of inocula of the bacterial strain. The test results indicated the LD50 of P. multocida serotype A field strain in BALB/c mice was 1.6 × 102 CFU by i.p. administration (Fig. 1C). Our results showed that the P. multocida serotype A field strain was classified as highly virulent.

Fig. 1
figure 1

Identification results of strain species and serotypes. (A) P. multocida species-specific polymerase chain reaction for the detection of KMT gene (~ 460 bp). M: DL2000 DNA marker; lane 1: DNA of P. multocida serotype A field strain; lane 2: no DNA (negative control); lane 3: C78-3 DNA (S. Choleraesuis wild-type control); lane 4: DH5α DNA (E. coli control). (B) Amplification of hyaD-hyaC gene (~ 1044 bp) specific to P. multocida serotype A. M: DL2000 DNA marker; lane 1: DNA of P. multocida field strain; lane 2: no DNA (negative control); lane 3: C78-3 DNA (S. Choleraesuis wild-type control); lane 4: DH5α DNA (E. coli control). (C) Survival rate of mice (n = 5) challenged with different doses of P. multocida serotype A

Construction of rSC0016(pS-PlpE) and preparation of the inactivated P. multocida serotype A vaccine and phenotypic analyses

The maps of the plasmids employed in the present study are presented in Fig. 2A. Successful construction of the plasmid pS-PlpE was confirmed by PCR amplification, EcoR I and Sal I digestion (Fig. 2B). The plasmid pS-PlpE that was constructed was further verified by DNA sequencing (data not shown), and the protein expression in the candidate vaccine strain was verified using polyclonal antiserum prepared as described in Methods. The expression of PlpE in the attenuated S. Choleraesuis vector is the basis for inducing protective immunity against P. multocida infections using the mouse infection model.

Fig. 2
figure 2

Plasmid maps and construction results, phenotypic characteristics of vaccine candidate strains. (A) Plasmid maps of pYA3493 and pS-PlpE. (B) Identification of recombinant plasmid pS-PlpE by PCR and restriction enzyme digestion. M: DL2000 or DL5000 DNA marker; Lane 1, 2, 3: EcoR I and Sal I digested pS-PlpE (different clones). (C) The expression of PlpE in rSC0016 was analyzed by WB. Lane 1: purified PlpE protein; lane 2: BL21(pET-28a). (D) Plasmid stability of pS-PlpE during passage. M: DL5000 DNA marker; Lane 1: 10 passages; lane 2: 20 passages; lane 3: 30 passages; lane 4: 40 passages; lane 5: 50 passages. (E) Growth curves of the rSC0016(pS-PlpE), rSC0016(pYA3493), C78-3 strains in the LB medium

As shown in Fig. 2C, a band of 40 kDa of the expected size of the recombinant protein could be detected in rSC0016(pS-PlpE), but not in rSC0016(pYA3493), the empty vector strain. To determine the stability of our plasmids upon serial passage over time, individual transformants were picked into LB medium and transferred into fresh medium every 12 h for 50 passages. At passages 10, 20, 30, 40 and 50, the plasmids were extracted and identified by double enzyme digestion (Fig. 2D). Our results demonstrated that the use of Asd as a plasmid retention system (balanced lethal system) was adequate to enable plasmid retention in the Asd− strain, rSC0016. The growth curve of the rSC0016(pS-PlpE) strain was also determined parallel to the rSC0016(pYA3493) strain and the wild-type S. Choleraesuis strain, C78-3. As revealed in Fig. 2E, the growth rate of wild-type strain C78-3 was significantly higher between 6 and 8 h when compared to other strains. It appears that rSC0016(pS-PlpE) grew somewhat slower than the rSC0016(pYA3493) between 6 and 8 h. However, the growth capacity was significantly higher in the rSC0016(pYA3493) compared to the rSC0016(pS-PlpE) strain at 10 h, the last time point. This indicates that foreign antigen expression may influence the growth capacity of the rSC0016 vector. After the inactivated vaccine was prepared from the wild type P. multocida serotype A strain, it was coated onto TSA solid medium containing 5% fetal bovine serum, and the results showed that no colonies grew (data not shown). The lack of colony growth on the solid media confirmed that the inactivation was sufficient, and the P. multocida inactivated vaccine was successfully prepared.

High levels of antigen-specific antibodies were elicited by immunization with rSC0016(pS-PlpE)

Specific antibody responses elicited in different immune groups were measured by titrating the serum and vaginal lavages of vaccinated and control mice against the PlpE protein. Compared with the PBS group or the rSC0016(pYA3493) empty vector group, mice orally immunized with rSC0016(pS-PlpE) and inoculated subcutaneously with the inactivated vaccine induced higher serum IgG titers against PlpE (Fig. 3A). Also, these levels were significantly higher in serum of mice after the booster immunization compared to those immunized with PBS or empty vector group. However, there was no significant difference between the rSC0016(pS-PlpE) and inactivated vaccine immunized groups. sIgA antibodies plays a critical role in mucosal immunity against P. multocida infection, so levels of sIgA were analyzed in vaginal lavage fluid collected from mice. As shown in Fig. 3B, both attenuated rSC0016 (pS-PlpE) and inactivated vaccine immunizations induced significantly higher levels of sIgA titers in mice than in the PBS and empty vector control groups. Although the mucosal immune response induced by rSC0016(pS-PlpE) strain was marginally higher than that of the inactivated vaccine immunization group, there was no significant difference. These data suggest that the rSC0016 (pS-PlpE) strain elicited significant specific humoral and mucosal immune responses, and the levels were comparable to those of the inactivated vaccines.

