key: cord-0002044-xlrnhmon authors: Wang, Miao; Pan, Li; Zhou, Peng; Lv, Jianliang; Zhang, Zhongwang; Wang, Yonglu; Zhang, Yongguang title: Protection against Foot-and-Mouth Disease Virus in Guinea Pigs via Oral Administration of Recombinant Lactobacillus plantarum Expressing VP1 date: 2015-12-02 journal: PLoS One DOI: 10.1371/journal.pone.0143750 sha: c280d46d6fa7e2c2938a5c09bd6b092e1058a9b0 doc_id: 2044 cord_uid: xlrnhmon Mucosal vaccination is an effective strategy for generating antigen-specific immune responses against mucosal infections of foot-and-mouth disease virus (FMDV). In this study, Lactobacillus plantarum strains NC8 and WCFS1 were used as oral delivery vehicles containing a pSIP411-VP1 recombinant plasmid to initiate mucosal and systemic immune responses in guinea pigs. Guinea pigs were orally vaccinated (three doses) with NC8-pSIP411, NC8-pSIP411-VP1, WCFS1-pSIP411, WCFS1-pSIP411-VP1 or milk. Animals immunized with NC8-pSIP411-VP1 and WCFS1-pSIP411-VP1 developed high levels of antigen-specific serum IgG, IgA, IgM, mucosal secretory IgA (sIgA) and neutralizing antibodies, and revealed stronger cell-mediated immune responses and enhanced protection against FMDV challenge compared with control groups. The recombinant pSIP411-VP1 effectively improved immunoprotection against FMDV in guinea pigs. Foot-and-mouth disease (FMD) is a highly contagious disease of livestock. Outbreaks of FMD cause severe financial losses [1] , and often lead to quarantining and export limitations in affected countries, as well as culling of herds. After a 2-12 day incubation period, affected animals develop acute onset of high fever, which is followed by vesicle formation in the mouth, pharynx and on the feet. Affected animals suffer pain, refuse their feed, and salivate extensively. The causal pathogen, FMDV, belongs to the genus Aphthovirus of the family Picornaviridae and includes seven serotypes, A, O, C, Asia 1, SAT1, SAT2 and SAT3. The virion has a high potential for genetic and antigenic variation. Cross-protective antibodies are not formed after infection or vaccination by other serotypes and subtypes of FMDV. This has confounded the efforts of vaccination programs for preventing the disease [2] . The viral genome is a positive Chemically competent E. coli DH5ɑ (Takara) was transformed applying the protocol provided by the manufacturer. Lactobacilli were electro-transformed according to the protocol of Aukrust et al. but with the following modifications [17] : (1) 2% instead of 1% glycine was used in the preparation of electro-competent lactobacilli, and (2) the cells were washed three times in wash buffer (0.5mM Na-phosphate, 0.1mM MgCl 2 , pH = 7.4), and resuspended in [16] E.coli DH5ɑ Cloning strains Takara L. plantarum NC8 (NC8 CCUG 61730) Host strain; plasmid-free [17] L. plantarum WCFS1 Host strain; plasmid-free [18] electroporation buffer (0.1M sucrose, 3mM MgCl 2 , pH = 7.4) and stored at -80°C. A stationary phase (12 h) culture of recipient Lactobacillus strains, NC8 and WCFS1, were inoculated (1/100 inoculum) (v/v) into 100 mL of MRS broth (containing 2% glycine) without antibiotics at 30°C without shaking. The cells were harvested at OD 600 0.3 (about 3 h), put on the ice for 10 minutes, centrifuged at 2,000 r/min for 10 minutes at 4°C and washed two times with an equal culture volume of ice-cold wash buffer. Then the cells were resuspended in 1 mL ice-cold sucrose-magnesium chloride electroporation buffer (SMEB). 10 μL of recombination plasmid DNA was mixed with 200 μL of ice-cold competent cells in SMEB. The mixture was placed on ice for 5 minutes and then was transferred into a prechilled cuvette (inter-electrode distance 0.2 cm, Bio-Rad). A single electrical pulse was 2,500 V/cm, 2.5 μF and delivered via Gene Pluster TM (Bio-Rad). The suspension was immediately put on ice for 5 minutes and 900 μL of recovery medium was added (MRS broth without antibiotics containing 2% glycine). The bacteria were then incubated for 3 h at 30°C without agitation and at last the recombinant strains were selected on MRS agar medium containing 5 μg/mL erythromycin. For analysis of VP1 expressed by pSIP411 in NC8 and