Abstract

The major etiologic agent that causes acute gastroenteritis worldwide in young animals and children is Group A rotavirus. Currently, commercially available vaccines do not often prevent porcine rotavirus (PRV) infection. In this study, we evaluated the efficacy of oral recombinant Lactobacillus vaccine against PRV in a mouse model. Lactobacillus plantarum NC8 was used as the host strain, and bacterial vectors were constructed, because the NC8 isolated has shown the capability to survive gastric transit and to colonize the intestinal tract of humans and other mammals. To explore the immunological mechanisms, lactic acid bacterial vectors were used to express VP7 antigen from PRV. We constructed an L. plantarum strain with surface-displayed VP7, named NC8-pSIP409-pgsA-VP7-DCpep. The expressed recombinant protein had a molecular weight of ∼37 kDa. The strain was used to immunize BALB/c mice to evaluate their immunomodulatory characteristics. Mice were orally immunized with recombinant L. plantarum NC8-pSIP409-pgsA-VP7-DCpep at a dose of 2 × 109 colony forming units/200 µl. The results showed that NC8-pSIP409-pgsA-VP7-DCpep significantly stimulated the differentiation of dendritic cells (DCs) in Peyer’s patches (PPs) and increased the serum levels of IL-4 and IFN-γ, as measured by enzyme-linked immunosorbent assay in mice treated with NC8-pSIP409-pgsA-VP7-DCpep. Compared to the empty vector group, NC8-pSIP409-pgsA-VP7-DCpep significantly increased the production of B220+ B cells in mesenteric lymph nodes (MLNs) and PPs and also increased the titer levels of the VP7-specific antibodies, including IgG and sIgA. The administration of NC8-pSIP409-pgsA-VP7-DCpep mediated relatively broad cellular responses. This study reveals that clear alternatives exist for PRV control strategies and provides information on PRV infection.

Introduction

There are two kinds of genetically engineered live vaccines: one is a gene deletion vaccine and the other is a vector vaccine. The gene-deficient vaccine uses genetic engineering methods to delete virulence-related genes. The virus forms pseudoviral particles and has an effective antigenic structure, but it cannot regain virulence and further replicate and multiply. The vector vaccine connects the protective antigen to the corresponding vector for expression. For example, the VP6 and VP7 genes can be inserted into a suitable vector, such as a viral vector, bacteria proteins, poxvirus, and adenovirus, using insect cells, Nicotiana benthamiana and transgenic potatoes [1–4] or bacteria, such as Salmonella, Escherichia coli, and lactic acid bacteria. The vector, as well as the antigen of interest which is expressed along with the replication of the vector, stimulates the body to produce protective antibodies [4–6]. Because most vaccines are injected subcutaneously into the muscle, this type of immunization can induce strong systemic B-cell and T-cell immune responses, but adaptive immune responses are relatively weak in the mucosal sites, such as the intestinal epithelium. In contrast, rotavirus enters the host through the oral mucosal pathway. Thus, entering the host is more likely to produce an adaptive immune response [4–6]. At present, the rotavirus protein vector vaccine is mainly an oral vaccine that uses lactic acid bacteria and E. coli as delivery vehicles. This newly constructed oral live vaccine has specificity, and the production of IgG and IgA antibodies provides complete protection against porcine rotavirus (PRV) [1–4].

PRV is recognized as a key source of acute gastroenteritis in young piglets as well as nursing and weaned piglets mainly if piglets are colostrum deficient. Of the seven serogroups (A–G), four (A, B, C, and E) rotavirus serogroups have been identified in swine, and Group A rotavirus is considered as the main rotavirus. The external capsid layer of PRV contains two viral proteins, VP4 and VP7, which contain 780 copies of the VP7 protein and are grouped as 260 trimers. The VP7 glycoprotein has 14 serotypes and is the main protein that induces the production of neutralizing antibodies, which has become the focus in vaccine research. The VP7 protein has immunogenicity and immunoreactivity, and it can induce the production of protective neutralizing antibodies [7–10]. Anti-VP7 antibodies are necessary for the protection against RV shedding. Thus, a global public health goal is the development of an effective and safe vaccine.

Lactobacillus is a facultative anaerobic, Gram-positive, microaerophilic, rod-shaped bacterium [11], and it has been confirmed to promote health and improve several health problems [12]. Lactobacillus is a food-grade bacterium. Lactobacillus plantarum is a common lactic acid bacterial strain and has a wide range of applications in industrial lactic acid fermentation and health care. Expression of a foreign antigen using a lactic acid bacterial strain induces mucosal (sIgA) and systemic (IgG) immune responses [13]. Lactic acid bacteria have the following advantages: (i) lactic acid bacteria are easy to culture, and some strains can construct a new expression vector system; (ii) the genetic manipulation method is simple, reproducible, and efficient; (iii) the genetic engineering products guarantee the relative safety of the food-grade expression system for direct oral administration; (iv) the highly regulatable promoter system is capable of expressing toxin genes; (v) the exogenous proteins obtain good expression inside and outside the cell; (vi) lactic acid bacteria effectively cause the body’s immune response that lasts for a long time; and (vii) lactic acid bacteria are safe, and the expressed foreign protein can be taken directly together with the bacteria without purification. Some studies have revealed that L. plantarum strains have probiotic effects on human health [14]. Some patients have been treated with recombinant lactobacilli that express exogenous antigens [15] with the dosages depending on the period of existence in the intestine and the expressed antigen. Many reports have suggested that the L. plantarum NC8 strain can be extensively used for vector-based expression [16]. Therefore, this strain can be used as an ideal antigen presentation vector. The expression of the VP7 gene of PRV as a recombinant fusion protein represents a relatively safe expression system for developing vaccines against PRV infection. To generate a suitable, safe, and efficient vaccine to prevent infection, a recombinant lactic acid bacterial strain that can express an antigen and thus activate the systemic and mucosa immune responses, for example L. plantarum NC8 strain, is needed [17–19]. Several experimental studies using L. plantarum NC8 strain have revealed that the strain induces an efficient immune response to a specific antigen because diverse antigens have been expressed in L. plantarum NC8 strains as probiotics [20–24].

Peptides targeting dendritic cells (DCpeps) play a central role in targeting peptides by directing innate immunity and regulating adaptive/acquired immunity. Phage display allows detection of the three DC-binding peptides containing 12 residues. Recent studies have demonstrated that DCpep binds with DCs from a wide range of species, including avian, chicken, canine, feline, and equine DCs [21,22]. In vitro, oral administration of NC8-pSIP409-NP-M1-DCpep in DCs from chicken is superior in the activation of autologous CD3+CD4+ and CD3+CD8+ T cells in the spleens of vaccinated chickens compared to control groups for avian influenza virus [21,22]. Oral administration of NC8-pSIP409-NP-M1-DCpep enhances T-cell-mediated immune responses as well as mucosal sIgA and IgG levels, providing protection against H9N2 AIV challenge. In addition, DCs pulsed with DCpep in vitro bind to hepatitis virus nonstructural protein 3 (NS3), resulting in better activation of autologous CD4+ and CD8+ T cells from hepatitis C virus-infected patients in comparison with NS3 alone. Thus, DCpep has been shown to be an effective mucosal adjuvant [22–24].