Fig. 3
figure 3

Detection of antibody titer in the immunized mice (n = 30). (A) PlpE-specific IgG antibody titer in serum determined by iELISA. (B) IgA titers detected by iELISA in vaginal lavage fluids. Specific anti-PlpE serum IgG1 (C) and IgG2a (D) titers. The results are expressed as the mean ± SD. Degrees of significance are indicated as follows: *P < 0.05; **P < 0.01; n.s., nonsignificant (P ≥ 0.05)

In mice, IgG2a is associated with the Th1-like immune response, while IgG1 is related to the Th2-like immune response. To further assess the types of immune responses in immunized mice, we examined IgG1 and IgG2a antibody subclass levels in the different groups (Fig. 3C, D). The results showed that mice immunized with rSC0016(pS-PlpE) and the inactivated vaccine induced increased levels of IgG1 and IgG2a, which were significantly higher than those immunized with PBS and empty vector. The inactivated vaccine induced higher, but not significant, IgG1 levels than rSC0016(pS-PlpE), while rSC0016(pS-PlpE) induced significantly higher IgG2a levels compared to the inactivated vaccinated group. The above results indicate that significant Th1- and Th2-type immune responses were induced in both the rSC0016 (pS-PlpE) and inactivated vaccine groups compared to the PBS and empty vector control groups.

rSC0016(pS-PlpE) induced Th-1 and Th-2 responses

Assessment of cell-mediated immune responses was done through measuring the IFN-γ and IL-4 cytokines by ELISA in culture supernatants fluid of splenic lymphocytes stimulated with PlpE (Fig. 4). The results revealed that the levels of IFN-γ and IL-4 in both the rSC0016(pS-PlpE) group and inactivated vaccine group were significantly higher compared with PBS and empty vector controls. These results show that both rSC0016(pS-PlpE) and inactivated vaccine immunization groups induced robust Th1 and Th2 type immune responses. A highly significant Th-1 response was observed, evident by the higher IFN-γ levels induced, in the rSC0016(pS-PlpE) group compared to the inactivated vaccine group’s response, while the inactivated vaccine group responded with a significant Th-2 response, shown by the higher IL-4 concentration, compared to the rSC0016(pS-PlpE) group, which would be expected for a parenterally administered antigen.

Fig. 4
figure 4

Levels of secreted IL-4 and IFN-γ were assayed by ELISA (n = 15). Splenic lymphocytes were used to evaluate cytokine secretion in vitro following re-stimulation with purified PlpE protein. The results are expressed as the mean ± SD. Degrees of significance are indicated as follows: *P < 0.05; **P < 0.01; n.s., nonsignificant (P ≥ 0.05)

Oral immunization with rSC0016(pS-PlpE) vaccine candidate strain increased survival and reduced pathological lung lesions in mice

Mice of each immunized group were intraperitoneally challenged with the isolated P. multocida serotype A three weeks after the second booster vaccination to verify the protective effect of the rSC0016(pS-PlpE) vaccine candidate strain. As shown in Fig. 5A, the results indicated that the survival rate was 80% in the rSC0016(pS-PlpE) group and 60% in the inactivated vaccine group. All mice in the control (empty vector-vaccinated and PBS-treated) groups died within 48 h after challenge. Pulmonary histopathologic changes upon P. multocida infection were assessed to further characterize the protective efficacy of the different vaccines compared to the PBS and empty vector control groups (Fig. 5B). Histological analysis of the lungs revealed that those in the rSC0016(pS-PlpE) and inactivated vaccine groups were similar to the non-infected blank control, with no obvious pathologic changes observed. In addition, the pathology scores for both the rSC0016(pS-PlpE) and inactivated vaccine groups were not significantly different from that of the non-infected blank control group (Fig. 5C). In contrast, compared with the blank control group, the lungs of mice in the PBS and empty vector groups challenged with the wild-type P. multocida strain displayed severe tissue injury, which mainly presented as mild inflammatory cell infiltration (red arrow), hyperemia (yellow arrow), and thickening of alveolar wall (black arrow) (Fig. 5B). Consistent with these histopathological changes, the lung lesion scores in the PBS and empty vector groups were significantly higher compared to those of the blank control group, rSC0016(pS-PlpE) group and inactivated vaccine group (Fig. 5C). These results suggest that rSC0016(pS-PlpE) vaccine may have protected mice against an i.p. challenge with wild-type P. multocida by preventing severe injury to the lungs.

Fig. 5
figure 5

Protective efficacy of developed vaccine. (A) Survival rates of mice after P. multocida challenge were determined (n = 30). (B) Histological changes of lung tissue in mice (n = 15). HE staining revealed that mild inflammatory cell infiltration (red arrow), hyperemia (yellow arrow), and thickening of alveolar wall (black arrow) were observed in the lung tissues of PBS and rSC0016(pYA3493) immunized mice, compared with the blank control group. As expected, there were no obvious differences in the pathological changes among the rSC0016(pS-PlpE), the inactivated vaccine, and the blank control groups. (C) Histological scores of mice lungs in each group

Discussion

P. multocida is an important zoonotic pathogen that has caused severe economic losses in the global swine industry and endangered public health around the world [28]. As with other opportunistic pathogens, P. multocida has the potential to proliferate and infect the lungs when host immunity is compromised due to stress or other pathogens infections [6, 7]. Because P. multocida can infect the host through mucosal tissues, the induction of protective immunity at mucosal tissues plays an important role in defense against pathogens [29]. At the same time, both cellular and humoral immune responses also play important roles in the host’s defense against P. multocida infections, which may be helpful to reduce the lungs damage and prompt their recovery [30]. In brief, the simultaneous induction of strong mucosal, humoral and cellular immune responses by vaccination against P. multocida colonization and infection is the goal for the development of a new vaccine.

Recently, numerous studies have shown that several P. multocida outer membrane proteins (OMPs) contribute to the pathogenicity and possess immunogenic and bactericidal properties [31, 32]. PlpE is an antigenic, surface-exposed and conserved OMP that is detected among the diverse serotypes of P. multocida investigated and has been mooted as a potential candidate for vaccine development [16, 33]. The use of attenuated Salmonella as a vector carrying heterologous antigen represents a novel strategy to preventing and controlling infectious diseases of humans and animals [24,25,26,27, 34]. In this study, a recombinant-attenuated S. Choleraesuis strain carrying the plpE gene inserted into the pYA3943 plasmid was constructed, and the expression and secretion of the PlpE protein from the rSC0016 strain carrying the plpE gene in the pYA3943 plasmid was demonstrated by WB analysis. The pYA3943 prokaryotic plasmid contains the pBR ori and asd genes, which produce a balanced lethal system to complement the chromosomal asd mutation strain. Our results show that the pS-PlpE plasmid can be stably maintained for over 50 passages in the rSC0016 vaccine candidate strain. These results to construct and characterize rSC0016(pS-PlpE) are consistent with results from previously reported literature [24, 27].