WCFS1, the recombinant strains were grown for 12 h in MRS broth containing 5 μg/mL erythromycin at 37°C without shaking. Then the overnight cultures were inoculated (1/100 inoculum) (v/v) in MRS medium and induced by adding SppIP to 100 ng/mL at OD 600 0.3 (about 3 h). Cells were harvested at OD 600 1.0 (about 7 h after induction) by centrifugation at 8,000 r/min at 4°C. The bacterial cells were washed three times with sterile phosphate-buffered saline (PBS, PH = 7.4) at 4°C and resuspended in lysis buffer (50mM Tris-HCl, 400U mutanolysin/mL, 20mg lysozyme/mL, 50 mM glucose, and Roche complete protease inhibitors) at 37°C for 1h. The bacteria were centrifuged at 8,000 r/min for 15 minutes at 4°C, and then the pellets were resuspended for analysis via 12% Sodium dodecyl sulphate-polyacrylamide gel electrophoresis (SDS-PAGE). Protein extracts were electro-transferred onto a nitrocellulose membrane for western blot analysis using swine anti-FMDV antibodies (prepared previously in our laboratory) at a dilution of 1:200 and horseradish-peroxidase (HRP)-conjugated goat anti-swine IgG antibody (Abbkine, Inc., USA) at a dilution of 1: 2,000, which was visualized with chemiluminescent substrate reagent according to the manufacturer's instructions. Cells (NC8-pSIP411, NC8-pSIP411-VP1, WCFS1-pSIP411 or WCFS1-pSIP411-VP1) grown at 37°C in MRS medium containing 5 μg/mL of erythromycin, induced at OD 600 0.3 and harvested at OD 600 1.0, were washed in PBS and resuspended in milk. Five groups (n = 6) of clean guinea pigs were orally dosed with the Lactobacillus strains with a 1.0 mL syringe-vaccinated every 7 days with 200 μL (10 9 CFU (colony forming unit) in 200 μL of milk for three Target Source sense primer P1 GAC ATG TCC TCC TGC ATC T VP1 gene This work antisense primer P2 CAC AAA TGT ACA GGG ATG GGT VP1 gene This consecutive days (i.e., the animals were vaccinated on days 1~3, 11~13 and 21~23), using milk as control groups (Table 3) . On days 0 (pre-immune), 10, 20 and 30, 500 μL of anticoagulated whole blood samples and 500 μL blood samples for serum tests were collected from whole blood of heart; 300 mg of fecal samples was obtained every 2 days in 300 μL sterile PBS (pH 7.4) containing 0.01 mol/L EDTA-Na 2 and then incubated for 12 h at 4°C. Saliva samples were also collected every 2 days from the mouth of the guinea pigs using sterile rayon balls and soaked in the PBS with 1 mM protease inhibitor. Clear extracts of all samples were stored at -80°C for subsequent analysis. Detection of antibodies using enzyme-linked immunosorbent assay (ELISA) Specific IgG in serum was tested using an indirect ELISA. 96-well EIA/RIA microplates (Costar; Corning, Inc.) were coated for 12 h at 4°C with FMDV-A antigen. After three times washing with PBS + Tween 20 (0.1%) (PBST), the wells were blocked for 2 h at 37°C with PBST containing 5% skim milk to prevent nonspecific binding and then were washed three times. Serum (1:10 dilution) was reacted with the coated wells for 1 h at 37°C. After the plates were washed three times, HRP-conjugated goat anti-guinea pig IgG antibody, diluted 1:5,000, was used to detect the bound antibodies, and the plates were incubated for an additional 50 minutes at 37°C. After another three washes, colour development was carried out using o-phenylenediamine (OPD) as the substrate, and optical absorbance was measured at 492 nm within 15 minutes after adding the stop buffer. For the ELISA tests in this study, cut off values were defined as the mean OD plus 3×standard deviation (SD) of samples from 30 guinea pigs used in this study before immunization. A Capture ELISA test was used for the detection of IgA and IgM in serum as well as sIgA in saliva and feces, according to the protocol of Satya et al. [19] . After 96-well microplates were coated for 12 h at room temperature (RT) with rabbit antiserum against FMDV (1:1,000 dilution) and washed three times with PBST, FMDV-A antigen (1:1,000 dilution) was added to each well and incubated for 1 h at 37°C. After a further wash step, test samples were added to the wells (serum 1:10 dilution, saliva 1:4 dilution and feces 1:2 dilution) and incubated for 1 h at 37°C. After the plates were washed three times, HRP-conjugated goat anti-guinea pig IgA/IgM antibody was used to detect bound antibodies, diluted 1:1,000, and incubated for additional 50 minutes at 37°C. After three times washes, colour development was carried out using OPD as the substrate, and optical absorbance was measured at 492 nm within 15 minutes after adding the stop buffer. Serum samples obtained from guinea pigs on day 30 were analyzed for neutralizing antibody titers using a neutralization assay with monolayers of BHK-21 cells [20] . 