Poly-γ-glutamic acid synthetase A (pgsA) is a protein in the polyglutamic acid synthetase system of Bacillus subtilis, which attaches to some enzymatic systems on cell walls. Recently, pgsA was demonstrated to be a useful exogenous protein on the surface of L. plantarum, resulting in a surface presented with bacteria [23]. Due to its high level of surface display, the system may be functional using Gram-positive bacteria, but studies have been restricted to Lactococcus lactis and Lactobacillus casei to date. Enzymes from different sources and the pgsA bacterial surface display systems have been developed, and this anchor motif has been displayed effectively on the surface of E. coli [25]. However, the genetically engineered vaccines composed of a single recombinant antigen are inadequately immunogenic, so it is essential to increase their immunogenicity by combining with suitable adjuvants, such as cytokines and immunoregulatory agents.

In this study, we constructed a recombinant L. plantarum strain NC8-pSIP409-pgsA-VP7-DCpep that can express the heterologous VP7-DCpep protein and found that the recombinant protein substantially improved the in vivo antibody levels in BALB/c mice.

Materials and Methods

Reagents, bacterial strain, virus, and plasmid vector

The DN30209 PRV isolate VP7 mRNA (complete cds, GenBank: JN388691.1) was purchased from Genewiz (Beijing, China). The pSIP409-pgsA expression vector and the L. plantarum NC8 strain were kindly provided by Professor A. Kolandaswamy (Madurai Kamaraj University, India) and stored in the Laboratory of Jilin Provincial Engineering Research Center of Animal Probiotics [26]. Horseradish peroxidase-conjugated goat anti-rabbit IgG was purchased from Cell Signaling Technology (Boston, USA).

Construction of recombinant plasmid

The VP7-DCpep fusion gene was cleaved from the T4-VP7-DCpep plasmid (Genewiz) by incubating 10 µl of template with 30 µl of ddH2O, 5 µl of 10× buffer solution, 2.5 µl of XbaI, and 2.5 µl of HindIII at 37°C for 4 h, followed by 0.8% agarose gel electrophoresis. The expected amplicons of VP7 (1029 bp) were excised from the gel and purified with a Genomic DNA Kit (Tiangen, Beijing, China) according to the manufacturer’s instructions.

The VP7-DCpep gene fragment was ligated into the pSIP409-pgsA vector overnight at 16°C and transformed into E. coli BL21 competent cells the next day. In brief, the vector was incubated with BL21 cells in an ice bath for 30 min, followed by incubation in a 42°C water bath for 90 s and in an ice bath for another 6 min. Then, 600 µl of Luria-Bertani (LB) medium was added, and the cells were shaken at 220 rpm for 1 h at 37°C. Then, 100 µl of bacterial suspension was taken out and applied to solid culture medium containing 50 μg/ml kanamycin. After overnight incubation at 37°C, colonies were cultured on a shaker at 180 rpm for 12 h at 37°C. Monoclones were selected and cultured in 5 ml of LB liquid medium containing 50 μg/ml kanamycin. A plasmid extraction kit (Tiangen) was used to extract the plasmid according to the manufacturer’s instructions. The pSIP409-pgsA-VP7 recombinant plasmid was then verified by XbaI/HindIII digestion at 37°C in a water bath. The enzyme products were detected by 0.8% agarose gel electrophoresis and then sent to Jilin Province Comate Bioscience Co., Ltd (Changchun, China) for sequencing.

Construction of recombinant L. plantarum NC8 strain

L. plantarum NC8 strain was transformed into competent cells as previously reported [27]. Then, the competent NC8 bacterial cells were electrotransformed with recombinant plasmid following a previously described protocol [28]. In brief, pSIP409-pgsA-VP7-DCpep was mixed with the L. plantarum NC8 competent cells in an ice bath. After 5 min, the mixture was transferred into a pre-cooled 0.2 cm electric shock cuvette, subject to electrotransformation by using an electric transformer (Gene Pulser XcellTM System; Bio-Rad, Hercules, USA), producing NC8-pSIP409-pgsA-VP7-DCpep recombinant strain. Then, 800 µl of prewarmed MRS liquid medium (Solarbio, Beijing, China) containing 0.5 M sucrose was added and then transferred into a 1.5 ml centrifuge tube for anaerobic culture at 30°C for 3 h. Then, 200 μl bacterial suspension was sprayed on MRS solid medium containing 10 μg/ml erythromycin (Em; Solarbio) and cultured under anaerobic conditions at 30°C until single colonies formed. A single colony was inoculated into 5 ml MRS liquid medium (Em+) and incubated overnight at 30°C under anaerobic conditions. Then, 800 µl of the NC8-pSIP409-pgsA-VP7-DCpep bacterial suspension was mixed with 80% glycerol (200 µl) to preserve the strain and frozen at −80°C.

Sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE)

The NC8-pSIP409-pgsA-DCpep strain was subcultured, and the second generation of bacteria was added to MRS liquid medium (Em+) at 1:100 inoculum. Anaerobic culture was performed in a 30°C incubator until the optical density (OD) value reached 0.3. Then, 20 μg/ml SppIP (Sangon Biotech, Shanghai, China) was added at 1:400 (v/v) to induce protein expression overnight. The overnight culture was centrifuged at 5000 g for 10 min at 4°C. The pellet was washed three times with cold phosphate buffered saline (PBS), digested in 500 µl of pyrolysis buffer (Beyotime, Shanghai, China) for 30 min on a shaker (120 rpm) at 37°C and then centrifuged at 10,000 g for 30 min. Then, 160 µl of the supernatant containing VP7 was added to 40 µl of 5× SDS buffer, denatured by boiling at 100°C for 5 min, and centrifuged at 8000 g for 2 min. Then, 15 µl of the protein sample was subject to SDS-PAGE (5% stacking gel and 12% separation gel). The gel was then incubated with Coomassie brilliant blue staining (Beyotime) and analyzed on an Amersham Imager (General Electric Company, Shanghai, China).

Western blot analysis

After SDS-PAGE, the proteins were electro-transferred to PVDF membrane (Millipore, Darmstadt, Germany). After the transfer was completed, the PVDF membrane was blocked using blocking solution (Beyotime) for 3 h at room temperature. Then, the polyvinylidene fluoride (PVDF) membrane was incubated with the 6 × His Epitope Tag monoclonal antibody (ThermoFisher Scientific, Waltham, USA) and then horseradish peroxidase-conjugated rabbit anti-mouse secondary antibody (ThermoFisher Scientific). After washing, the blot was visualized by enhanced chemiluminescence (ECL; ThermoFisher Scientific) and analyzed on the Amersham Imager.

Flow cytometry analysis

The expression of VP7-DCpep fusion protein by NC8-pSIP409-pgsA-VP7-DCpep recombinant strain was confirmed by analyzing the fluorescence intensity on the bacterial surface. Briefly, NC8-pSIP409-pgsA-VP7-DCpep was induced as described above, and NC8-pSIP409-pgsA was used as a negative control. The bacterial suspension was centrifuged for 1 min at 14,000 g, and the bacterial cells was washed three times with PBS containing 0.5% bovine serum albumin. The cells were collected and incubated with 6 × His Epitope Tag monoclonal antibody (1:100 dilution), followed by incubation with fluorescein isothiocyanate-labeled rabbit anti-mouse IgG secondary antibody (1:100 dilution; ThermoFisher Scientific), and then detected by a BD LSR Fortessa flow cytometer (BD Bioscience, San Jose, USA). The data were analyzed using FlowJo 7.6.1 software [29].