Besides expressing the target protein, the ability of the candidate vaccine strain to induce the antigen-specific antibody is equally important. In this study, mice orally inoculated with rSC0016(pS-PlpE) and administered the inactivated vaccine subcutaneously induced significantly higher levels of PlpE-specific IgG antibodies, while mice that received the empty vector, or PBS orally had no detectable specific antibodies. It is now widely accepted that clearance of pathogens is mediated by IgG that promote uptake and killing by professional phagocytes, mainly activated macrophages [35]. However, previous studies reported that the capsule of P. multocida is an important physical barrier that protects the bacteria to escape from the killing system of macrophages [36, 37]. Additionally, a previous study demonstrated that IgG levels against the PlpE protein are not indicative of protection, so the production of other antibody types should be further evaluated [13]. sIgA is the predominant immunoglobulin secreted at the mucosal surfaces and is the central aim of mucosal immune response [38]. Several studies have shown that sIgA can bind to pathogens and prevent them from attaching to or penetrating superficial mucosa layer, such as Streptococcus pneumoniae, Neisseria meningitidis, and Bordetella pertussis [39,40,41]. Moreover, enhanced mucosal immunity also resulted in higher protection against P. multocida [29, 42]. Our results revealed that after immunization, the level of sIgA measured from the rSC0016(pS-PlpE) group was slightly higher than levels in mice subcutaneously provided the inactivated vaccine, and was significantly higher than that of the empty vector or PBS groups. The increased level of sIgA in the rSC0016(pS-PlpE) group may be due to the method of immunizing by the oral route. Accumulating evidences suggests that oral vaccines can induce a more robust mucosal immunity than parenteral immunization [19]. However, the present study showed that the level of IgG, IgA, and IgG1 antibodies produced after subcutaneous immunization of mice with inactivated vaccine was comparable to the rSC0016(pS-PlpE) group, and the IgG2a level was significantly higher in the rSC0016(pS-PlpE) group at 5-weeks post oral vaccination. The significantly increased level of IgG2a for the rSC0016(pS-PlpE) group may be associated with the fact that the PlpE protein expressed by the recombinant vaccine is a very potent surface-exposed and secreted lipoprotein making it readily available to the host immune system. Previous studies have shown that lipoproteins serve as good mucosal adjuvants, stimulating high levels of IgG and secretory IgA production without additional adjuvants [43,44,45,46].

Cellular immune responses also play an important role in mitigating P. multocida infections [30]. IgG2a and IgG1 are used as indicators for Th1 and Th2 responses, respectively, thus the vaccine-induced T-cell phenotype can be evaluated by the levels of both IgG1 and IgG2a isotypes [47]. We observed the production of IgG1 and IgG2a isotypes in both rSC0016 (pS-PlpE) and inactivated vaccine immunized groups. These results suggest that both vaccines induced the Th1- and Th2-type immune responses against the PlpE antigen. Studies show that the generation of immune types may be related to the vaccination route and the formulation of the vaccine (live vs. inactivated) [48]. In previous studies, recombinant attenuated Salmonella vectors were more likely to induce Th1 immune types [24, 27]. IFN-γ and IL-2 are generally related to Th1-type cellular immune responses and can induce IgG2a production [49, 50]. IL-4 levels are generally related to Th2 type cellular immune responses and can stimulate IgG1 production [51, 52]. Similarly, both the rSC0016 (pS-PlpE) and inactivated vaccine immunization groups induced high levels of IFN-γ and IL-4 production. These results show that the rSC0016(pS-PlpE) recombinant vaccine induced potent, mixed Th1/Th2 cellular immune responses. In a previous study, the immunogenicity and protective effects of a P. multocida ghost vaccine was investigated, and the results showed that the ghost vaccine induced a higher level of cellular immune response compared to inactivated vaccines, which, in turn, protected mice against P. multocida infection, decreased tissue lesions and lowered bacterial loads [53]. Therefore, high levels of cellular immune response may also contribute to protection against P. multocida infection.

To assess whether the immune response induced by the vaccine candidate strain rSC0016 (pS-PlpE) was protective, mice were challenged with wild-type P. multocida by intraperitoneal injection three weeks post booster vaccination. Our data demonstrated that immunization with the rSC0016(pS-PlpE) vaccine twice provided 80% protection, compared with 60% protection provided by the inactivated vaccine. This is consistent with a previous study that parenteral inactivated vaccines only provide partial protection compared to live attenuated vaccines against P. multocida infection in mice [11]. In addition, compared with the inactivated vaccine group, the rSC0016(pS-PlpE) vaccine candidate strain also delayed the time to death in mice. This agrees with data from other studies, suggesting that formalin can destroy subtle antigenic structure on the outer-membrane of P. multocida, which leads to a lower immunogenic response [10, 18]. We further investigated pulmonary histopathology upon P. multocida infection for assessing the efficacy of rSC0016(pS-PlpE) to reduce lung tissue damage. The results showed that mice in the rSC0016(pS-PlpE) vaccine and the inactivated vaccine immunization groups had significantly reduced lung lesions and lung inflammation compared to the empty vector- and PBS-inoculated groups. These results were further validated by an evaluation of the pathology scores. In contrast to the blank control group, no significant changes of alveolar structural damage were observed in both the rSC0016(pS-PlpE) and inactivated vaccine groups. However, inflammatory cell infiltration, hyperemia, and alveolar wall thickening were observed in the PBS and empty vehicle immunization groups. Histopathological results showed that the rSC0016(pS-PlpE) vaccine candidate immunized group significantly reduced the pathological damage to the lungs of mice after challenge, which may be related to the high survival rate after P. multocida challenge.

Conclusions

In summary, we constructed a novel live vector vaccine strain of S. Choleraesuis expressing PlpE of P. multocida and explored its potential application as a live vaccine in a mouse infection model. Oral inoculation of rSC0016(pS-PlpE) induced good induction of humoral immune responses, mixed Th1 and Th2 immune responses, and most importantly, mucosal immune responses in mice vaccinated twice with the live recombinant vaccine. Furthermore, immunized mice in the rSC0016(pS-PlpE) group exhibited significantly fewer lung lesions and 80% survival against lethal challenge with the wild-type P. multocida strain. These results suggest that attenuated Salmonella Choleraesuis may represent a valuable platform for the development of an oral vaccine for P. multocida infections, and for use as an effective strategy against multiple serotypes. Nonetheless, it is crucial to note that the outcomes observed in mice cannot be extrapolated to pigs [54, 55]. Hence, further studies are necessary to verify the protective efficacy of rSC0016(pS-PlpE) in pigs.

