Recombinant Neisseria surface protein A is a potential vaccine candidate against Neisseria meningitides serogroup B

  • Authors:
    • Shangyun Ying
    • Jun He
    • Minjun Yu
    • Yukuai Zhang
    • Suhong Deng
    • Lusi Zhang
    • Meihua Xie
    • Sihai Hu
  • View Affiliations

  • Published online on: June 13, 2014     https://doi.org/10.3892/mmr.2014.2325
  • Pages: 1619-1625
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Neisseria meningitidis is the pathogen of epidemic encephalomyelitis and is responsible for permanent damage to the brain and nervous system. In the present study, the prokaryotic expression vector pGEX-6p-1/neisseria surface protein A (NspA) was constructed and the immune protective effect was investigated with the purified recombinant rNspA. Female BALB/c mice were immunized by intraperitoneal inoculation of rNspA, glutathione S-transferase (GST) or phosphate-buffered saline (PBS). The protection experiment in mice demonstrated that the protection rate of the rNspA group was 85% against the N. meningitidis strain MC58, and a serum bactericidal assay in vitro revealed that the serum bactericidal titer of the rNspA group reached 1:64 following three immunizations. The levels of specific immunoglobulin (Ig) A (SIgA), IgG, IgG1, IgG2a, IgG2b and IgG3 of mice in the rNspA group peaked at week six and were higher than those in the mice in the GST and PBS groups. The levels of stimulation index, interleukin-4 and interferon-γ in the culture supernatant of the spleen lymphocytes of the rNspA group increased in a time-dependent manner and were higher than those of the mice in the GST and PBS groups over the same period. The results suggested that rNspA may induce increased specific humoral and cellular immune responses, and that it is effectively protective against N. meningitidis serogroup B in mice. The present study offered novel evidence that may lead to the development of a novel effective N. meningitidis serogroup B vaccine.

Introduction

Neisseria meningitidis, the pathogen of epidemic cerebrospinal meningitis, induces permanent damage to the brain and nervous system (1,2). This pathogen is also responsible for the development of invasive diseases caused by N. meningitidis, including septicemia, pneumonia and arthritis (35). Epidemic cerebrospinal meningitis has become a major public health problem, and developing novel methods to effectively control and prevent it has attracted worldwide attention.

Vaccinations are one of the major, most effective strategies for the control and prevention of diseases caused by N. meningitidis (4). According to the molecular structure and antigenicity of capsular polysaccharide, N. meningitidis is divided into 13 serogroups (A, B, C, D, H, I, K, L, X, Y, Z, 29E and W135), among which N. meningitidis serogroup A, B and C cause ≤90% of meningitis cases (6,7). At present, vaccines in specific to N. meningitidis serogroup A, C, Y and W135 based on capsular polysaccharides have been successively developed, which have been proved to effectively control the prevalence of epidemic cerebrospinal meningitis caused by the corresponding serogroup (810). However, recent studies have revealed that the probability of a significant increase in the prevalence of epidemic cerebrospinal meningitis caused by N. meningitidis serogroup B is high (11,12). Despite the fact that capsular polysaccharide occurs on the surface of N. meningitidis serogroup B, it is likely to trigger a cross reaction, as its structure is similar to mammalian gangliosides. Therefore, vaccines that mimic the capsular polysaccharide of N. meningitidis serogroup B are not suitable for development due to the risk of autoimmune disease (13,14).

It has been identified in recent years that Neisseria surface protein A (NspA) is a low molecular membrane protein that exists in the surface of all N. meningitidis and consists of 525 nucleotides, whose antigenicity is evolutionarily conservative (15). Protein crystal structure analysis demonstrated that NspA is composed of eight peptides with an antiparallel β-tubular structure among which, the annular extracellular section forms a relatively prolonged adhesion area. The area is mainly composed of hydrophobic residues and an anchored molecule, and therefore, it is hypothesized that NspA may functionally interact with hydrophobic materials (16). Despite the precise functioning of NspA remaining to be elucidated, it is known that its gene sequence is similar to the opacity (Opa) protein in the outer membrane protein family, and accordingly, it has the function of indirect adhesion to the host cells (17). Studies utilizing isotope-labeled anti-NspA monoclonal antibodies have confirmed that NspA is located on the surface of complete cells and is one of the most typical membrane proteins of N. meningitidis (18). Therefore, NspA is considered to be a candidate antigen for the development of a vaccine for epidemic cerebrospinal meningitis caused by N. meningitidis serogroup B.