Inoculation of mice with recombinant NC8 strain

To study the immune responses to recombinant NC8-pSIP409-pgsA-VP7-DCpep, rotavirus antibody-free SPF adult female BALB/c mice (7 weeks of age, weighing 25–30 g) were obtained from Huafukang Biotechnology Co., Ltd (Beijing, China). Briefly, mice were randomly divided into three groups (15 mice in each group): NC8-pSIP409-pgsA-VP7-DCpep (Group A), NC8-pSIP409-pgsA (Group B), and PBS (Group C). All mice received an oral dose of 2 × 109 colony forming units in 200 µl PBS for three consecutive days (days 0, 1, and 2). On days 14, 15, and 16, a booster immunization was administered, and a second booster was administered at days 28, 29, and 30. In Group C, only PBS was administered. All experiment protocols were approved by the Institutional Animal Care and Use Committee of Jilin Agricultural University (No. 20190520003).

Sample collection

Blood sample was collected from the tail on days 14 and 28 and from the eye on day 42. Serum was obtained by centrifugation and stored at −80°C until use. On day 42 after immunization, mice were weighed. Three mice from each group were sacrificed by cervical dislocation. The heart, liver, spleen, lung, and kidney (two) were weighed, and the organ index was calculated using the following equation: organ index = (organ weight/mouse body weight) × 100%.

Single-cell suspension preparation

On a super-clean platform, Peyer’s patches (PPs), mesenteric lymph nodes (MLNs), and spleens were collected and ground in a 35 mm sterile dish containing a 200-mesh steel mesh with 1 ml of RPMI 1640 medium (ThermoFisher Scientific). The cell mixture was washed twice with RPMI 1640 medium, and the supernatant was discarded. Erythrocyte lysate buffer (Beyotime) was added to lyse red blood cells. After washing, the cell suspension was passed through a nylon membrane (Solarbio) and diluted with PBS, and the number of leukocytes was then counted. A segment of duodenum was collected from the mice and washed with PBS containing phenylmethanesulfonyl fluoride (PMSF). The intestinal lavage fluid was collected and frozen at −80°C.

Detection of NC8-pSIP409-pgsA-VP7-DCpep on B cells by flow cytometry

For the surface staining of B cells, the PP and MLN cell suspensions were adjusted to 1×104 cells/μl, incubated with anti-B220-APC antibody (BD Biosciences, San Jose, USA) for 20 min to detect B cells, and then washed twice with PBS at 4°C with shaking at 2000 rpm for 5 min. Then, 300 μl of the cell suspension was placed into a fluorescence-activated cell sorting tube and subject to flow cytometric analysis on the BD LSR Fortessa flow cytometer to detect B cells.

Enzyme-linked immunosorbent assay analysis

IL-4 and IFN-γ in serum and sIgA in intestinal lavages fluid were detected by using the enzyme-linked immunosorbent assay (ELISA) kits for mouse IL-4, IFN-γ, and sIgA, respectively (R&D Systems, Minneapolis, USA). The specific anti-His tag IgG in serum was detected by a commercial ELISA kit (GenScript, Nanjing, China). The protocols were according to the manufacturer’s instructions. The OD value of each well was detected with a microplate reader (BioTek, Winooski, USA) at 450 nm.

Challenge of mice with the DN30209 Chinese PRV strain

Four weeks after the last oral immunization, three mice from each group were challenged with 4 × 104 FFU(100) 50% infective dose (ID50) of the DN30209 Chinese PRV (a kind gift from Prof. Xiaofeng Ren, Northeast Agricultural University, Harbin, China) by oral gavage to evaluate vaccine-induced protection. The survival and body weight changes of mice after PRV challenge were monitored.

Histopathological examination

Because VP7 can invade intestinal epithelial cells, we selected the small intestine of mice to detect histopathological changes. Small intestine, liver, spleen, and lung were also harvested and fixed with 4% formaldehyde at room temperature for at least 48 h. Paraffin sections were made and stained with hematoxylin and eosin (H&E; Beyotime), and histological examination was performed under a fluorescence microscope (DMI8; Leica, Germany), and representative images of small intestine, liver, lung, and spleen were taken.

Statistical analysis

FlowJo 7.6.1 software was used for flow cytometric analysis, and GraphPad Prism 5 software was used for data analysis. One-way ANOVA was used for statistical analysis of the differences between groups. P<0.05 was considered of significant difference.

Results

Cloning, expression, and purification of the rotaviral VP7 gene in the prokaryotic expression system

VP7 protein is a suitable target for the development of vaccines against rotavirus. In this study, we cloned the VP7 gene from the DN30209 Chinese PRV isolate, and sequencing and BLAST results showed that the cloned VP7 gene represents the DN30209 Chinese PRV isolate (GenBank accession No. JN388691.1) [26]. The schematic representations of the rotaviral full length VP7 are shown in Supplementary Fig. S1.

The VP7-DCpep fusion gene with His tag was successfully released by the XbaI/HindIII double enzyme digestion, resulting in a 1029 bp fragment visualized via 0.8% agarose gel electrophoresis (Supplementary Fig. S2). The pSIP409-pgsA-VP7-DCpep recombinant plasmid was verified by restriction enzyme digestion using the XbaI/HindIII, followed by detection via 0.8% agarose gel electrophoresis. Two bands were observed with the expected size (Supplementary Fig. S2).

SDS-PAGE result showed that there was no obvious specific band at 37 kDa representing VP7-DCpep fusion protein in the group of NC8-pSIP409-pgsA-VP7-DCpep (Fig. 1A, lanes 1–4). This may be attributed to the low expression level of VP7-DCpep and low sensitivity of Coomassie brilliant blue staining method. However, Western blot analysis showed that there was specific band for NC8-pSIP409-pgsA-VP7-DCpep and no band for NC8-pSIP409-pgsA, which was a negative control (Fig. 1B). These results indicated that VP7 protein was expressed on the cell wall surface of recombinant L. plantarum NC8.

Detection of VP7 protein expression (A) Expression analysis of NC8-pSIP409-pgsA-VP7-DCpep by Coomassie brilliant blue-stained SDS-PAGE of L. plantarum NC8. For the analysis, 10 µl was loaded on 10% SDS-PAGE gel, which was run at 150 V. Lane M: marker; lane 1: induced NC8-pSIP409-pgsA; lanes 2, 3, and 4: 4, 6, and 8 h after induction of NC8-pSIP409-pgsA-VP7-DCpep, respectively. (B) Western blot analysis of the expression of recombinant NC8-pSIP409-pgsA-VP7-DCpep. Lane M, low molecular weight protein marker; lanes 1 and 2, induced NC8-pSIP409-pgsA; lane 3, induced NC8-pSIP409-pgsA-VP7-DCpep.
Figure 1.

Detection of VP7 protein expression (A) Expression analysis of NC8-pSIP409-pgsA-VP7-DCpep by Coomassie brilliant blue-stained SDS-PAGE of L. plantarum NC8. For the analysis, 10 µl was loaded on 10% SDS-PAGE gel, which was run at 150 V. Lane M: marker; lane 1: induced NC8-pSIP409-pgsA; lanes 2, 3, and 4: 4, 6, and 8 h after induction of NC8-pSIP409-pgsA-VP7-DCpep, respectively. (B) Western blot analysis of the expression of recombinant NC8-pSIP409-pgsA-VP7-DCpep. Lane M, low molecular weight protein marker; lanes 1 and 2, induced NC8-pSIP409-pgsA; lane 3, induced NC8-pSIP409-pgsA-VP7-DCpep.

Confocal microscopy results showed that there was green fluorescence on the bacterial cell surface representing SppIP-induced NC8-pSIP409-pgsA-VP7-DCpep (Fig. 2B). Correspondingly, confocal microscopy showed that the SppIP-induced NC8-pSIP409-pgsA-VP7-DCpep strain expressing VP7 resulted in green fluorescence on the cells compared to the NC8-pSIP409-pgsA empty vector group that showed no green fluorescence (Fig. 2A). These results indicated that VP7 protein was expressed on the outer side of the cell wall.