Methods

Mice and ethics statement

Female Balb/c mice were purchased from Yangzhou University Comparative Medicine Centre (Jiangsu, China). All animal experiments were performed in strict accordance with the animal welfare standards of the Animal Research Committee Guidelines of Jiangsu Province (License Number: SYXK (SU) 2017–0007), and approved by the Ethics Committee for Animal Experimentation of Yangzhou University. The mice were euthanized by CO2 method when necessary. For the animal experiments, all efforts were taken to minimize the suffering of the animals. All experiments with animals were performed in accordance with the ARRIVE guidelines.

Bacterial strains, plasmids, and culture conditions

The strains and plasmids used in this work are listed in Table 1. Escherichia coli (E. coli) DH5α competent cells were used for subcloning experiments. Bacterial strains E. coli χ7213 and plasmids pYA3493 were kindly provided by R. Curtiss III (The Biodesign Institute, Arizona State University, Tempe, AZ). This plasmid encodes β-lactamase type II signals that, when fused in frame to an antigen gene of interest, directs antigen secretion to the periplasm and culture supernatant. P. multocida serotype A wild-type clinical strain separated in our laboratory were used as templates with full length primers of plpE-F and plpE-R (Table 2) to amplify plpE gene. Trypticase Soy Agar (TSA) or Trypicase Soy Broth (TSB) with 5% (v/v) fetal bovine serum was used for P. multocida strain cultivation. The S. Choleraesuis strain C78-3 (CVCC79103) was purchased from China Veterinary Culture Collection Center (CVCC, Beijing, China). E. coli and S. Choleraesuis strain C78-3 were cultured in Luria-Bertani (LB) liquid media or on LB agar (1.5% agar). In the case of rSC0016 strain, 2,6-diaminopimelic acid (DAP; 50 μg/mL) was added to the LB medium. When required, antibiotics and supplements were used at the following concentrations: Ampicillin (Amp; 50 μg/mL), Chloramphenicol (Cm; 50 μg/mL), Kanamycin (Kan; 50 μg/mL), D-mannose (0.2% wt/vol), L-arabinose (0.2% wt/vol) and D-lactose (1% wt/vol).

Table 1 Bacterial strains and plasmids used in this study
Table 2 The primers information

Bacterial identification, genotyping and virulence assay

P. multocida serotype A field strain was isolated from diseased pigs in Jiangsu Province, China. The genomic DNA of bacteria was extracted from overnight cultures of P. multocida isolate using the boiling method [56]. Serotype-specific primers or capsular typing primers were synthesized via referring to previous literature to identify P. multocida clinical strain [57]. The CFU of P. multocida field strain was calculated and then subjected to 10-fold dilution from 5.0 × 104 to 5.6 × 100 CFU. Six-week-old female BALB/c mice were randomly divided into 5 groups and mice (n = 5) in each group were challenged intraperitoneally (i.p.) with 0.1 mL of various dose of bacteria, and survival was recorded over a 14-day period. The 50% lethal doses (LD50) of the challenge strains were calculated according to the method of Reed and Muench [58].

Protein expression, purification and polyclonal antibodies

The target gene plpE was subcloned into the pET-28a plasmid (Novagen) to generate the recombinant plasmid pET-28a-PlpE. The recombinant plasmid was then confirmed by restriction analysis using EcoR I and Sal I (Takara), PCR and sequencing by TsingKe Biological Technology. Next, BL21 (DE3) (Invitrogen) was transformed with pET-28a-PlpE, which was cultured at 37 °C until the OD600 reached 0.5, and IPTG (Sangon) was added to a final concentration of 0.5 mM to induce PlpE expression. The PlpE protein was affinity purified by Ni-affinity chromatography on Ni-NTA matrix as described previously [24]. Protein concentration was estimated using the BCA Protein Assay Kit (Thermofisher) and confirmed by Western Blotting (WB) with anti-His mAb (GenScript). Polyclonal antiserums of PlpE were prepared by injecting six-week-old female BALB/c mice intramuscularly with purified PlpE protein. Each injection contained 50 μg protein mixed 1:1 (V/V) with the Quick Antibody-Mouse 3 W adjuvant (Biodragon Immunotechnologies) as recommended by the producers and booster doses with the same formulated PlpE were given at two weeks later. After a week of the boost immunization, blood was collected through submandibular vein puncture for serum preparation and tested by iELISA.

Construction of candidate vaccine strain and phenotyping properties

The plpE gene was amplified by PCR using primers that introduced EcoR I and Sal I restriction sites at the ends of the fragment. The resulting gene fragment was then inserted into the pYA3493 prokaryotic expression plasmid in the proper multiple cloning site to fuse PlpE with the β-lactamase type II secretion signal sequence, enabling PlpE expression. The success of the construction of pS-PlpE (pYA3493-PlpE) plasmid was verified by PCR, enzyme digestion, and sequencing. Upon confirmation, recombinant pS-PlpE plasmid and empty plasmid pYA3493 were introduced into rSC0016 strain by electroporation and Asd+ transformants were selected on LB plates. Expression of recombinant PlpE protein in the vaccine vector was verified by Western blotting. The plasmid stability of the rSC0016(pS-PlpE) was determined by a continuous passage experiment (one passage every 12 h) as described previously [27]. rSC0016(pYA3493) and C78-3 were used as control strains. Growth curves results were averaged across the three trials.