In the present study, the constructed prokaryotic expression vector pGEX-6p-1/NspA was transfected into Escherichia (E.) coli BL21 to express the rNspA. Humoral immunity and cellular immunity levels induced by purified rNspA inoculated in BALB/c mice were detected and the immunoprotective effect was evaluated to provide a vaccine candidate aganist N. meningitidis serogroup B.

Materials and methods

Materials

The DNA Ladder (1 Kb), BamHI, NotI and T4 ligase were purchased from NEB (Hitchin, UK). Protein Marker was purchased from Fermentas (Burlington, ON, Canada). Plasmid Miniprep kit and Polymerase Chain Reaction (PCR) Product Purification kit were purchased from Omega (Norcross, GA, USA). Glutathione S-transferase (GST) Purification resin was purchased from Merck KGaA (Darmstadt, Germany). ELISA kit was purchased from eBioscience. N. meningitidis Serogroup B Diagnostic antisera were purchased from BD Biosciences (Franklin Lakes, NJ, USA).

Strains, cultivation and experimental animals

pGEX-6p-1 vector, E. coli strain JM109, Escherichia coli strain BL21 were used as conventional recombinant experiments. N. meningitidis serogroup B strain MC58 was purchased from American Type Culture Collection (ATCC; Manassas, VA, USA). Four- to five-week old, specific-pathogen free female BALB/c mice and newborn rabbits were purchased from the Faculty of Experimental Animals at the University of South China (Hengyang, China). The animals were raised on a normal diet at 25°C and 50% humidity on a 12-h light/dark cycle. The study was approved by the Ethics Committee of the University of South China (Hengyang, China).

Construction and identification of pGEX-6p-1/NspA

BamHI and NotI were selected as the restriction sites for the upstream and downstream primers, respectively. The NspA fragment was amplified by PCR using the N. meningitidis serogroup B genomic DNA as templates and the primers 5′-CGGGATCCATGAAAAAAGCACTTGCCGCACTG-3′ and 5′-TTGCGGCCCGCTAACCGCCGACAGTCGCTAC-3′ were used. Following enzyme digestion and ligation, pGEX-6P-1/NspA was constructed. The construct was confirmed by restriction digestion and sequencing.

Expression, purification and identification of rNspA

pGEX-6P-1/NspA was transformed into E. coli BL21 and induced by isopropyl β-d-1-thiogalactopyranoside (IPTG). Following ultrasonication, the supernatant was collected and GST affinity chromatography purification was conducted to obtain purified recombinant NspA protein (rNspA). The rNspA protein was stored at −70°C after its concentration was detected with the Bradford method. Mouse serum following rNspA immunization was used as the primary antibody and horseradish peroxidase-labeled sheep anti-mouse IgG (Beyotime Institute of Biotechnology, Haimen, China) was used as the secondary antibody. Western blot analysis was used to indentify rNspA.

Model construction of MC58 strain infection

A total of 40 female 9–10-week old BALB/c mice were randomly divided into four groups, with ten mice in each group. A 0.9% NaCl solution was used to dilute the bacterial suspension. N. meningitidis strain MC58 suspension of optical density (OD) A600=0.005 (a concentration equivalent to 4,000 CFU/ml) was immediately injected into the abdominal cavity of the mice. The control group was injected with phosphate-buffered saline (PBS) of the same amount. Within 72 h following the attack, the disease and survival rate of the mice was monitored. Peritoneal fluid in the dead mice and blood in the surviving mice were obtained to inoculate in chocolate agar plates (BD Biosciences) and within 12–24 h, the growth was observed. Colonies were identified by gram staining, biochemical and specificity PCR amplification.

Animal immunization and specimen collection

A total of 60 female BALB/c mice (4–5 weeks old) were selected and randomly divided into three groups, with 20 mice in each group. Immunization with 100 μg rNspA (rNspA group), 100 μg GST (GST group), PBS (control group) were administered intraperitoneally. Prior to each immunization, 50 μl of Freund’s incomplete adjuvant was added to the mixture, and immunization was performed every two weeks, for a total of three times. The mouse reproductive tract fluid and mouse tail venous blood were collected in week 0, 2, 4, 6 and 8. Following immunization, a suspension of N. meningitidis strain MC58 bacteria was utilized to bacterially challenge the mice. Two weeks following the final immunization, the mice were dissected and the spleens were obtained. A 200 nylon gauze mesh was used for filtering to produce a cell suspension.