Immunofluorescence analysis of VP7 protein expression in recombinant L. plantarum VP7 protein expression was analyzed by confocal fluorescence microscopy in (A) NC8-pSIP409-pgsA group and (B) recombinant NC8-pSIP409-pgsA-VP7-DCpep group. Magnification, 630 ×.
Figure 2.

Immunofluorescence analysis of VP7 protein expression in recombinant L. plantarum VP7 protein expression was analyzed by confocal fluorescence microscopy in (A) NC8-pSIP409-pgsA group and (B) recombinant NC8-pSIP409-pgsA-VP7-DCpep group. Magnification, 630 ×.

Flow cytometry results showed that the peak value of the NC8-pSIP409-pgsA-VP7-DCpep group was 28.4, while that of the NC8-pSIP409-pgsA group was 16 (Fig. 3A,B), and there was an obvious shift to right due to the expression of VP7 protein on the surface of L. plantarum (Fig. 3A). These data further indicated the expression of VP7 on the surface of recombinant L. plantarum NC8 strain.

VP7 protein expression at the surface of L. plantarum (A) VP7 protein expression at the surface of L. plantarum was detected by flow cytometry. (B) Statistical analysis of the VP7 protein expression. ***P<0.001.
Figure 3.

VP7 protein expression at the surface of L. plantarum (A) VP7 protein expression at the surface of L. plantarum was detected by flow cytometry. (B) Statistical analysis of the VP7 protein expression. ***P<0.001.

NC8-pSIP409-pgsA-VP7-DCpep promoted immune cells in mice

After 42 days of immunization, the MLN and PP junctions of three mice in each group were collected. The number of B220+ B cells in MLNs and PPs was detected by flow cytometry, with gating strategy shown in Fig. 4A,B. The results showed that the percentage of B220+ B cells in both MLNs (Fig. 4C; P<0.001) and PPs (Fig. 4D; P<0.01) in the NC8-pSIP409-pgsA-VP7-DCpep group was significantly higher than that in the NC8-pSIP409-pgsA group. These data indicated that NC8-pSIP409-pgsA-VP7-DCpep promoted the population of B cells.

The number of B220+ B cells in MLNs and PPs was detected by flow cytometry (A) FSC-A SSC-A subset. (B) B220+ subset. (C) MLN B220+ percentage of lymphocytes. (D) PPs B220+ percentage of lymphocytes. *P<0.05, **P<0.01, ***P<0.001.
Figure 4.

The number of B220+ B cells in MLNs and PPs was detected by flow cytometry (A) FSC-A SSC-A subset. (B) B220+ subset. (C) MLN B220+ percentage of lymphocytes. (D) PPs B220+ percentage of lymphocytes. *P<0.05, **P<0.01, ***P<0.001.

NC8-pSIP409-pgsA-VP7-DCpep induced specific antibodies in mice

To further evaluate the role of the NC8-pSIP409-pgsA-VP7-DCpep recombinant strain in humoral immunity, ELISA was used to detect the levels of the IFN-γ and IL-4 cytokines in mouse serum. The results showed that compared to the NC8-pSIP409-pgsA group, the levels of IL-4 (Fig. 5A) and IFN-γ (Fig. 5B) in the NC8-pSIP409-pgsA-VP7-DCpep group were increased significantly (P<0.001).

Serum cytokine levels detected by ELISA (A) IL-4 cytokine levels in mouse serum. (B) IFN-γ cytokine levels in mouse serum.***P<0.001. ns, not significant.
Figure 5.

Serum cytokine levels detected by ELISA (A) IL-4 cytokine levels in mouse serum. (B) IFN-γ cytokine levels in mouse serum.***P<0.001. ns, not significant.

As shown in Fig. 6A,B, the expression levels of specific IgG (anti-His tag) and mucosal antibody sIgA in the NC8-pSIP409-pgsA-VP7-DCpep group were also significantly higher than those in the NC8-pSIP409-pgsA group (P<0.001). These data indicated that oral administration of NC8-pSIP409-pgsA-VP7-DCpep induced mucosal immune response.

Specific antibodies detected by ELISA The expression of specific anti-His tag  (A) IgG content in serum and (B) sIgA in intestinal lavage fluid. *P<0.05, ***P<0.001.
Figure 6.

Specific antibodies detected by ELISA The expression of specific anti-His tag  (A) IgG content in serum and (B) sIgA in intestinal lavage fluid. *P<0.05, ***P<0.001.

NC8-pSIP409-pgsA-VP7-DCpep was safe for immunization in mice

In the evaluation of the safety of the recombinant NC8-pSIP409-pgsA-VP7-DCpep, we found that the mice in the NC8-pSIP409-pgsA-VP7-DCpep group had normal hair, normal food intake, normal urine output, normal fecal output, and the same body weight as those in the PBS group within a week after immunization. The weight of the mice in the NC8-pSIP409-pgsA-VP7-DCpep recombinant strain group was increased as shown in Fig. 7A,B. On day 42, there was no significant difference in the index of heart, liver, spleen, lung, and kidney (two viscera) among the NC8-pSIP409-pgsA-VP7-DCpep, NC8-pSIP409-pgsA, and PBS groups.

Safety evaluation of recombinant NC8-pSIP409-pgsA-VP7-DCpep in mice Effects on (A) body weight and (B) organ indexes after immunization. ns, not significant.
Figure 7.

Safety evaluation of recombinant NC8-pSIP409-pgsA-VP7-DCpep in mice Effects on (A) body weight and (B) organ indexes after immunization. ns, not significant.

Because PRV mainly invades small intestinal epithelial cells, we selected mouse duodenum, liver, lung, and spleen from five viscera for pathological analysis (Fig. 8AL). The intestinal villi in the NC8-pSIP409-pgsA-VP7-DCpep, NC8-pSIP409-pgsA, and PBS groups were intact, and the liver structure was clear. Moreover, the alveolar boundary in the lung was clear, and the red and white pulp boundary in spleen was clear. No pathological changes were found in these three groups, indicating that the immunization of mice with NC8-pSIP409-pgsA-VP7-DCpep was safe.

Pathological changes in mouse induced by recombinant NC8-pSIP409-pgsA-VP7-DCpep revealed by HE staining (A–C) Small intestine. (D–F) Liver. (G–I) Lung. (J–L) Spleen. A, D, G, and J represent mice treated with PBS; B, E, H, and K represent mice immunized with empty vector; and C, F, I, and L represent mice immunized with recombinant NC8-pSIP409-pgsA-VP7-DCpep. Scale bar: 200 μm.
Figure 8.

Pathological changes in mouse induced by recombinant NC8-pSIP409-pgsA-VP7-DCpep revealed by HE staining (A–C) Small intestine. (D–F) Liver. (G–I) Lung. (J–L) Spleen. A, D, G, and J represent mice treated with PBS; B, E, H, and K represent mice immunized with empty vector; and C, F, I, and L represent mice immunized with recombinant NC8-pSIP409-pgsA-VP7-DCpep. Scale bar: 200 μm.

NC8-pSIP409-pgsA-VP7-DCpep immunization protected mice against PRV challenge

To assess the protection activity of the NC8-pSIP409-pgsA-VP7-DCpep for mice against PRV infection post vaccination, a virus challenge experiment was conducted. By oral gavage, the immunized mice were challenged with 100 ID50 of the DN30209 Chinese PRV isolate strain per mouse for 3 weeks following the last vaccination. All mice survived after PRV challenge. However, mice treated with the NC8-pSIP409-pgsA-VP7-DCpep vaccine demonstrated improved body weight compared to those in the vaccine control groups receiving NC8-pSIP409-pgsA or PBS (Fig. 9A,B). These results demonstrated that oral immunization with NC8-pSIP409-pgsA-VP7-DCpep has promising protection against the PRV.