Vaccine preparation

The vaccine strains used in this study were prepared as the method described in previous studies [24, 27]. Briefly, rSC0016(pS-PlpE), rSC0016(pYA3493) were grown in LB broth to a cell density of approximately 1 × 109 CFU/mL (late log phase). The growth culture for the vaccine strains were supplemented with 0.2% arabinose and 0.2% mannose (Sigma). Bacterial cells were harvested by centrifugation at 7,000 × g for 10 min at 25 °C, and the pellet was resuspended in 300 μL of phosphate-buffered saline (PBS). The number of CFU were determined by plating serial dilutions onto solidified LB medium. Meanwhile, The P. multocida inactivated vaccine was prepared according to a published protocol as a control [53]. The P. multocida serotype A wild-type strain was incubated for 12 h at 37 °C in 5 mL TSB medium containing 5% (v/v) fetal bovine serum (Gibco). The revived culture was then added to same media as described above and incubated for 24 h at 37 °C. the bacterial suspension was then enumerated and the suspension adjusted, such that it contained 2 × 109 CFU/mL. The bacterial suspension was inactivated by incubation with 0.4% formaldehyde (v/v) at 37 °C for 24 h. The inactivated bacterial pellet was mixed with an equal volume of oil/water adjuvant and emulsified by using emulsification mixer (Fluko, China) to generate the inactivated P. multocida vaccine. The sterility test was also qualified to ensure that the inactivated vaccine did not contain live bacteria.

Vaccination and challenge procedure

The 195 female BALB/c mice at six-week-old were randomly divided into 5 groups. Group 1 (n = 45) and 2 (n = 45) were orally inoculated with 1 × 109 CFU of rSC0016(pS-PlpE) or rSC0016(pYA3493), respectively, and group 3 (n = 45) was oral immunization with PBS as an infected control [59]. The Group 4 (n = 45) of mice were inoculated subcutaneously with 0.2 mL of P. multocida vaccine as a control vaccine [60]. Finally, Group 5 serves as a blank control (without any interventions, mainly for pathological histological analysis). Booster immunization was carried out three weeks after the primary immunization following the same vaccine formulation and methods as with primary inoculation. Three weeks after the boost immunization, the immunized BALB/c mice in different groups were infected by intraperitoneal injection with the virulent strain of P. multocida serotype A [60]. All animal experiments were repeated at least three times, and the data were pooled and analyzed together.

Measuring antibody responses

To measure humoral and mucosal immune responses to vaccination, sera and vaginal lavages were collected seven days post-immunization (primary and boost immunization) for iELISA [24, 27]. To this end, 100 μL of purified PlpE proteins (1 ug/ml) as a capture molecule was coated in 96-well ELISA plates at 4 °C overnight. The ELISA plates were washed five times with PBST (350 μL/well) and blocked with 100 μL/well 5% skim milk for 2 h at room temperature. Test samples (serum or vaginal lavages) were twofold diluted in PBST (350 μL/well), added in duplicate, and incubated for 2 h at 37 °C. Following incubation, the plates were washed with PBST (350 μL/well), and HRP-conjugated goat anti-mouse IgG, IgG1, IgG2a, and IgA (Sigma, St. Louis, MO, USA) were added into the plates (100 μL/well) followed by 2 h incubation at 37 °C. The plates were visualized with 3,3′,5,5′-tetramethy1 benzidine (TMB) (Solarbio, Beijing, China) in the dark for 15 min at 37 °C, and the reaction was stopped with 2 M H2SO4. The antibody titre was expressed as the greatest serum dilution where OD450 was at least twice that of control sample at the same dilution.

Cytokine assays

The spleens from each group of mice were collected one week after the boost immunization, and the spleen lymphocytes were isolated according to the instructions of the mouse spleen lymphocyte separation kit (Dakewe Biotech, China). Next, spleen lymphocytes were seeded into 96-well plates at a density of 5 × 105 cells, and cultured with recombinant PlpE proteins (5 μg/mL) for 48 h. The concentration of IFN-γ and IL-4 in the culture supernatants was measured by Mouse IFN-γ and IL-4 ELISA kits (Solarbio, Beijing, China) following the manufacturer instructions. All assays were performed in triplicate.

Pathological examination

Histopathological examination (n = 15) was performed as previously described [24, 27]. The lung tissues from the mice were fixed in 4% paraformaldehyde, and then dehydrated, embedded in paraffin and sectioned. Eventually, the sections were stained with Hematoxylin and Eosin (HE). The representative sections of lung were examined and scored to assess tissue pathology, as previously described [61]. In brief, histopathological scores were mainly based on interstitial inflammation, vascular endotheliitis, bronchitis, edema, serous effusion and thrombosis. All parameters were scored separately from 0 (absence of lesions) to 3 (severe lesions).

Statistical analysis

Statistical comparisons among multiple groups were performed by one-way analysis variance (ANOVA). Analyses were conducted using Graph Pad Prism 8.0 software (Graph Pad Software Inc., CA, USA). A P-value of less than or equal to 0.05 (*) were considered significant difference, and a P-value of less than or equal to 0.01 (**) represents a highly significant difference. P-values larger than 0.05 were considered to be not significant (n.s.).

Data Availability

The original PCR and whole blot pictures are available in Supplementary material (1) The DNA sequences used are listed in Supplementary material (2) All data generated or analyzed during this study are available from the corresponding author by request.

Abbreviations

P. multocida :

Pasteurella multocida

S. Choleraesuis:

Salmonella Choleraesuis

CFU:

Colony-forming units

HE:

Hematoxylin and Eosin

i.p.:

Intraperitoneally

iELISA:

Indirect enzyme-linked immunosorbent assay

IPTG:

Isopropyl-β-d-thiogalactoside

LB:

Luria-Bertani broth

LD50 :

50% lethal dose

Ni-NTA:

Nickel-nitrilotriacetic acid

OMPs:

Outer membrane proteins

PAR:

Progressive atrophic rhinitis

PN:

pneumonic pasteurellosis

PlpE:

Pasteurella lipoprotein E

s.c.:

Subcutaneously

Th1:

Type 1 T helper

Th2:

Type 2 T helper

TSA:

Trypticase Soy Agar

TSB:

Trypicase Soy Broth

WB:

Western Blot

References

  1. Wilson BA, Ho M. Pasteurella multocida: from zoonosis to cellular microbiology. Clin Microbiol Rev. 2013;26(3):631–55.

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Harper M, Boyce JD, Adler B. Pasteurella multocida pathogenesis: 125 years after Pasteur. FEMS Microbiol Lett. 2006;265(1):1–10.

    CAS  PubMed  Google Scholar 

  3. Carter GR. Studies on Pasteurella multocida. I. A hemagglutination test for the identification of serological types. Am J Vet Res. 1955;16(60):481–4.