Serum bactericidal assay (SBA)

The N. meningitidis strain MC58 suspension was mixed with rabbit complement (Pel-Freez Biologicals, Rogers, AR, USA) at a 1:1 ratio, the mixture was combined with the immune serum in a two-fold diluted concentration and cultured for 1 h at 37°C. The chocolate agar plates were then inoculated and incubated at 37°C overnight. At the same time, the diagnostic sera of N. meningitidis serogroup B were used as a positive control and PBS as a negative control. The negative control included bacterial suspension plus complement, inactivated bacteria plus complement, and serum plus bacteria plus heating deactivation complement. Deactivation complement was considered as the reference. For the serum with a bactericidal rate >50%, the reciprocal of the highest dilution multiple was as bactericidal antibody titers of serum antibody.

Immune activity assay

rNspA as the antigen was coated on 96-well plates and vaginal lavage fluid of the mice was obtained. The indirect ELISA method was used to detect specific immunoglobulin A (SIgA) levels induced by the rNspA. The serum of immune mice was collected and the indirect ELISA method was used to detect specific IgG, IgG1, IgG2a, IgG2b and IgG3 antibody levels. A total of 10 μg rNspA was used to stimulate spleen cells of the immune mice and an CCK-8 colorimetric assay was used to detect the proliferation index of the spleen lymphocytes. Three days following cultivation, the cells were collected to perform lysis by ultrasonication. The indirect ELISA method was used to detect the levels of interferon (IFN)-γ and interleukin (IL)-4.

Statistical analysis

All data are expressed as the mean ± standard deviation. Independent sample t-tests were used to perform the comparison in pairs within the groups. Multiple groups were compared using repeated data variance t-test. Statistical analysis was performed using the statistical software program, SPSS, version 13.0 (SPSS, Inc., Chicago, IL, USA). P<0.05 was considered to indicate a statistically significant difference between values.

Results

Expression, purification and identification of rNspA

The purity and native conformation of recombinant proteins is essential for the mice to be able to produce specific protective antibodies and to reduce non-specific interference. As demonstrated in Fig. 1A, the exploration consequences of induced expression demonstrated that the rNspA had the highest expression in the cell supernatant under the conditions of 30°C and 1 mmol/l IPTG. By purification of the GST resin, two proteins with molecular weights of 40 and 44 kDa, equal to the estimated values, were observed. As shown in Fig. 1B, western blot analysis with rNspA immune serum as the primary antibody identified two proteins close to 45 kDa. The results indicated that rNspA is effectively expressed in E. coli BL21.

Protective effects of rNSPA and serum bactericidal analysis

The BALB/c mouse model infected with the N. meningitidis strain MC58 was successfully constructed for the evaluation of specific protective effects induced by the candidate antigens. As demonstrated in Table I, at 72 h, the protection rate of the rNspA group was 85%, while that of the mice in the GST and PBS groups was 0%. The results revealed that rNspA had significant protective effects on mice against the N. meningitidis strain MC58.

Table I

Immune protection evaluation of BALB/c mice after rNspA immunization.

Table I

Immune protection evaluation of BALB/c mice after rNspA immunization.

Surviving micea (n)

GroupsSBA24 h48 h≥72 hSurvival %
rNspA64**20/2020/2017/2085**
GST28/200/200/200
PBS-6/200/200/200

a Post-bacterial challenge. The data are represented as the number of surviving mice in each group within 72 h.

** P<0.01, for the comparisons between the rNspA and PBS groups and between the NspA and GST groups the results were significantly different. Data are presented as the mean ± standard deviation. The values were calculated from three independent experiments.

{ label (or @symbol) needed for fn[@id='tfn3-mmr-10-03-1619'] } rNspA, recombinant neisseria surface protein A; SBA, serum bactericidal assay; PBS, phosphate-buffered saline; GST, glutathione S-transferase.

The SBA is internationally recommended as the gold standard for the evaluation of serological immune effects, which was used for the detection of functional antibodies with bactericidal activity in serum. As shown in Table I, following SBA method optimization, the serum bactericidal titer of rNspA group reached 1:64 following three immunizations; however, that of the GST group only reached 1:2 and that of the PBS group was 0. The results indicated that rNspA-induced immune serum had a complement-dependent bactericidal effect in vitro and was highly protective in mice against the N. meningitidis strain MC58.