50 μL sera samples Table 3 . Immunization groups. Number Immunizing dose (10 9 CFU/mL) Vaccines were added to each well by double ratio dilution method (from 1:2 to 1:512) with DMEM (Gibco) containing 2% FBS in a 96-well plate (before adding the sera samples, the serum was inactivated by incubating at 56°C in 30 minutes). Samples obtained from the guinea pigs immunized orally with the empty vector or milk were used as controls. 50 μL of FMDV, adjusted to 100 TCID 50 by DMEM containing 2% FBS, was added to each cell of the plates containing serially diluted serum. The antibody and virus mixture were mixed and incubated at 37°C for 1 h. Then 100 μL of BHK cells (10 6 cells/mL) was added to the antibody-virus mixture and incubated with 5% CO 2 at 37°C. After 5 days incubation, CPE was measured by microscopy and each sera dilution were counted when the negative sera control and the blank control had no CPE. After the supernatant medium was discarded, 50 μL stationary liquid (10% formaldehyde) were added to each well incubated for 30 minutes and then stained with 10% formaldehyde containing 0.05% methylene blue solution for another 30 min. Read-Muench method was used to calculate the neutralizing antibody titer. The neutralizing antibody titers were calculated as the log 10 of the reciprocal of the final serum dilution that neutralized 100 TCID 50 of virus in 50% of the wells. Five guinea pigs from each immunization group (and controls) were dissected under aseptic condition, and single-lymphocyte suspensions were prepared from the spleen on day 30. Cells were labeled with carboxyfluorescein succinimidyl ester (CFSE) as described previously. CFSElabeled cells were washed three times in PBS containing 3% FBS and then resuspended in Roswell Park Memorial Institute 1640 medium (RPMI-1640) plus 10% FBS, 15 mM HEPES and 0.05 mM ß-mercaptoethanol. 1 mL cell suspensions were added to each well of the 24-well plates at 1.0~2.0×10 6 cells/mL. The cells were stimulated with 10 μg/mL concanavalin A (con A) (positive control), 10 μg/mL inactivated FMDV antigens (specific antigen stimulation) and culture medium (negative control), respectively, each group containing three parallel wells, incubated with 5% CO 2 at 37°C. After 60 h of incubation, the lymphocytes were collected and concentrated by centrifugation at 1,500 r/min for 10 minutes, and then washed three times in PBS and finally resuspended in 300 μL PBS. The data for T cell proliferation was acquired with a FACSAria flow cytometer (BD, USA). Blood samples on day 30 obtained with the anticoagulation agent EDTA were stained with R. Phycoerythrin (RPE)-conjugated mouse anti-guinea pig CD4 antibody and fluorescein isothiocyanate (FITC) conjugated mouse anti-guinea pig CD8 antibody for 30 minutes at room temperature (RT), using blood samples with RPE and FITC conjugated mouse IgG1 antibodies as negative control and blood as blank control. Subsequently, FACS lysing solution (BD FACS TM ) was added to each sample to lyse blood cells for 15 minutes at RT. The cell pellets were resuspended by PBS after three washes, and detected by a FACSAria flow cytometer. Forty days after the first immunization, anesthetized guinea pigs were challenged subcutaneously and intradermally in the left rear leg with 0.2 mL 100ID 50 of FMDV serotype-A. After 7 days of challenge, protection against FMDV was examined by clinical signs as follows: guinea pigs showing FMD-compatible lesions only at the original injection site were judged to be protected, and those showing any FMD clinical signs in the other three feet were judged to be unprotected [20] . Data