    CAS  PubMed  Google Scholar 

  4. Rimler RB, Rhoades KR. Serogroup F, a new capsule serogroup of Pasteurella multocida. J Clin Microbiol. 1987;25(4):615–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Peng Z, Wang H, Liang W, Chen Y, Tang X, Chen H, Wu B. A capsule/lipopolysaccharide/MLST genotype D/L6/ST11 of Pasteurella multocida is likely to be strongly associated with swine respiratory disease in China. Arch Microbiol. 2018;200(1):107–18.

    CAS  PubMed  Google Scholar 

  6. Lung O, Ohene-Adjei S, Buchanan C, Joseph T, King R, Erickson A, Detmer S, Ambagala A. Multiplex PCR and microarray for detection of Swine Respiratory Pathogens. Transbound Emerg Dis. 2017;64(3):834–48.

    CAS  PubMed  Google Scholar 

  7. Turni C, Meers J, Parke K, Singh R, Yee S, Templeton J, Mone NK, Blackall PJ, Barnes TS. Pathogens associated with pleuritic pig lungs at an abattoir in Queensland Australia. Aust Vet J. 2021;99(5):163–71.

    CAS  PubMed  Google Scholar 

  8. Snyder E, Credille B. Mannheimia haemolytica and Pasteurella multocida in bovine respiratory disease: how are they changing in response to efforts to control them? Veterinary Clin North Am Food Anim Pract. 2020;36(2):253–68.

    Google Scholar 

  9. Liu D, Chai T, Xia X, Gao Y, Cai Y, Li X, Miao Z, Sun L, Hao H, Roesler U, et al. Formation and transmission of Staphylococcus aureus (including MRSA) aerosols carrying antibiotic-resistant genes in a poultry farming environment. Sci Total Environ. 2012;426:139–45.

    PubMed  Google Scholar 

  10. Ahmad TA, Rammah SS, Sheweita SA, Haroun M, El-Sayed LH. Development of immunization trials against Pasteurella multocida. Vaccine. 2014;32(8):909–17.

    PubMed  Google Scholar 

  11. Guan LJ, Song JJ, Xue Y, Ai X, Liu ZJ, Si LF, Li MY, Zhao ZQ. Immune Protective Efficacy of China’s Traditional Inactivated and Attenuated Vaccines against the Prevalent Strains of Pasteurella multocida in Mice. Vaccines 2021, 9(10).

  12. Dessalegn B, Bitew M, Asfaw D, Khojaly E, Ibrahim SM, Abayneh T, Gelaye E, Unger H, Wijewardana V. Gamma-Irradiated Fowl Cholera Mucosal Vaccine: potential vaccine candidate for safe and effective immunization of Chicken against Fowl Cholera. Front Immunol. 2021;12:768820.

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Mostaan S, Ghasemzadeh A, Asadi Karam MR, Ehsani P, Sardari S, Shokrgozar MA, Abolhassani M, Nikbakht Brujeni G. Pasteurella multocida PlpE protein polytope as a potential subunit vaccine candidate. Vector borne and zoonotic diseases (Larchmont NY). 2021;21(11):870–4.

    Google Scholar 

  14. Okay S, Ozcengiz E, Ozcengiz G. Immune responses against chimeric DNA and protein vaccines composed of plpEN-OmpH and PlpEC-OmpH from Pasteurella multocida A:3 in mice. Acta Microbiol Immunol Hung. 2012;59(4):485–98.

    CAS  PubMed  Google Scholar 

  15. Zhang Y, Lin L, Yang J, Lv Q, Wang M, Wang F, Huang X, Hua L, Wang X, Chen H, et al. Two Bordetella bronchiseptica attenuated vaccine candidates confer protection against lethal challenge with B. Bronchiseptica and Pasteurella multocida toxin in mouse models. Vaccine. 2022;40(27):3771–80.

    CAS  PubMed  Google Scholar 

  16. Hatfaludi T, Al-Hasani K, Gong L, Boyce JD, Ford M, Wilkie IW, Quinsey N, Dunstone MA, Hoke DE, Adler B. Screening of 71 P. multocida proteins for protective efficacy in a fowl cholera infection model and characterization of the protective antigen PlpE. PLoS ONE. 2012;7(7):e39973.

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Wu JR, Shien JH, Shieh HK, Chen CF, Chang PC. Protective immunity conferred by recombinant Pasteurella multocida lipoprotein E (PlpE). Vaccine. 2007;25(21):4140–8.

    CAS  PubMed  Google Scholar 

  18. Okay S, Özcengiz E, Gürsel I, Özcengiz G. Immunogenicity and protective efficacy of the recombinant Pasteurella lipoprotein E and outer membrane protein H from Pasteurella multocida A:3 in mice. Res Vet Sci. 2012;93(3):1261–5.

    CAS  PubMed  Google Scholar 

  19. Gayet R, Bioley G, Rochereau N, Paul S, Corthésy B. Vaccination against Salmonella infection: the mucosal way. Microbiol Mol biology reviews: MMBR 2017, 81(3).

  20. Galen JE, Curtiss R. The delicate balance in genetically engineering live vaccines. Vaccine. 2014;32(35):4376–85.

    CAS  PubMed  Google Scholar 

  21. Curtiss R, Wanda SY, Gunn BM, Zhang X, Tinge SA, Ananthnarayan V, Mo H, Wang S, Kong W. Salmonella enterica serovar typhimurium strains with regulated delayed attenuation in vivo. Infect Immun. 2009;77(3):1071–82.

    CAS  PubMed  Google Scholar 

  22. Kang HY, Srinivasan J, Curtiss R. Immune responses to recombinant pneumococcal PspA antigen delivered by live attenuated Salmonella enterica serovar typhimurium vaccine. Infect Immun. 2002;70(4):1739–49.

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Wang S, Kong Q, Curtiss R. New technologies in developing recombinant attenuated Salmonella vaccine vectors. Microb Pathog. 2013;58:17–28.

    CAS  PubMed  Google Scholar 

  24. Li Q, Lv Y, Li YA, Du Y, Guo W, Chu D, Wang X, Wang S, Shi H. Live attenuated Salmonella enterica serovar Choleraesuis vector delivering a conserved surface protein enolase induces high and broad protection against Streptococcus suis serotypes 2, 7, and 9 in mice. Vaccine. 2020;38(44):6904–13.