SIgA of mice immunized with rNspA

SIgA levels were detected to determine whether the antigen may activate the mucosal immune system in mice. As revealed in Fig. 2, the levels of SIgA in the rNspA group demonstrated an upward trend with increasing time; however, the levels of SIgA decreased at week eight. At week six, following immunization, the levels of SIgA of mice in the rNspA group peaked and the antibody titer was ≤1:2,800. The levels of SIgA in the mice in the rNspA group were higher than those of the mice in the GST and PBS groups over the same time period, while no significant difference was identified between the mice in the GST and PBS groups. The results revealed that rNspA may induce high levels of mucosal immune responses in mice.

Humoral immunity in mice immunized with rNspA

The total specific IgG reflected the response levels of mice on humoral immunity and that the types of cell-mediated immunity may be reflected by the IgG subclasses. As demonstrated in Fig. 3, the levels of IgG, IgG1 and IgG2a in the rNspA group exhibited an upward trend as time increased; however, the levels of IgG, IgG1 and IgG2a decreased at week eight. At week six following immunization, the levels of IgG, IgG1 and IgG2a of the mice in the rNspA group peaked and the antibody titer was 1:8,800, 1:6,400 and 1:5,120, respectively. The levels of IgG, IgG1, IgG2a in the mice in the rNspA group were higher than those in the mice in the GST and PBS groups over the same period, while no significant difference was identified between the mice in the GST and PBS groups. The serum IgG2a/IgG1 ratios in the rNspA group at week 2, 4, 6 and 8 following immunization were 0.795 (0.307/0.386), 0.637 (0.373/0.586), 0.710 (0.490/0.690) and 0.624 (0.414/0.663), respectively. All ratios were <1. The results suggested that the rNspA-immunized mice produced a high level of humoral immune response, which was dominated by the Th2-type.

IgG2b and IgG3 have complement-mediated opsonization in the humoral immune response

As shown in Fig. 3D and E, the IgG2b antibody titers of the mice in the rNspA group reached a peak (1:3,200) at week four. The IgG3 antibody titer of the mice in the rNspA group at week six reached a peak (1:2,800). The levels of IgG2b and IgG3 in the mice in the rNspA group were higher than those in the mice in the GST and PBS groups over the same period, while no significant difference was identified between the mice in the GST and PBS groups. These results suggested that the rNspA-immunized mice produced high levels of IgG2b and IgG3, which may mediate complementary activation and regulation so as to improve the immune defense against N. meningitidis.

Cellular immunity levels in mice immunized with rNspA

The immune response state of the spleen lymphocytes in mice immunized with rNspA was determined through detection of the spleen lymphocyte stimulation index (SI). As illustrated in Fig. 4, following stimulation of the spleen lymphocytes of the mice by rNspA, the SI value of the mice in the rNspA group (1.61±0.04) was significantly higher than that of the mice in the GST (0.63±0.01) and the PBS (0.49±0.02) groups, while no significant difference was identified between the mice in the GST and PBS groups. The results suggested that the immune response of spleen lymphocytes may be activated in the mice immunized with rNspA.

Levels of IL-4 and IFN-γ in the culture supernatant of spleen lymphocytes

IL-4 stimulates B-cell proliferation and is involved in the differentiation of Th2 cells in the body’s immune response. IFN-γ has immunomodulatory functions, which activate macrophages, thus producing cytotoxic effects and improving the activity and cytotoxicity of T lymphocytes. As demonstrated in Fig. 5, the levels of IL-4 and IFN-γ in the culture supernatant of spleen lymphocytes in the rNspA group exhibited an upward trend as the time increased and at week six following immunization, the levels of IL-4 and IFN-γ in the mice in the rNspA group reached a peak. The levels of IL-4 and IFN-γ in the mice in the rNspA group were higher than those of the mice in the GST and PBS groups over the same period of time, while no significant difference was identified between the mice in the GST and PBS groups. The results revealed that the mice immunized with rNspA produced a certain level of cellular immune responses.