Oral immunization with NC8-pSIP409-pgsA-VP7-DCpep has promising protection against PRV challenge (A) Percent survival and (B) body weight changes of mice after infection.
Figure 9.

Oral immunization with NC8-pSIP409-pgsA-VP7-DCpep has promising protection against PRV challenge (A) Percent survival and (B) body weight changes of mice after infection.

Histopathological changes were little in mice after challenge following immunization

Because VP7 can invade intestinal epithelial cells, we selected the small intestine of mice to detect histopathological changes. Small intestine, liver, spleen, and lung were harvested from five viscera and fixed at room temperature with 4% formaldehyde for at least 48 h. Paraffin sections were made and stained with H&E for pathological analysis (Fig. 10AL). The intestinal villi in the NC8-pSIP409-pgsA-VP7-DCpep recombinant group were intact, and the liver structure was clear. Moreover, the alveolar boundary in lung was clear, and the red and white pulp boundary in spleen was clear. No pathological changes were observed, indicating that the immunization of mice with NC8-pSIP409-pgsA-VP7-DCpep was safe and resulted in an immune response (Fig. 10C). In the NC8-pSIP409-pgsA empty carrier group and PBS group, moderate villous atrophy and fusions as well as moderate crypt hyperplasia were observed throughout the small intestine sections (Fig. 10A). These data indicated that NC8-pSIP409-pgsA-VP7-DCpep was effective for protecting mice from rotavirus challenge.

Histopathological changes in mice infected with the DN30209 Chinese PRV isolate strain following immunization (A–C) Small intestine. (D–F) Liver. (G–I) Lung. (J–L) Spleen. A, D, G, and J represent mice treated with PBS; B, E, H, and K represent mice immunized with empty vector; and C, F, I, and L represent mice immunized with recombinant NC8-pSIP409-pgsA-VP7-DCpep.
Figure 10.

Histopathological changes in mice infected with the DN30209 Chinese PRV isolate strain following immunization (A–C) Small intestine. (D–F) Liver. (G–I) Lung. (J–L) Spleen. A, D, G, and J represent mice treated with PBS; B, E, H, and K represent mice immunized with empty vector; and C, F, I, and L represent mice immunized with recombinant NC8-pSIP409-pgsA-VP7-DCpep.

Discussion

In the prevention of infections, mucosal immunity is the primary barrier present in the mouth, genital tracts and respiratory system. Traditional vaccines are mainly immunized by intramuscular injection. However, oral vaccines are more convenient and time-saving. As delivery vectors for oral vaccines, bacterial vectors have attracted attention due to their advantages of easy cultivation and immunity. Attenuated pathogens, such as Salmonella and Listeria, can be used as vaccine carriers [30]. However, the application of attenuated pathogenic bacteria is limited because of their independent ancestry. However, probiotics, such as lactic acid bacteria, can be used in animals and humans because of their safety characteristics. L. lactis and Lactobacillus lactis are the most studied L. lactis vaccines [13,31]. The induction of mucosal immunity is the major effect stimulated by oral immunization because it induces sIgA production and stimulates immunity systemically. However, pathogens, such as Streptococcus pyogenes and Streptococcus pneumoniae, are also generally recognized as safe [32,33]. Some specific strains of lactic acid bacteria, Lactobacillus, Streptococcus, and Bifidobacteria are probiotics in humans and animals, maintaining gastrointestinal homeostasis, affecting other organs to produce biologically active metabolites and regulating immune parameters and intestinal tract [32,33]. Permeability plays an important role. Lactobacillus can be effectively colonized in the host cavity and plays a vital role in maintaining the balance of the natural flora. Adhesion is an important property for recombinant lactic acid bacterial vaccines because adhesion ensures long-term presence of the vaccines in the host and may extend the time the antigen enters the immune system [34]. The natural adjuvant activity of lactic acid bacteria makes it attractive in the development of oral vaccines. More importantly, lactic acid bacteria can express a variety of foreign antigens and have a certain protective effect. All of the abovementioned antigens expressed in lactic acid bacteria have a certain protective effect, which makes lactic acid bacteria more attractive and a promising host for oral vaccine production and delivery [32,33]. A live expression vector can both correctly carry and survive in the intestinal environment and express a heterogenic antigen [35,36]. We examined the possible binding and stability of orally administered NC8-pSIP409-pgsA-VP7-DCpep in the intestine of mice. NC8-pSIP409-pgsA is an ideal expression system, as it survives in the intestinal mucosa and is safe. Additionally, the use of NC8-pSIP409-pgsA in the food industry represents a beneficial use of these types of organisms. NC8-pSIP409-pgsA is one of the most utilized species in food industry, medicine, and fermentation. NC8-pSIP409-pgsA rapidly grows and is simple to process, making it a perfect expression vector for exogenous gene products [36,37]. Oral vaccination in mice with the NC8-pSIP409-pgsA strain as an expression vector for the rotavirus VP7 surface antigen was effective and safe. Oral immunization with the recombinant strain elicited specific humoral and cellular immunity.

Many protective antigens can be expressed in L. lactis [38,39] and Lactobacillus lactis [40–42]. In addition, L. lactis in Lactobacillus can express rotavirus proteins, such as VP6 and VP7, which allows more effective delivery of the plasmid DNA to the host cells, improving the expression of exogenous antigens and enhancing humoral immune response. In our lab, we mainly use L. plantarum NC8 as host bacteria to deliver protective antigens, such as avian influenza, Newcastle disease, transmissible gastroenteritis, and porcine epidemic diarrhea [26,43–46]. The construction of recombinant-expressing heterologous pathogen antigens has been performed in several lactobacilli, such as the cholera toxin B subunit, transmissible gastroenteritis coronavirus spike glycoprotein, and the S. pneumoniae antigens PsaA and PspA [47,48]. For mucosal immunization, organisms have been revealed to be good live vehicles for the delivery of antigens. In the present study, we established that the NC8-pSIP409-pgsA-VP7-DCpep recombinant strain may be used to produce protective anti-VP7 antibodies and stimulate mucosal and systemic immunization following mucosal vaccination in a mouse model.

In addition, induction of antigen-presenting cells and immunostimulatory responses can be regulated by probiotic vaccines, such as DCs and macrophages [49]. Some studies have noted that the immunostimulatory role has tissue-specific differences in DC level and in stimulating the responses of T helper cells [50,51]. Splenic DCs specifically induce Th1 responses, while DCs isolated from mucosal tissues are involved in the promotion of Th2 responses. Our focus was on the variation in the IFN-γ and IL-4 levels in the serum. When activated in vivo, DCs stimulate T cells to differentiate into helper T cells, such as Th1 and Th2, which play an important role in secreting IFN-γ and IL-4 cytokines, respectively. After the activation of DCs, VP7-expressing L. plantarum cells stimulate T cells to secrete more IL-4, which results in T cell differentiating into Th2. This result is consistent with previously reports showing that the production of IFN-γ and IL-4 cytokines was increased [52–54]. Therefore, we speculate that although the VP7-expressing L. plantarum vector may be limited as a DNA vaccine vector, the presence of VP7-DCpep may play an adjuvant role in the co-expression of exogenous antigens.