    CAS  PubMed  Google Scholar 

  25. Li Y, Wang S, Scarpellini G, Gunn B, Xin W, Wanda SY, Roland KL, Curtiss R. Evaluation of new generation Salmonella enterica serovar typhimurium vaccines with regulated delayed attenuation to induce immune responses against PspA. Proc Natl Acad Sci USA. 2009;106(2):593–8.

    CAS  PubMed  Google Scholar 

  26. Li Y, Wang S, Xin W, Scarpellini G, Shi Z, Gunn B, Roland KL, Curtiss R. A sopB deletion mutation enhances the immunogenicity and protective efficacy of a heterologous antigen delivered by live attenuated Salmonella enterica vaccines. Infect Immun. 2008;76(11):5238–46.

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Li YA, Ji Z, Wang X, Wang S, Shi H. Salmonella enterica serovar Choleraesuis vector delivering SaoA antigen confers protection against Streptococcus suis serotypes 2 and 7 in mice and pigs. Vet Res. 2017;48(1):89.

    PubMed  PubMed Central  Google Scholar 

  28. Wilkie IW, Harper M, Boyce JD, Adler B. Pasteurella multocida: Diseases and Pathogenesis. In: Pasteurella multocida: Molecular Biology, Toxins and Infection edn. Edited by Aktories K, Orth JHC, Adler B. Berlin, Heidelberg: Springer Berlin Heidelberg; 2012: 1–22.

  29. Kang TL, Velappan RD, Kabir N, Mohamad J, Rashid NN, Ismail S. The ABA392/pET30a protein of Pasteurella multocida provoked mucosal immunity against HS disease in a rat model. Microb Pathog. 2019;128:90–6.

    CAS  PubMed  Google Scholar 

  30. Doan TD, Wang HY, Ke GM, Cheng LT. N-terminus of Flagellin Fused to an Antigen improves vaccine efficacy against Pasteurella multocida infection in chickens. Vaccines 2020, 8(2).

  31. Luo Q, Kong L, Dong J, Zhang T, Wang H, Zhang R, Lu Q, Chen H, Shao H, Jin M. Protection of chickens against fowl cholera by supernatant proteins of Pasteurella multocida cultured in an iron-restricted medium. Avian pathology: journal of the WVPA. 2019;48(3):221–9.

    CAS  Google Scholar 

  32. Leeanan TEK, Pannoi R, Anuntasomboon S, Thongkamkoon P, Thamchaipenet P. OmpA protein sequence-based typing and virulence-associated gene profiles of Pasteurella multocida isolates associated with bovine haemorrhagic septicaemia and porcine pneumonic pasteurellosis in Thailand. BMC Vet Res. 2017;13(1):243.

    PubMed  PubMed Central  Google Scholar 

  33. Adler B, Bulach D, Chung J, Doughty S, Hunt M, Rajakumar K, Serrano M, van Zanden A, Zhang Y, Ruffolo C. Candidate vaccine antigens and genes in Pasteurella multocida. J Biotechnol. 1999;73(2–3):83–90.

    CAS  PubMed  Google Scholar 

  34. Zhang XL, Jeza VT, Pan Q. Salmonella typhi: from a human pathogen to a vaccine vector. Cell Mol Immunol. 2008;5(2):91–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Takaya A, Yamamoto T, Tokoyoda K. Humoral immunity vs. Salmonella. Front Immunol. 2019;10:3155.

    CAS  PubMed  Google Scholar 

  36. Maheswaran SK, Thies ES. Influence of encapsulation on phagocytosis of Pasteurella multocida by bovine neutrophils. Infect Immun. 1979;26(1):76–81.

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Rimler RB, Register KB, Magyar T, Ackermann MR. Influence of chondroitinase on indirect hemagglutination titers and phagocytosis of Pasteurella multocida serogroups a, D and F. Vet Microbiol. 1995;47(3–4):287–94.

    CAS  PubMed  Google Scholar 

  38. Gutzeit C, Magri G, Cerutti A. Intestinal IgA production and its role in host-microbe interaction. Immunol Rev. 2014;260(1):76–85.

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Hellwig SM, van Spriel AB, Schellekens JF, Mooi FR, van de Winkel JG. Immunoglobulin A-mediated protection against Bordetella pertussis infection. Infect Immun. 2001;69(8):4846–50.

    CAS  PubMed  PubMed Central  Google Scholar 

  40. van der Pol W, Vidarsson G, Vilé HA, van de Winkel JG, Rodriguez ME. Pneumococcal capsular polysaccharide-specific IgA triggers efficient neutrophil effector functions via FcalphaRI (CD89). J Infect Dis. 2000;182(4):1139–45.

    PubMed  Google Scholar 

  41. Vidarsson G, van Der Pol WL, van Den Elsen JM, Vilé H, Jansen M, Duijs J, Morton HC, Boel E, Daha MR, Corthésy B, et al. Activity of human IgG and IgA subclasses in immune defense against Neisseria meningitidis serogroup B. J Immunol (Baltimore Md: 1950). 2001;166(10):6250–6.

    CAS  Google Scholar 

  42. Kharb S, Charan S. Mucosal immunization provides better protection than subcutaneous immunization against Pasteurella multocida (B:2) in mice preimmunized with the outer membrane proteins. Vet Res Commun. 2011;35(7):457–61.

    PubMed  Google Scholar 

  43. Brown LE, Jackson DC. Lipid-based self-adjuvanting vaccines. Curr Drug Deliv. 2005;2(4):383–93.

    CAS  PubMed  Google Scholar 

  44. Mao L, Liu C, Liu JY, Jin ZL, Jin Z, Xue RY, Feng R, Li GC, Deng Y, Cheng H, et al. Novel synthetic lipopeptides as potential mucosal Adjuvants enhanced SARS-CoV-2 rRBD-Induced Immune Response. Front Immunol. 2022;13:833418.

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Moyle PM, Toth I. Self-adjuvanting lipopeptide vaccines. Curr Med Chem. 2008;15(5):506–16.