Discussion

N. meningitidis is the pathogen of epidemic encephalomyelitis and predominantly affects children between the ages of six months to two years, who are particularly susceptible due to their underdeveloped immune systems. The mortality rate is high following infection (13,14). Currently, among all N. meningitidis groups, the capsular polysaccharide in N. meningitidis serogroup B has a similar structure to that of the human tissue, and therefore, a vaccine based on a capsular polysaccharide from N. meningitidis serogroup B may not be used clinically as it may trigger an autoimmune disease. Therefore, to date, an effective vaccine against N. meningitidis serogroup B remains to be developed (3,4).

The establishment of an animal infection model with N. meningitidis serogroup B significantly facilitates the pathogenic study of N. meningitidis in a host and allows the evaluation of protective effects of specific candidate antigens (19). The pathogenic factors of N. meningitidis to the animal host include a capsule, fimbriae, immunoglobulin A1 protease and lipooligosaccharides (20). Mortality ensues following N. meningitidis infection in BALB/c mice (21), and therefore, BALB/c mice were selected as an animal model for N. meningitidis MC58 infection in the present study. The mortality rates of the animals were observed four weeks following infection and these data were used for evaluation of the immunoprotective effects of rNspA in mice.

As a result of the rapid development of recombinant DNA technology and reverse volcanology, new candidate antigens for N. meningitidis serogroup B are being continuously discovered; in particular, studies have focused on outer membrane proteins (22,23). NspA protein is a low molecular membrane protein that exists in the surface of all N. meningitidi. More importantly, NspA protein is highly conserved with stable antigenicity, which are optimal properties for a potential candidate vaccine antigen (16).

Previous studies have demonstrated that rNspA exists in different forms at different temperatures, e.g. 22 kDa at 95°C, 16 and 22 kDa at 105°C and 16 kDa at 125°C (23). rNspA is a small protein and its fusion with the GST tag may improve its immunogenicity. In the present study, two distinct bands were identified when rNspA was either treated at 95°C or 105°C, which may be explained by the temperature-dependent breakage of disulfide bonds. These results were consistent with the molecular size of rNspA reported by Martin et al (23).

Previous studies have revealed that rNspA may offer 80% protection against N. meningitidis strain 608B and 100% protection against N. meningitidis strain 164B in mice (24). A study reported that rNspA may offer 80% protection against N. meningitidis strain H44/76 (25) and the present study revealed that it offered 85% protection against N. meningitidis serogroup B strain MC58. These findings suggested that rNspA induced protective immunity in mice and subsequently reduced mortality in mice. During an in vitro serum bactericidal test, it was identified that the serum immunized with rNspA exhibited bactericidal effects when compared with the GST and PBS immunized mice. The mortality rate of N. meningitidis strain MC58 was >50% using the serum from primary immunized mice at a dilution ratio of 1:32 and following three immunizations. Serum at a dilution of 1:64 also eliminated 50% of N. meningitidis MC58 under the mediation of a complement system.

Mucosa is the first line of defense against invading microbes and secretory IgA effectively neutralizes pathogens (26). The SIgA titer in the lavage fluid of the reproductive tract was as high as 1:2,800 at week six in the rNspA group, which indicated that rNspA induced efficient local mucosal immunity and provided mucosal protection. N. meningitidis colonizes in nasopharyngeal mucosa and effective mucosal immunity is important for the clearance of N. meningitidis (27,28).

The serum IgG titer was as high as 1:8,800 following rNspA immunization in mice and a high level of specific IgG and its subclasses IgG1, IgG2a, IgG2b and IgG3 were detected, which indicated that rNspA may induce enhanced humoral immune responses. Several studies have demonstrated that IgG2a in serum reflects the Th1 cellular immune response and IgG1 reflects the Th2 humoral immune response (29). Following three immunizations, the IgG2a/IgG1 ratio in serum was always <1, which indicated the rNspA-induced immune response was of the Th2 type. In the cell-mediated immune response, the rNspA group demonstrated an improved effect in activating spleen lymphocytes as compared with the GST and PBS groups. Th1 cells mainly secrete type I cytokines, including IFN-γ and TNF-β, which facilitate Th0 differentiation towards Th1. Th2 cells mainly secrete type II cytokines, including IL-4 and IL-5, which facilitate Th0 differentiation towards Th2 (30). In the present study, high levels of IFN-γ and IL-4 were detected in the supernatant of mice spleen lymphocytes. These results indicated that rNspA induced cellular immunity and humoral immunity (mainly Th2) that may have a useful protective effect in mice against the N. meningitidis strain MC58.