handling and analysis and graphic representation were performed using GraphPad Prism 6.0 software (San Diego, CA). The data were analyzed to express the mean ±SD as the error value and one-tailed t test was used for comparison. Differences were considered to be statistically significant when P value was less than 0.05. Expression of VP1 by L. plantarum The mucosal immunity of guinea pigs was studied by measuring the anti-FMDV-VP1 IgA response in serum. Before oral immunization on day 0, there was no substantial difference in mucosal IgA levels between the experimental (NC8-pSIP411-VP1 and WCFS1-pSIP411-VP1) and control groups (NC8-pSIP411, WCFS1-pSIP411 and milk) (Fig 2(C) ) (p>0.05). Seven days following the first immunization (day 10), anti-FMDV-VP1 IgA reached high levels in experimental groups, NC8-pSIP411-VP1 and WCFS1-pSIP411-VP1 (p<0.05), while there was no obvious increase of anti-FMDV-VP1 antibody in the control groups that received pSIP411 or milk (p>0.05). Similar results for anti-FMDV-VP1 sIgA antibody were obtained for fecal and salivary samples in the immunized guinea pigs (Fig 2(A) and 2(B) ). After a second immunization (day 20), the concentration of serum IgA in the experimental groups, NC8-pSIP411-VP1 and WCFS1-pSIP411-VP1, appeared lower than the original level after the first immunization (p<0.05). When compared to the mucosal IgA levels, no significant anti-FMDV-VP1 antibodies were observed in the control groups (p>0.05). The concentrations of anti-FMDV-VP1-specific IgM from immunized guinea pigs were also determined (Fig 2(E) ) (p<0.05). After the third immunization, IgM antibodies rose to the highest level in NC8-pSIP411-VP1 and WCFS1-pSIP411-VP1 groups (p<0.05). Although there was no increase in the negative milk control (p>0.05), there again was a modest signal in NC8-pSIP411 and WCFS1-pSIP411 immunized animals. The FMDV-specific systemic immune responses of guinea pigs were studied by measuring the anti-FMDV-VP1 IgG response in serum. As the results show (Fig 2(D) ), there was no substantial difference in IgG levels between the experimental and control groups on day 0 (p>0.05). Seven days following the first immunization (day 10), anti-FMDV-VP1 IgG obtained high levels in experimental groups, NC8-pSIP411-VP1 and WCFS1-pSIP411-VP1 (p<0.05). The concentration of IgG in the experimental groups rose further after the second immunization (day 20) and reached the highest level after the third immunization (day 30) (p<0.05). No significant elicitation of anti-FMDV-VP1 IgG antibodies was observed in the milk negative control group (p>0.05). Slight increases in IgG were again noted in the NC8-pSIP411 and WCFS1-pSIP411 groups. Taken together, these results indicate that orally administered NC8-pSIP411-VP1 and WCFS1-pSIP411-VP1 were able to elicit both FMDV-specific systemic and mucosal antibody responses. Moreover, the mucosal immune responses occurred earlier than systemic responses but also declined more rapidly. To investigate cellular immune responses after oral immunization with Lactobacillus strains incorporating the recombinant plasmid, CD4 + and CD8 + T cells were assayed by flow cytometry in blood samples obtained after the third vaccination. As demonstrated in Fig 3 the CD4 + T cells and CD8 + cells in both NC8-pSIP411-VP1 and WCFS1-pSIP411-VP1 groups rose to a higher level than the control groups. A single-cell suspension of lymphocytes was prepared from immunized guinea pigs on day 30. After 60 h cultivation, the suspensions were stimulated by concanavalin A as a positive control-resulting in evident proliferation, and by medium as a negative control-with anticipated dormancy evident. Both NC8-pSIP411-VP1 and WCFS1-pSIP411-VP1 groups produced significantly high levels of T cell proliferation when stimulated by activated FMDV as a specific antigen (Fig 4) , while no obvious proliferation occurred in the control groups. These results indicate that these Lactobacillus strains expressing VP1 protein could induce antigen-specific T cell responses. The anti-viral neutralizing serum antibody titers of immunized guinea pigs were assessed (Fig 2(F) ). Serum samples were obtained from