Antibody-mediated passive immunization can provide protection against infection by invading pathogens [55]. Therefore, it is important to develop novel passive immunization approaches to prevent PRV infection. PRV VP7 is capable of inducing neutralizing antibodies and mediating adaptive immune responses [7]. Our results demonstrated that the use of NC8-pSIP409-pgsA-VP7-DCpep as the PRV VP7 antigen following oral vaccination in mice was efficient and safe. Following immunization with NC8-pSIP409-pgsA-VP7-DCpep, all mice were in a good condition. None of the mice showed any illness, and none of the mice died. Because no studies have been conducted on recombinant NC8-pSIP409-pgsA-VP7-DCpep, we mainly explored the role of VP7-expressing L. plantarum in cellular and humoral immunity, such as differentiation of helper T cells and B cells. According to the ELISA results, the antibody titers of VP7-specific IgG (P=0.0012) and sIgA (P<0.001) in serum were increased in mice treated with NC8-pSIP409-pgsA-VP7-DCpep compared to mice treated with the empty vector. NC8-pSIP409-pgsA-VP7-DCpep increased the number of B220+ B cells in the PP junction, promoted the production of VP7-specific antibodies, and improved the specific humoral immune response. The production of B220+ B cells was also significantly increased in MLNs (P<0.001) and in PPs (P<0.001).

We mainly explored the role of VP7-expressing L. plantarum in cellular and humoral immunity, such as activation of helper T cells and B cells. First, VP7-expressing L. plantarum enhanced the infection protection in vivo, as confirmed by flow cytometry. At the same time, VP7-expressing L. plantarum increased the expression of IL-4 by approximately 3 folds, suggesting that the infection protection may be related to the production of IL-4. It has been reported that IL-4 is related to the efficiency of the vaccine [56,57]. L. plantarum strains can be used as delivery vectors for oral vaccines due to their probiotic properties, and the VP7 protein can change the probiotic characteristics of L. plantarum. Moreover, the safety of this system has also attracted attention. Pathological analysis confirmed that the expression of VP7 on L. plantarum surface was safe and did not cause pathological damage. L. plantarum expressing VP7 activates DCs in vivo and promotes the differentiation of CD4+ T cells into Th1 and Th2, and it further promotes the expression of B220+ B cells in PP junctions, promotes the production of VP7-specific antibodies, and improves the specific humoral immune response. Future studies should concentrate on investigating the NC8-pSIP409-pgsA-VP7-DCpep recombinant strain vaccine in a porcine model of the disease as a means of evaluating the efficiency of the formulation against infections in its natural host.

Supplementary Data

supplementary data is available at Acta Biochimica et Biophysica Sinica online.

Funding

This work was supported by the grants from the National Key Research and Development Program of China (Nos. 2017YFD0501000, 2017YFD0501200, and 2017YFD0500400), the National Natural Science Foundation of China (No. 31672528) and the Science and Technology Development Program of Jilin Province (Nos. 20180201040NY, 20190301042NY, and 20200402041NC).

Conflict of Interest

The authors declare that they have no conflict of interest.

References

1.

Khodabandehloo
 
M
,
Shahrabadi
 
MS
,
Keyvani
 
H
,
Bambai
 
B
,
Sadigh
 
Z
.
Recombinant outer capsid glycoprotein (VP7) of rotavirus expressed in insect cells induces neutralizing antibodies in rabbits
.
Iran J Public Health
 
2012
,
41
:
73
84
.

2.

Pêra
 
FFPG
,
Mutepfa
 
DLR
,
Khan
 
AM
,
Els
 
JH
,
Mbewana
 
S
,
van Dijk
 
AAA
,
Rybicki
 
EP
, et al.  
Engineering and expression of a human rotavirus candidate vaccine in Nicotiana benthamiana
.
Virol J
 
2015
,
12
: 205. doi:

3.

Wu
 
YZ
,
Li
 
JT
,
Mou
 
ZR
,
Fei
 
L
,
Ni
 
B
,
Geng
 
M
,
Jia
 
ZC
, et al.  
Oral immunization with rotavirus VP7 expressed in transgenic potatoes induced high titers of mucosal neutralizing IgA
.
Virology
 
2003
,
313
:
337
342
. doi:

4.

Wang
 
L
,
Huang
 
JA
,
Nagesha
 
HS
,
Smith
 
SC
,
Phelps
 
A
,
Holmes
 
I
,
Martyn
 
JC
, et al.  
Bacterial expression of the major antigenic regions of porcine rotavirus VP7 induces a neutralizing immune response in mice
.
Vaccine
 
1999
,
17
:
2636
2645
. doi:

5.

Li
 
Y
,
Guan
 
L
,
Liu
 
X
,
Liu
 
W
,
Yang
 
J
,
Zhang
 
X
,
Wang
 
F
, et al.  
Oral immunization with rotavirus VP7-CTB fusion expressed in transgenic Arabidopsis thaliana induces antigen-specific IgA and IgG and passive protection in mice
.
Exp Ther Med
 
2018
,
15
:
4866
4874
.

6.

Desselberger
 
U
,
Huppertz
 
H-I
.
Immune responses to rotavirus infection and vaccination and associated correlates of protection
.
J Infect Dis
 
2011
,
203
:
188
195
. doi:

7.

Blutt
 
SE
,
Crawford
 
SE
,
Warfield
 
KL
,
Lewis
 
DE
,
Estes
 
MK
,
Conner
 
ME
.
The VP7 outer capsid protein of rotavirus induces polyclonal B-cell activation
.
J Virol
 
2004
,
78
:
6974
6981
. doi:

8.

Kim
 
Y
,
Chang
 
KO
,
Kim
 
WY
,
Saif
 
LJ
.
Production of hybrid double- or triple-layered virus-like particles of group A and C rotaviruses using a baculovirus expression system
.
Virology
 
2002
,
302
:
1
8
. doi:

9.

Rosen
 
BI
,
Parwani
 
AV
,
Lopez
 
S
,
Flores
 
J
,
Saif
 
LJ
.
Serotypic differentiation of rotaviruses in field samples from diarrheic pigs by using nucleic acid probes specific for porcine VP4 and human and porcine VP7 genes
.
J Clin Microbiol
 
1994
,
32
:
311
317
. doi:

10.

Gatti
 
MSV
,
Ferraz
 
MMG
,
Rácz
 
ML
,
de Castro
 
AFP
.
Rotavirus excretion in naturally infected pigs with and without diarrhoea
.
Vet Microbiol
 
1993
,
37
:
187
190
. doi:

11.

Wang
 
Z
,
Yu
 
Q
,
Gao
 
J
,
Yang
 
Q
.
Mucosal and systemic immune responses induced by recombinant Lactobacillus spp. expressing the hemagglutinin of the avian influenza virus H5N1
.
Clin Vaccine Immunol
 
2012
,
19
:
174
179
. doi:

12.

Dicks
 
LM
,
Botes
 
M
.
Probiotic lactic acid bacteria in the gastro-intestinal tract: health benefits, safety and mode of action
.
Benef Microbes
 
2010
,
1
:
11
29
. doi:

13.

Bermudez-Humaran
 
LG
,
Kharrat
 
P
,
Chatel
 
JM
,
Langella
 
P
.
Lactococci and lactobacilli as mucosal delivery vectors for therapeutic proteins and DNA vaccines
.
Microb Cell Fact
 
2011
,
10
: S4. doi:

14.

Mercenier
 
A
,
Pavan
 
S
,
Pot
 
B
.
Probiotics as biotherapeutic agents: present knowledge and future prospects
.
Curr Pharm Des
 
2003
,
9
:
175
191
. doi:

15.

Gilbert
 
C
,
Robinson
 
K
,
le Page
 
RW
,
Wells
 
JM
.
Heterologous expression of an immunogenic pneumococcal type 3 capsular polysaccharide in Lactococcus lactis
.
Infect Immun
 
2000
,
68
:
3251
3260
. doi:

16.