    CAS  PubMed  Google Scholar 

  46. Nikam PS, Kingston JJ, Belagal Motatis AK. Oral co-administration of bivalent protein r-BL with U-Omp19 elicits mucosal immune responses and reduces S. Typhimurium shedding in BALB/c mice. Immunol Lett. 2021;231:61–7.

    CAS  PubMed  Google Scholar 

  47. Stevens TL, Bossie A, Sanders VM, Fernandez-Botran R, Coffman RL, Mosmann TR, Vitetta ES. Regulation of antibody isotype secretion by subsets of antigen-specific helper T cells. Nature. 1988;334(6179):255–8.

    CAS  PubMed  Google Scholar 

  48. Zimmermann P, Curtis N. Factors that influence the Immune response to vaccination. Clin Microbiol Rev 2019, 32(2).

  49. Constant SL, Bottomly K. Induction of Th1 and Th2 CD4+ T cell responses: the alternative approaches. Annu Rev Immunol. 1997;15:297–322.

    CAS  PubMed  Google Scholar 

  50. Schroder K, Hertzog PJ, Ravasi T, Hume DA. Interferon-gamma: an overview of signals, mechanisms and functions. J Leukoc Biol. 2004;75(2):163–89.

    CAS  PubMed  Google Scholar 

  51. Swain SL, Weinberg AD, English M, Huston G. IL-4 directs the development of Th2-like helper effectors. J Immunol (Baltimore Md: 1950). 1990;145(11):3796–806.

    CAS  Google Scholar 

  52. Habiela M, Seago J, Perez-Martin E, Waters R, Windsor M, Salguero FJ, Wood J, Charleston B, Juleff N. Laboratory animal models to study foot-and-mouth disease: a review with emphasis on natural and vaccine-induced immunity. J Gen Virol. 2014;95(Pt 11):2329–45.

    PubMed  PubMed Central  Google Scholar 

  53. Ran X, Meng XZ, Geng HL, Chang C, Chen X, Wen X, Ni H. Generation of porcine Pasteurella multocida ghost vaccine and examination of its immunogenicity against virulent challenge in mice. Microb Pathog. 2019;132:208–14.

    CAS  PubMed  Google Scholar 

  54. Kennedy MJ, Yancey RJ, Sanchez MS, Rzepkowski RA, Kelly SM, Curtiss R. Attenuation and immunogenicity of ∆cya ∆crp derivatives of Salmonella choleraesuis in Pigs. Infect Immun. 1999;67(9):4628–36.

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Stabel TJ, Mayfield JE, Morfitt DC, Wannemuehler MJ. Oral immunization of mice and swine with an attenuated Salmonella choleraesuis [delta cya-12 delta(crp-cdt)19] mutant containing a recombinant plasmid. Infect Immun. 1993;61(2):610–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Nnolim NE, Mpaka L, Okoh AI, Nwodo UU. Biochemical and molecular characterization of a Thermostable Alkaline Metallo-Keratinase from Bacillus sp. Nnolim-K1. Microorganisms 2020, 8(9).

  57. Townsend KM, Boyce JD, Chung JY, Frost AJ, Adler B. Genetic organization of Pasteurella multocida cap loci and development of a multiplex capsular PCR typing system. J Clin Microbiol. 2001;39(3):924–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Reed LJ, Muench H. A simple method of estimating 50% endpoints. Am J Hygiene. 1938;27:493–7.

    Google Scholar 

  59. Zhou G, Tian Y, Tian J, Ma Q, Huang S, Li Q, Wang S, Shi H. Oral immunization with attenuated Salmonella Choleraesuis expressing the P42 and P97 antigens protects mice against Mycoplasma hyopneumoniae challenge. Microbiol Spectr. 2022;10(6):e0236122.

    PubMed  Google Scholar 

  60. Li Y, Xiao J, Chang YF, Zhang H, Teng Y, Lin W, Li H, Chen W, Zhang X, Xie Q. Immunogenicity and protective efficacy of the recombinant Pasteurella multocida lipoproteins VacJ and PlpE, and outer membrane protein H from P. multocida A:1 in ducks. Front Immunol. 2022;13:985993.

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Wu C, Qin X, Li P, Pan T, Ren W, Li N, Peng Y. Transcriptomic analysis on responses of murine lungs to Pasteurella multocida infection. Front Cell Infect Microbiol. 2017;7:251.

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We would like to thank Dr Curtiss III for kindly providing χ7213, pRE112, pYA3493.

Funding

This work was supported by the National Natural Science Foundation of China (grant numbers 32172802, 31672516, 32002301, 31172300, 30670079), the State Key Laboratory of Genetically Engineered Veterinary Vaccines (No. AGVSKL-ZD202004), the China Postdoctoral Science Foundation [No. 2019M661953], and project funded by the Priority Academic Program Development of Jiangsu Higher Education Institutions (PAPD) and supported by the 111 Project D18007. This study was also supported by the Postgraduate Research & Practice Innovation Program of Jiangsu Province (Yangzhou University; No. SJCX22_1804). The funding bodies have not been involved in the design of the study as well as the collection, analysis, and interpretation of the data and manuscript writing.

Author information

Authors and Affiliations

Authors

Contributions

GDZ and JST were responsible for implementation of the assays, interpreting the data and writing the first draft; YCT and QFM were involved in certain assays; Quan Li, Shifeng Wang and Huoying Shi were involved in writing and editing this paper; Huoying Shi was involved in experiment design, was responsible for the interpretation of the data, and monitoring the exploration process; All authors read and approved the final manuscript.

Corresponding author

Correspondence to Huoying Shi.

Ethics declarations

Ethics approval and consent to participate

All animal experiments were performed in strict accordance with the animal welfare standards of the Animal Research Committee Guidelines of Jiangsu Province (License Number: SYXK (SU) 2017–0007), and approved by the Ethics Committee for Animal Experimentation of Yangzhou University. All experiments with animals were performed in accordance with the ARRIVE guidelines.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Supplementary Material 2

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhou, G., Tian, J., Tian, Y. et al. Recombinant-attenuated Salmonella enterica serovar Choleraesuis vector expressing the PlpE protein of Pasteurella multocida protects mice from lethal challenge. BMC Vet Res 19, 128 (2023). https://doi.org/10.1186/s12917-023-03679-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12917-023-03679-0

Keywords