vaccinated animals on day 30 after the first immunization. Antibody responses of guinea pigs orally immunized with NC8-pSIP411-VP1 or WCFS1-pSIP411-VP1 exhibited higher FMDV-neutralizing activity than the control groups' responses, as shown by cytopathic effect (CPE) inhibition, indicating that NC8-pSIP411-VP1 and WCFS1-pSIP411-VP1 could induce neutralizing antibodies to FMDV. The guinea pigs were challenged with 100 GPID 50 (50% guinea pigs infectious dose) of homotypic virus on day 30 after the first immunization, and the protective effects of the oral vaccines were evaluated. During ten days continuous observation post-challenge, the percentages of animals in each group exhibiting clinical signs (fever, swollen soles, and secondary vesicles) are as follows: 1/5 in NC8-pSIP411-VP1 group, 3/5 in NC8-pSIP411 group, 0/5 in WCFS1-pSIP411-VP1 group, 2/5 in WCFS1-pSIP411 and 4/5 in milk group as shown in Table 4 . NC8-pSIP411-VP1 and WCFS1-pSIP411-VP1 vaccination provided at least partial protection against FMDV. Although vaccines have been useful for controlling and even eradicating FMD from parts of the world since the early 1900s, the disease still infects millions of animals each year and remains the main sanitary barrier to the commerce of animals and animal products. Vaccination is the main prophylactic method of preventing FMDV infection. Many types of vaccines have been designed-almost entirely parenterally administered-including inactivated antigen vaccines, live attenuated vaccines, DNA vaccines, empty capsid vaccines and synthetic peptide vaccines [21] . Inactivated virus vaccines can elicit high levels of neutralizing antibodies and offer efficacious protection against homologous serotypes [22] . However, these vaccines have some shortcomings, including reversion to virulence, insufficient stability, and creation of viral carrier state. These vaccines cannot prevent the virus from entering the body via mucosa [15] . FMDV infection generally occurs at mucosal surfaces, which makes mucosal immunization with specific vaccine antigens an attractive option for protection against infection. Intranasal delivery of cationic PLGA nano/microparticles loaded with various FMDV DNA vaccine formulations encoding IL-6 as a molecular adjuvant enhanced protective immunity against infection by aerosolized FMDV [20] . Another study also suggested that intranasal delivery of Chi-PLGA-DNA nanoparticles with FMDV antigen resulted in high levels of mucosal, systemic and cell-mediated immunity and could reduce disease severity and virus excretion as well as delay clinical symptoms [23] . . "*" stands for statistically significant differences relative to the milk control and "#" represents statistically significant differences relative to the WCFS1/NC8-pSIP411 control (P<0.05). Abbreviations: Ig, immunoglobulin; SD, standard deviations. doi:10.1371/journal.pone.0143750.g002 Many available approaches stimulate efficient mucosal responses, such as delivering protein or peptide antigens in association with appropriate adjuvants (e.g., liposomes and ISCOMs) [24, 25] , stimulating cellular or humoral immune responses in particulate form linked to latex microspheres [26] and using recombinant live vectors (e.g., attenuated virus or bacteria) [27] [28] [29] [30] [31] . Using lactobacilli as carriers to deliver exogenous vaccines antigens into the cytosol efficiently appears to be one of the most successful strategies [14, 32] . WCFS1, the first genome annotated Lactobacillus strain, was found that it possessed the functionality of homologous signal peptides sequence [33] . NC8, isolated from grass silage, possessed unusual property of naturally plasmid free among L. plantarum strains [34] . These two L. plantarum can colonize the GI-tract, and are recognized as emerging candidates for the expression of recombinant proteins as well as for genetic and metabolic cell engineering [16] , relevant to both medical and industrial biotechnology [35, 36] . Oral administration of lactobacilli with recombinant proteins in situ has many advantages, such as safe, convenient, less stress among animals and reduction in adverse reaction [37, 