The complement binding site in Fc fragments of IgG2b and IgG3 is exposed once they are bound to rNspA, which binds Clq and strongly activates the activity of the complement (31,32). As principal in vivo opsonin, the Fc fragment of IgG may bind to Fc receptors (FcRs) on the surface of phagocytes and FcR-bound IgG1 and IgG3 exhibit a strong opsonizing activity (33). High levels of specific IgG, IgG2a, IgG2b and IgG3 were detected in the rNspA group serum and high levels of IgG2b and IgG3 may activate the complement and enhance opsonization and a bactericidal effect (34). Furthermore, previously reported results demonstrated that rNspA, as an antigen, may induce complement-dependent bactericidal activity and enhance the body’s resistance to microbe invasion (24,26).

In conclusion, the present study demonstrated that rNspA induced higher and specific mucosal, humoral and cellular immune responses. Furthermore, rNspA-induced antibody regulated complement-dependent bactericidal activity and mediated resistance to the N. meningitidis strain MC58. The present study offers new evidence that may aid in the development of an effective N. meningitidis serogroup B vaccine and further study is required to investigate the potential clinical applications of these results.

Acknowledgements

This study was supported by the National Natural Science Foundation of China (no. 81172890), the Construct Program of the Key Discipline in Hunan Province and Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control (no. 2014-5-2012-312). The authors are grateful to Mrs. Chunxue Lu, Mrs. Lili Chen and Mr. Xiaoxing You for their excellent technical assistance and advice.

References

1 

Braunstein M, Rajkumar P, Claus CL, Vaccarelli G, Moore AJ, Wang D and Anderson MK: HEBAlt enhances the T-cell potential of fetal myeloid-biased precursors. Int Immunol. 22:963–972. 2010. View Article : Google Scholar : PubMed/NCBI

2 

van de Beek D, De Gans J, Spanjaard L, Weisfelt M, Reitsma JB and Vermeulen M: Clinical features and prognostic factors in adults with bacterial meningitis. N Engl J Med. 351:1849–1859. 2004.

3 

Harrison LH, Mohan N and Kirkpatrick P: Meningococcal group A, C, Y and W-135 conjugate vaccine. Nat Rev Drug Discov. 9:429–430. 2010. View Article : Google Scholar : PubMed/NCBI

4 

Khatami A and Pollard AJ: The epidemiology of meningococcal disease and the impact of vaccines. Expert Rev Vaccines. 9:285–298. 2010. View Article : Google Scholar : PubMed/NCBI

5 

Águeda S, Campos T and Maia A: Prediction of bacterial meningitis based on cerebrospinal fluid pleocytosis in children. Braz J Infect Dis. 17:401–404. 2013.PubMed/NCBI

6 

Raymond J: Neisseria meningitidis: characterisation and epidemiology. Arch Pediatr. 19(Suppl 2): S55–S60. 2012.(In French).

7 

Ferguson LE, Hormann MD, Parks DK and Yetman RJ: Neisseria meningitidis: presentation, treatment, and prevention. J Pediatr Health Care. 16:119–124. 2002. View Article : Google Scholar : PubMed/NCBI

8 

Findlow J, Lowe A, Deane S, Balmer P, van den Dobbelsteen G, Dawson M, Andrews N and Borrow R: Effect of sequence variation in meningococcal PorA outer membrane protein on the effectiveness of a hexavalent PorA outer membrane vesicle vaccine in toddlers and school children. Vaccine. 23:2623–2627. 2005. View Article : Google Scholar : PubMed/NCBI

9 

van den Dobbelsteen GP, van Dijken HH, Pillai S and van Alphen L: Immunogenicity of a combination vaccine containing pneumococcal conjugates and meningococcal PorA OMVs. Vaccine. 25:2491–2496. 2007.PubMed/NCBI

10 

Aaberge IS, Oster P, Helland OS, Kristoffersen AC, Ypma E, Høiby EA, Feiring B and Nøkleby H: Combined administration of meningococcal serogroup B outer membrane vesicle vaccine and conjugated serogroup C vaccine indicated for prevention of meningococcal disease is safe and immunogenic. Clin Diagn Lab Immunol. 12:599–605. 2005.