Sorvig
 
E
,
Gronqvist
 
S
,
Naterstad
 
K
,
Mathiesen
 
G
,
Eijsink
 
VG
,
Axelsson
 
L
.
Construction of vectors for inducible gene expression in Lactobacillus sakei and L plantarum
.
FEMS Microbiol Lett
 
2003
,
229
:
119
126
. doi:

17.

Mathipa
 
MG
,
Thantsha
 
MS
.
Probiotic engineering: towards development of robust probiotic strains with enhanced functional properties and for targeted control of enteric pathogens
.
Gut Pathog
 
2017
,
9
: 28. doi:

18.

Michon
 
C
,
Langella
 
P
,
Eijsink
 
VGH
,
Mathiesen
 
G
,
Chatel
 
JM
.
Display of recombinant proteins at the surface of lactic acid bacteria: strategies and applications
.
Microb Cell Fact
 
2016
,
15
: 70. doi:

19.

Shonyela
 
SM
,
Wang
 
G
,
Yang
 
W
,
Yang
 
G
,
Wang
 
C
.
New progress regarding the use of lactic acid bacteria as live delivery vectors, treatment of diseases and induction of immune responses in different host species focusing on Lactobacillus species
.
World J Vaccines
 
2017
,
74
: 33.

20.

Yang
 
G
,
Jiang
 
Y
,
Yang
 
W
,
Du
 
F
,
Yao
 
Y
,
Shi
 
C
,
Wang
 
C
.
Effective treatment of hypertension by recombinant Lactobacillus plantarum expressing angiotensin converting enzyme inhibitory peptide
.
Microb Cell Fact
 
2015
,
14
: 202. doi:

21.

Yang
 
WT
,
Shi
 
SH
,
Yang
 
GL
,
Jiang
 
YL
,
Zhao
 
L
,
Li
 
Y
,
Wang
 
CF
.
Cross-protective efficacy of DCs targeting conserved influenza virus antigen expressed by Lactobacillus plantarum
.
Sci Rep
 
2016
,
6
: 39665. doi:

22.

Yang
 
WT
,
Yang
 
GL
,
Shi
 
SH
,
Liu
 
YY
,
Huang
 
HB
,
Jiang
 
YL
,
Wang
 
JZ
, et al.  
Protection of chickens against H9N2 avian influenza virus challenge with recombinant Lactobacillus plantarum expressing conserved antigens
.
Appl Microbiol Biotechnol
 
2017
,
101
:
4593
4603
. doi:

23.

Yang
 
G
,
Yao
 
J
,
Yang
 
W
,
Jiang
 
Y
,
Du
 
J
,
Huang
 
H
,
Gu
 
W
, et al.  
Construction and immunological evaluation of recombinant Lactobacillus plantarum expressing SO7 of Eimeria tenella fusion DC-targeting peptide
.
Vet Parasitol
 
2017
,
236
:
7
13
. doi:

24.

Yang
 
WT
,
Yang
 
GL
,
Shi
 
SH
,
Liu
 
YY
,
Huang
 
HB
,
Jiang
 
YL
,
Wang
 
JZ
, et al.  
Protection of chickens against H9N2 avian influenza virus challenge with recombinant Lactobacillus plantarum expressing conserved antigens
.
Appl Microbiol Biotechnol
 
2017
,
101
:
4593
4603
.

25.

Narita
 
J
,
Okano
 
K
,
Tateno
 
T
,
Tanino
 
T
,
Sewaki
 
T
,
Sung
 
MH
,
Fukuda
 
H
, et al.  
Display of active enzymes on the cell surface of Escherichia coli using PgsA anchor protein and their application to bioconversion
.
Appl Microbiol Biotechnol
 
2006
,
70
:
564
572
. doi:

26.

Huang
 
KY
,
Yang
 
GL
,
Jin
 
YB
,
Liu
 
J
,
Chen
 
HL
,
Wang
 
PB
,
Jiang
 
YL
, et al.  
Construction and immunogenicity analysis of Lactobacillus plantarum expressing a porcine epidemic diarrhea virus S gene fused to a DC-targeting peptide
.
Virus Res
 
2018
,
247
:
84
93
. doi:

27.

Hanahan
 
D
.
Studies on transformation of Escherichia coli with plasmids
.
J Mol Biol
 
1983
,
166
:
557
580
. doi:

28.

Dower
 
WJ
,
Miller
 
JF
,
Ragsdale
 
CW
.
High efficiency transformation of E.coli by high voltage electroporation
.
Nucleic Acids Res
 
1988
,
16
:
6127
6145
. doi:

29.

Kikuchi
 
Y
,
Kunitoh-Asari
 
A
,
Hayakawa
 
K
,
Imai
 
S
,
Kasuya
 
K
,
Abe
 
K
,
Adachi
 
Y
, et al.  
Oral administration of Lactobacillus plantarum strain AYA enhances IgA secretion and provides survival protection against influenza virus infection in mice
.
PLoS One
 
2014
,
9
: e86416. doi:

30.

Lin
 
IY
,
Van
 
TT
,
Smooker
 
PM
.
Live-attenuated bacterial vectors: tools for vaccine and therapeutic agent delivery
.
Vaccines (Basel)
 
2015
,
3
:
940
972
. doi:

31.

Allain
 
T
,
Mansour
 
NM
,
Bahr
 
MM
,
Martin
 
R
,
Florent
 
I
,
Langella
 
P
,
Bermudez-Humaran
 
LG
.
A new lactobacilli in vivo expression system for the production and delivery of heterologous proteins at mucosal surfaces
.
FEMS Microbiol Lett
 
2016
,
363
:
117
125
. doi:

32.

Tsai
 
YT
,
Cheng
 
PC
,
Pan
 
TM
.
The immunomodulatory effects of lactic acid bacteria for improving immune functions and benefits
.
Appl Microbiol Biotechnol
 
2012
,
96
:
853
862
. doi:

33.

de Vrese
 
M
,
Schrezenmeir
 
J
.
Probiotics, prebiotics, and synbiotics
.
Adv Biochem Eng Biotechnol
 
2008
,
111
:
1
66
.

34.

Szatraj
 
K
,
Szczepankowska
 
AK
,
Chmielewska-Jeznach
 
M
.
Lactic acid bacteria-promising vaccine vectors: possibilities, limitations, doubts
.
J Appl Microbiol
 
2017
,
123
:
325
339
. doi:

35.

Serradell
 
MC
,
Rupil
 
LL
,
Martino
 
RA
,
Prucca
 
CG
,
Carranza
 
PG
,
Saura
 
A
,
Fernández
 
EA
, et al.  
Efficient oral vaccination by bioengineering virus-like particles with protozoan surface proteins
.
Nat Commun
 
2019
,
10
: 361. doi:

36.

Maffey
 
L
,
Vega
 
CG
,
Miño
 
S
,
Garaicoechea
 
L
,
Parreño
 
V
.
Anti-VP6 VHH: an experimental treatment for rotavirus A-associated disease
.
PLoS One
 
2016
,
11
: e0162351. doi:

37.

Myrbråten
 
IS
,
Wiull
 
K
,
Salehian
 
Z
,
Håvarstein
 
LS
,
Straume
 
D
,
Mathiesen
 
G
,
Kjos
 
M
.
CRISPR interference for rapid knockdown of essential cell cycle genes in Lactobacillus plantarum
.
mSphere
 
2019
,
4
:
e00007
e00019
. doi:

38.