38] . Besides, lactobacilli as live vectors can persist longer in the GI-tract and some of them have intrinsic probiotic properties [39, 40] . The gene used in this study, VP1 gene, has been the hot area of research in molecular biology of FMDV for years. VP1 protein exposed on the surface of viral particles is the main antigen of neutralizing antibody responses and plays an important part in forming the virus particles [41] . Over the past decades, the partial or complete VP1 coding sequence has been used in the development of engineering vaccines accommodating the large-scale production of recombinant protein, efficiently produced and expressed in various systems, such as E. coli, transgenic plants, yeasts, baculovirus and mammalian cells [19, [42] [43] [44] [45] [46] . The sequence of VP1 gene was optimized based on the pool and codon used by L. plantarum, which reduced the GC content from 59.53% to 47.49% and without changing the gene expression, and improved the translational performance [47] . In this study, pSIP expression systems were used to express FMDV-VP1 protein. Studies are currently underway to employ lactic acid bacteria vectors to produce vaccines of many viral proteins, such as the hemagglutinin (HA) gene of H9N2 avian influenza virus, anti-rotavirus protein (Llama VHH antibody fragments), and bioactive porcine interferon-alpha [48] [49] [50] . Two L. plantarum strains were chosen in this study to carry the pSIP plasmids. Compared to the results of western blotting of these two strains, it was found that the molecular weight of target proteins expressed by WCFS1, was a little bigger than proteins expressed by NC8, on account of post-translational modification (such as phosphorylation and acetylation). Milk was used to resuspend strains in this study in order to improve taste for animals and make the oral administration safe and easy. In addition, we choose 37°C as induction temperature to imitate internal temperature of guinea pigs (the suitable temperature range of L. plantarum is 30-37°C). In the respiratory tract, oral administration of recombinant L. plantarum can induce sIgA antibodies which are considered to be a major effector in the adaptive immune defense of the respiratory mucosa. IgA antibody emerges when antigens invade mucosa, (earlier than IgG antibody), indicating that IgA functions at the early stage of viral infection. This current study demonstrated that high levels of FMDV-specific IgA were produced in serum, saliva and feces, supporting the importance of IgA antibody responses in preventing the entry and replication of FMDV. However, the full mechanisms underlying the decrease in respiratory tract infections and other symptoms are still uncertain. The production of IgG antibody occurred later that IgA, but levels became higher and lasted longer than IgA. Taken together, the results suggested that mucosal responses induced by recombinant L. plantarum were interactive with systemic responses. Our results are the first to report the immunization effects in guinea pigs using pSIP system with two L. plantarum host strains via oral immunization. The data we obtained clearly demonstrated that oral administration of VP1 proteins expressed by NC8 and WCFS1 could induce both mucosal responses and systemic responses and provide good protection against FMDV challenge. The mucosal administration of the vaccine strains resulted in a detectable FMDVspecific IgG and IgA responses. However, because of frequent mutations in VP1 gene region of field isolates, VP1 protein based immunization cannot produce an effective and stable immune response over a long period of time. Researches are seeking alternative genes which are carrying effective immuno-dominant epitopes and with less mutation. For example, VP2 of FMDV is believed to carry immuno-dominant epitopes, and publications revealed that VP2 protein shows important immunological characteristics in the development of FMDV vaccines [51] . Those studies are still in preliminary stage, and further studies in our laboratory will explore immune effects of other FMDV gene segments in combination immunologic adjuvants (for instance, IFN) expressed by