11 

Zhou H, Gao Y, Xu L, Li M, Li Q, Li Y, Liang X, Luo H, Kan B, Xu J and Shao Z: Distribution of serogroups and sequence types in disease-associated and carrier strains of Neisseria meningitidis isolated in China between 2003 and 2008. Epidemiol Infect. 140:1296–1303. 2012.PubMed/NCBI

12 

Yang L, Shao Z, Zhang X, Xu L, Peng J, Xu X, Liang X, Qi Y and Jin Q: Genotypic characterization of Neisseria meningitidis serogroup B strains circulating in China. J Infect. 56:211–218. 2008. View Article : Google Scholar : PubMed/NCBI

13 

Panatto D, Amicizia D, Lai PL and Gasparini R: Neisseria meningitidis B vaccines. Expert Rev Vaccines. 10:1337–1351. 2011. View Article : Google Scholar

14 

Silva GP, Cruz SC, Cruz AC and Milagres LG: Short-term and long-term antibody response by mice after immunization against Neisseria meningitidis B or diphtheria toxoid. Braz J Med Biol Res. 46:148–153. 2013. View Article : Google Scholar

15 

Martin D, Cadieux N, Hamel J and Brodeur BR: Highly conserved Neisseria meningitidis surface protein confers protection against experimental infection. J Exp Med. 185:1173–1183. 1997. View Article : Google Scholar

16 

Halperin SA, Langley JM, Smith B, Wunderli P, Kaufman L, Kimura A and Martin D: Phase 1 first-in-human studies of the reactogenicity and immunogenicity of a recombinant meningococcal NspA vaccine in healthy adults. Vaccine. 25:450–457. 2007. View Article : Google Scholar : PubMed/NCBI

17 

Vandeputte-Rutten L, Bos MP, Tommassen J and Gros P: Crystal structure of Neisserial surface protein A (NspA), a conserved outer membrane protein with vaccine potential. J Biol Chem. 278:24825–24830. 2003. View Article : Google Scholar : PubMed/NCBI

18 

Tsolakos N, Lie K, Bolstad K, Maslen S, Kristiansen PA, Hoiby EA, Wallington A, Vipond C, Skehel M, Tang CM, Feavers IM, Wedege E and Wheeler JX: Characterization of meningococcal serogroup B outer membrane vesicle vaccines from strain 44/76 after growth in different media. Vaccine. 28:3211–3218. 2010. View Article : Google Scholar

19 

Zimmer SM and Stephens DS: Serogroup B meningococcal vaccines. Curr Opin Investig Drugs. 7:733–739. 2006.

20 

Tamargo B, Márquez Y, Ramírez W, Cedré B, Fresno M and Sierra G: New proteoliposome vaccine formulation from N. meningitidis serogroup B, without aluminum hydroxide, retains its antimeningococcal protectogenic potential as well as Th-1 adjuvant capacity. BMC Immunol. 14(Suppl 1): S122013.PubMed/NCBI

21 

Richmond P, Kaczmarski E, Borrow R, Findlow J, Clark S, McCann R, Hill J, Barker M and Miller E: Meningococcal C polysaccharide vaccine induces immunologic hyporesponsiveness in adults that is overcome by meningococcal C conjugate vaccine. J Infect Dis. 181:761–764. 2000. View Article : Google Scholar

22 

Cassataro J, Velikovsky CA, Bruno L, Estein SM, de la Barrera S, Bowden R, Fossati CA and Giambartolomei GH: Improved immunogenicity of a vaccination regimen combining a DNA vaccine encoding Brucella melitensis outer membrane protein 31 (Omp31) and recombinant Omp31 boosting. Clin Vaccine Immunol. 14:869–874. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Martin D, Brodeur BR, Hamel J, Couture F, de Alwis U, Lian Z, Martin S, Andrews D and Ellis RW: Candidate Neisseria meningitidis NspA vaccine. J Biotechnol. 83:27–31. 2000. View Article : Google Scholar : PubMed/NCBI

24 

Cadieux N, Plante M, Rioux CR, Hamel J, Brodeur BR and Martin D: Bactericidal and cross-protective activities of a monoclonal antibody directed against Neisseria meningitidis NspA outer membrane protein. Infect Immun. 67:4955–4959. 1999.PubMed/NCBI

25 

Lewis LA, Ngampasutadol J, Wallace R, Reid JE, Vogel U and Ram S: The meningococcal vaccine candidate neisserial surface protein A (NspA) binds to factor H and enhances meningococcal resistance to complement. PLoS Pathog. 6:e10010272010. View Article : Google Scholar