Torkashvand
 
A
,
Bahrami
 
F
,
Adib
 
M
,
Ajdary
 
S
.
Mucosal and systemic immune responses elicited by recombinant Lactococcus lactis expressing a fusion protein composed of pertussis toxin and filamentous hemagglutinin from Bordetella pertussis
.
Microb Pathog
 
2018
,
120
:
155
160
. doi:

39.

Su
 
H
,
Li
 
P
,
Yang
 
C
,
Jin
 
M
,
Chen
 
H
,
Zhou
 
H
.
Prevalence and genetic evolution of EA-H1N1 swine influenza virus in China
.
Scientia Sinica Vitae
 
2018
,
48
:
1287
1294
. doi:

40.

Maqsood
 
I
,
Shi
 
W
,
Wang
 
L
,
Wang
 
X
,
Han
 
B
,
Zhao
 
H
,
Nadeem
 
AM
, et al.  
Immunogenicity and protective efficacy of orally administered recombinant Lactobacillus plantarum expressing VP2 protein against IBDV in chicken
.
J Appl Microbiol
 
2018
,
125
:
1670
1681
. doi:

41.

Komatsu
 
A
,
Igimi
 
S
,
Kawana
 
K
.
Optimization of human papillomavirus (HPV) type 16 E7-expressing lactobacillus-based vaccine for induction of mucosal E7-specific IFNgamma-producing cells
.
Vaccine
 
2018
,
36
:
3423
3426
. doi:

42.

Sun
 
Y
,
Qian
 
J
,
Xu
 
X
,
Tang
 
Y
,
Xu
 
W
,
Yang
 
W
,
Jiang
 
Y
, et al.  
Dendritic cell-targeted recombinantLactobacilli induce DC activation and elicit specific immune responses against G57 genotype of avian H9N2 influenza virus infection
.
Vet Microbiol
 
2018
,
223
:
9
20
. doi:

43.

Yang
 
WT
,
Yang
 
GL
,
Yang
 
X
,
Shonyela
 
SM
,
Zhao
 
L
,
Jiang
 
YL
,
Huang
 
HB
, et al.  
Recombinant Lactobacillus plantarum expressing HA2 antigen elicits protective immunity against H9N2 avian influenza virus in chickens
.
Appl Microbiol Biotechnol
 
2017
,
101
:
8475
8484
. doi:

44.

Jiang
 
Y
,
Hu
 
J
,
Guo
 
Y
,
Yang
 
W
,
Ye
 
L
,
Shi
 
C
,
Liu
 
Y
, et al.  
Construction and immunological evaluation of recombinant Lactobacillus plantarum expressing HN of Newcastle disease virus and DC-targeting peptide fusion protein
.
J Biotechnol
 
2015
,
216
:
82
89
. doi:

45.

Jin
 
YB
,
Yang
 
WT
,
Shi
 
CW
,
Feng
 
B
,
Huang
 
KY
,
Zhao
 
GX
,
Li
 
QY
, et al.  
Immune responses induced by recombinant Lactobacillus plantarum expressing the spike protein derived from transmissible gastroenteritis virus in piglets
.
Appl Microbiol Biotechnol
 
2018
,
102
:
8403
8417
. doi:

46.

Yang
 
WT
,
Li
 
QY
,
Ata
 
EB
,
Jiang
 
YL
,
Huang
 
HB
,
Shi
 
CW
,
Wang
 
JZ
, et al.  
Immune response characterization of mice immunized with Lactobacillus plantarum expressing spike antigen of transmissible gastroenteritis virus
.
Appl Microbiol Biotechnol
 
2018
,
102
:
8307
8318
. doi:

47.

LeCureux
 
JS
,
Dean
 
GA
.
Lactobacillus mucosal vaccine vectors: immune responses against bacterial and viral antigens
.
mSphere
 
2018
,
3
:
e00061
-
18
. doi:

48.

Oliveira
 
ML
,
Areas
 
AP
,
Campos
 
IB
,
Monedero
 
V
,
Perez-Martinez
 
G
,
Miyaji
 
EN
,
Leite
 
LC
, et al.  
Induction of systemic and mucosal immune response and decrease in Streptococcus pneumoniae colonization by nasal inoculation of mice with recombinant lactic acid bacteria expressing pneumococcal surface antigen A
.
Microbes Infect
 
2006
,
8
:
1016
1024
. doi:

49.

Drakes
 
M
,
Blanchard
 
T
,
Czinn
 
S
.
Bacterial probiotic modulation of dendritic cells
.
Infect Immun
 
2004
,
72
:
3299
3309
. doi:

50.

Banchereau
 
J
,
Steinman
 
RM
.
Dendritic cells and the control of immunity
.
Nature
 
1998
,
392
:
245
252
. doi:

51.

Everson
 
MP
,
Lemak
 
DG
,
McDuffie
 
DS
,
Koopman
 
WJ
,
McGhee
 
JR
,
Beagley
 
KW
.
Dendritic cells from Peyer’s patch and spleen induce different T helper cell responses
.
J Interferon Cytokine Res
 
1998
,
18
:
103
115
. doi:

52.

Suradhat
 
S
,
Braun
 
RP
,
Lewis
 
PJ
,
Babiuk
 
LA
,
van Drunen Littel-van den Hurk
 
S
,
Griebel
 
PJ
,
Baca-Estrada
 
ME
.
Fusion of C3d molecule with bovine rotavirus VP7 or bovine herpesvirus type 1 glycoprotein D inhibits immune responses following DNA immunization
.
Vet Immunol Immunopathol
 
2001
,
83
:
79
92
. doi:

53.

Kaufhold
 
RM
,
Field
 
JA
,
Caulfield
 
MJ
,
Wang
 
S
,
Joseph
 
H
,
Wooters
 
MA
,
Green
 
T
, et al.  
Memory T-cell response to rotavirus detected with a gamma interferon enzyme-linked immunospot assay
.
J Virol
 
2005
,
79
:
5684
5694
. doi:

54.

Liu
 
YY
,
Yang
 
WT
,
Shi
 
SH
,
Li
 
YJ
,
Zhao
 
L
,
Shi
 
CW
,
Zhou
 
FY
, et al.  
Immunogenicity of recombinant Lactobacillus plantarum NC8 expressing goose parvovirus VP2 gene in BALB/c mice
.
J Vet Sci
 
2017
,
18
:
159
167
. doi:

55.

Casadevall
 
A
,
Dadachova
 
E
,
Pirofski
 
LA
.
Passive antibody therapy for infectious diseases
.
Nat Rev Microbiol
 
2004
,
2
:
695
703
. doi:

56.

Mani
 
R
,
Gupta
 
M
,
Malik
 
A
,
Tandon
 
R
,
Prasad
 
R
,
Bhatnagar
 
R
,
Banerjee
 
N
.
Adjuvant potential of poly-alpha-l-glutamine from the cell wall of mycobacterium tuberculosis
.
Infect Immun
 
2018
,
86
:
e00537
-
18
. doi:

57.

Matsuzaki
 
C
,
Takagaki
 
C
,
Higashimura
 
Y
,
Nakashima
 
Y
,
Hosomi
 
K
,
Kunisawa
 
J
,
Yamamoto
 
K
, et al.  
Immunostimulatory effect on dendritic cells of the adjuvant-active exopolysaccharide from Leuconostoc mesenteroides strain NTM048
.
Biosci Biotechnol Biochem
 
2018
,
82
:
1647
1651
. doi:

Author notes

Seria Masole Shonyela, Chunwei Shi contributed equally to this work.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/journals/pages/open_access/funder_policies/chorus/standard_publication_model)

Supplementary data