LAB via mucosal routes in different animal models, especially in large animals. In the past two decades, studies mucosally administered recombinant LAB (rLAB) using animal models have successfully demonstrated major health benefits, and the field has recently moved into the era of human clinical trials, which is enough to prove the potential of rLAB as therapeutic tools for their safe and efficient use. Despite good application prospect of rLAB as therapeutic delivery vehicles, there are still many questions remaining unanswered, such as the dose of therapeutic protein delivered in situ by the rLAB after oral administration, the more effective immunization route (the intranasal route or the oral route), the best period of life (neonate or adult) [52] . This study is in the early stages of the FMD oral vaccines and needs more tests to enhance the immune effect. Further studies will be carried on larger animals which are susceptible to FMDV, such as swine and bovine, and the studies are required to consider immune effect comparison of the intranasal immunization and the oral immunization, the coexpression of larger fragments and cytokines. Many experts anticipate that, in the next decade, more applications of rLAB as therapeutic tools will be developed and optimized because LAB delivery is now a true realistic option in both human and animal medicine [13, 53, 54] . In conclusion, the oral live carrier vaccine is a promising strategy for the presentation of FMDV antigen to provide an effective, inexpensive and stable alternative to current approaches of immunization for FMDV [55] . Poverty impacts of foot-and-mouth disease and the poverty reduction implications of its control. The Veterinary record Efficacy of synthetic peptide candidate vaccines against serotype-A foot-and-mouth disease virus in cattle Biosecurity and bioterrorism: biodefense strategy, practice, and science Construction of recombinant Lactobacillus casei efficiently surface displayed and secreted porcine parvovirus VP2 protein and comparison of the immune responses induced by oral immunization Novel vaccine development strategies for inducing mucosal immunity. Expert review of vaccines The normal Lactobacillus flora of healthy human rectal and oral mucosa Identification of Lactobacillus plantarum genes that are induced in the gastrointestinal tract of mice Spatial and temporal expression of Lactobacillus plantarum genes in the gastrointestinal tracts of mice Lactobacillus plantarum-survival, functional and potential probiotic properties in the human intestinal tract To pre-challenge lactic acid bacteria with simulated gastrointestinal conditions is a suitable approach to studying potential probiotic properties Tuning constitutive recombinant gene expression in Lactobacillus plantarum Dendritic cell targeting of Bacillus anthracis protective antigen expressed by Lactobacillus acidophilus protects mice from lethal challenge Mucosal delivery of therapeutic and prophylactic molecules using lactic acid bacteria Immunogenicity of recombinant classic swine fever virus CD8(+) T lymphocyte epitope and porcine parvovirus VP2 antigen coexpressed by Lactobacillus casei in swine via oral vaccination Vaccination strategies for mucosal immune responses High-level, inducible gene expression in Lactobacillus sakei and Lactobacillus plantarum using versatile expression vectors. Microbiol-Sgm Transformation of Lactobacillus by electroporation Complete genome sequence of Lactobacillus plantarum WCFS1. Proceedings of the National Academy of Sciences of the United States of America Secretory IgA as an indicator of oro-pharyngeal foot-and-mouth disease virus replication and as a tool for post vaccination surveillance Intranasal Delivery of Cationic PLGA Nano/Microparticles-Loaded FMDV DNA Vaccine Encoding IL-6 Elicited Protective Immunity against FMDV Challenge Novel approaches to foot-and-mouth disease vaccine development. Developments in biologicals Foot and mouth disease virus vaccines Induction of mucosal immune responses and protection of cattle against direct-contact challenge by intranasal delivery with foot-and-mouth disease virus antigen mediated by