26 

Arenas J, Nijland R, Rodriguez FJ, Bosma TN and Tommassen J: Involvement of three meningococcal surface-exposed proteins, the heparin-binding protein NhbA, the α-peptide of IgA protease and the autotransporter protease NalP, in initiation of biofilm formation. Mol Microbiol. 87:254–268. 2013.PubMed/NCBI

27 

Vaughan AT, Gorringe A, Davenport V, Williams NA and Heyderman RS: Absence of mucosal immunity in the human upper respiratory tract to the commensal bacteria Neisseria lactamica but not pathogenic Neisseria meningitidis during the peak age of nasopharyngeal carriage. J Immunol. 182:2231–2240. 2009. View Article : Google Scholar

28 

Davenport V, Groves E, Horton RE, Hobbs CG, Guthrie T, Findlow J, Borrow R, Naess LM, Oster P, Heyderman RS and Williams NA: Mucosal immunity in healthy adults after parenteral vaccination with outer-membrane vesicles from Neisseria meningitidis serogroup B. J Infect Dis. 198:731–740. 2008. View Article : Google Scholar : PubMed/NCBI

29 

Trotter CL, Yaro S, Njanpop-Lafourcade BM, Drabo A, Kroman SS, Idohou RS, Sanou O, Bowen L, Findlow H, Diagbouga S, Gessner BD, Borrow R and Mueller JE: Seroprevalence of bactericidal, specific IgG antibodies and incidence of meningitis due to group A Neisseria meningitidis by age in Burkina Faso 2008. PLoS One. 8:e554862013.PubMed/NCBI

30 

Zhao F, Wang S, Zhang X, Gu W, Yu J, Liu S, Zeng T, Zhang Y and Wu Y: Protective efficacy of a Treponema pallidum Gpd DNA vaccine vectored by chitosan nanoparticles and fused with interleukin-2. Can J Microbiol. 58:117–123. 2012. View Article : Google Scholar

31 

Haghi F, Peerayeh SN, Siadat SD and Montajabiniat M: Cloning, expression and purification of outer membrane protein PorA of Neisseria meningitidis serogroup B. J Infect Dev Ctries. 5:856–862. 2011.PubMed/NCBI

32 

da Hora VP, Conceição FR, Dellagostin OA and Doolan DL: Non-toxic derivatives of LT as potent adjuvants. Vaccine. 29:1538–1544. 2011.PubMed/NCBI

33 

Jung DJ, An JH, Kurokawa K, Jung YC, Kim MJ, Aoyagi Y, Matsushita M, Takahashi S, Lee HS, Takahashi K and Lee BL: Specific serum Ig recognizing staphylococcal wall teichoic acid induces complement-mediated opsonophagocytosis against Staphylococcus aureus. J Immunol. 189:4951–4959. 2012. View Article : Google Scholar

34 

Kelly DF and Rappuoli R: Reverse vaccinology and vaccines for serogroup B Neisseria meningitidis. Adv Exp Med Biol. 568:217–223. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2014
Volume 10 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ying S, He J, Yu M, Zhang Y, Deng S, Zhang L, Xie M and Hu S: Recombinant Neisseria surface protein A is a potential vaccine candidate against Neisseria meningitides serogroup B. Mol Med Rep 10: 1619-1625, 2014
APA
Ying, S., He, J., Yu, M., Zhang, Y., Deng, S., Zhang, L. ... Hu, S. (2014). Recombinant Neisseria surface protein A is a potential vaccine candidate against Neisseria meningitides serogroup B. Molecular Medicine Reports, 10, 1619-1625. https://doi.org/10.3892/mmr.2014.2325
MLA
Ying, S., He, J., Yu, M., Zhang, Y., Deng, S., Zhang, L., Xie, M., Hu, S."Recombinant Neisseria surface protein A is a potential vaccine candidate against Neisseria meningitides serogroup B". Molecular Medicine Reports 10.3 (2014): 1619-1625.
Chicago
Ying, S., He, J., Yu, M., Zhang, Y., Deng, S., Zhang, L., Xie, M., Hu, S."Recombinant Neisseria surface protein A is a potential vaccine candidate against Neisseria meningitides serogroup B". Molecular Medicine Reports 10, no. 3 (2014): 1619-1625. https://doi.org/10.3892/mmr.2014.2325