nanoparticles Ovalbumin injected with complete Freund's adjuvant stimulates cytolytic responses Induction of cytotoxic T lymphocytes in vivo with protein antigen entrapped in membranous vehicles Efficient major histocompatibility complex class I presentation of exogenous antigen upon phagocytosis by macrophages Attenuated Mengo virus: a new vector for live recombinant vaccines Intragastric administration of Lactobacillus casei expressing transmissible gastroentritis coronavirus spike glycoprotein induced specific antibody production Synthetic oligonucleotide expressed by a recombinant vaccinia virus elicits therapeutic CTL Expression of Streptococcus pneumoniae antigens, PsaA (pneumococcal surface antigen A) and PspA (pneumococcal surface protein A) by Lactobacillus casei. FEMS microbiology letters Induction of immune responses in mice after intragastric administration of Lactobacillus casei producing porcine parvovirus VP2 protein Use of live bacterial vaccine vectors for antigen delivery: potential and limitations Genetically engineered Lactobacillus plantarum WCFS1 constitutively secreting heterologous oxalate decarboxylase and degrading oxalate under in vitro Genome sequence of the naturally plasmid-free Lactobacillus plantarum strain NC8 (CCUG 61730) A Food-Grade System for Inducible Gene Expression in Lactobacillus plantarum Using an Alanine Racemase-Encoding Selection Marker Novel surface display system for heterogonous proteins on Lactobacillus plantarum Lactococcus lactis as a live vector for mucosal delivery of therapeutic proteins. Hum Vaccines Lactococci and lactobacilli as mucosal delivery vectors for therapeutic proteins and DNA vaccines. Microbial cell factories Probiotics and the gut microbiota in intestinal health and disease Identification of one novel candidate probiotic Lactobacillus plantarum strain active against influenza virus infection in mice by a large-scale screening Induction of protective immune response against both PPRV and FMDV by a novel recombinant PPRV expressing FMDV VP1 High-yield production of the VP1 structural protein epitope from serotype O foot-and-mouth disease virus in Escherichia coli High expression level of a foot and mouth disease virus epitope in tobacco transplastomic plants Expression of Foot-and-Mouth Disease Virus Capsid Proteins in Silkworm-Baculovirus Expression System and Its Utilization as a Subunit Vaccine Expressions of Bovine IFN-gamma and Foot-and-Mouth Disease VP1 antigen in P. pastoris and their effects on mouse immune response to FMD antigens Adenovirus-vectored type Asia1 foot-andmouth disease virus (FMDV) capsid proteins as a vehicle to display a conserved, neutralising epitope of type O FMDV Up-regulation of MDP and tuftsin gene expression in Th1 and Th17 cells as an adjuvant for an oral Lactobacillus casei vaccine against antitransmissible gastroenteritis virus Immunoprotection against influenza virus H9N2 by the oral administration of recombinant Lactobacillus plantarumNC8 expressing hemagglutinin in BALB/c mice Co-expression of anti-rotavirus proteins (llama VHH antibody fragments) in Lactobacillus: development and functionality of vectors containing two expression cassettes in tandem Expression of bioactive porcine interferon-alpha in Lactobacillus casei Effects of amino acid substitutions in the VP2 B-C loop on antigenicity and pathogenicity of serotype Asia1 foot-and-mouth disease virus Recombinant lactic acid bacteria as mucosal biotherapeutic agents Novel applications of recombinant lactic acid bacteria in therapy and in metabolic engineering Targeting diseases with genetically engineered Lactococcus lactis and its course towards medical translation. Expert opinion on biological therapy Recombinant Lactobacillus plantarum expressing and secreting heterologous oxalate decarboxylase prevents renal calcium oxalate stone deposition in experimental rats The authors wish to thank Prof. Lars Axelsson, Nofima, Norway for providing plasmid pSIP411; Culture Collection, University of Gothenburg (CCUG), Sweden for providing L. plantarum NC8 strain; Prof. Michiel Kleerebezem, NIZO food research, the Netherlands for providing L. plantarum WCFS1 strain.