Skip to main content
Erschienen in: Malaria Journal 1/2017

Open Access 01.12.2017 | Research

Naturally acquired humoral and cellular immune responses to Plasmodium vivax merozoite surface protein 8 in patients with P. vivax infection

verfasst von: Yang Cheng, Bo Wang, Siriruk Changrob, Jin-Hee Han, Jetsumon Sattabongkot, Kwon-Soo Ha, Patchanee Chootong, Feng Lu, Jun Cao, Myat Htut Nyunt, Won Sun Park, Seok-Ho Hong, Chae Seung Lim, Takafumi Tsuboi, Eun-Taek Han

Erschienen in: Malaria Journal | Ausgabe 1/2017

Abstract

Background

Thirty-one glycosylphosphatidylinositol (GPI)-anchored proteins of Plasmodium vivax, merozoite surface protein 1 (MSP1), MSP1 paralogue, MSP4, MSP5, MSP8, and MSP10 have been reported from homologs of Plasmodium falciparum by gene annotation with bioinformatics tools. These GPI-anchored proteins contain two epidermal growth factor (EGF)-like domains at its C-terminus. Here, P. vivax merozoite surface protein 8 (PvMSP8) are considered as potential targets of protective immunity.

Methods

Recombinant PvMSP8 (rPvMSP8) was expressed, purified, and used for the assessment of humoral and cellular immune responses in P. vivax-infected patients and immune mice. Moreover, the target epitope of ant-PvMSP8 antibodies and subcellular localization of PvMSP8 was also determined.

Results

The rPvMSP8 was successfully expressed and purified as soluble form as ~55 kDa. PvMSP8 was localized to the outer circle of pigments associated with the food vacuole. The rPvMSP8 protein had a high antigenicity (73.2% in sensitivity and 96.2% in specificity) in patients infected with P. vivax. IgG2 antibody subtype was the predominantly responses to this antigen. Antibody response to PvMSP8 increased up to day 7 and after that slightly decreased within a month. The longevity of anti-PvMSP8 antibody was stably sustained up to 12-year recovery patient samples. Most anti-PvMSP8 antibodies recognized two epitopes that were located outside the C-terminal EGF-like domain. The cellular immune response in P. vivax-exposed individuals produced high levels of IFN-γ and IL-10 upon PvMSP8 antigen stimulation in vitro.

Conclusions

All data in this study suggest that PvMSP8 antigen has a potential to induce both humoral and cellular immune responses in patients with P. vivax infection. The subcellular localization of PvMSP8 confirmed that it was associated with the parasite food vacuole in blood-stage parasites. A further characterization of this protein will be useful for blood stage P. vivax vaccine development.

Background

The merozoite surface protein 8 (MSP8) is one of the glycosylphosphatidylinositol (GPI)-anchored proteins of blood-stage malaria parasites. It contains a signal sequence at N-terminus and two epidermal growth factor (EGF)-like domains at C-terminus with significant homology to those of Plasmodium species [14]. In previous reports, the immunization with full-length Plasmodium yoelii MSP8 (PyMSP8) fused with P. yoelii MSP1-19 induced MSP8-restricted T cell response and high and sustained levels of protective PyMSP1-19- and PyMSP8-specific antibody responses [5]. As a malaria vaccine candidate, the conserved, immunogenic T-cell epitope located in the C terminus of Plasmodium falciparum MSP8 (PfMSP8) (ΔAsn/Asp) is useful for protective efficacy, together with the fusion partner of poor immunogens such as PfMSP1-19/MSP8 (ΔAsn/Asp) [6]. The dominant B cells epitopes were mapped onto the C terminus of PfMSP8 antigen, which supports a high immunogenicity of PfMSP8 for further vaccine development [6]. However, another previous P. falciparum MSP8 knock-out study showed that it is not required for asexual stage parasite growth and replication [7]. Thus these results indicated that the immunogenicity of MSP8 may different from Plasmodium species.
PfMSP8 has been confirmed to localize to the parasitophorous vacuole (PV) of infected erythrocytes. Intriguingly, its C terminus is found in the food vacuole (FV) [8]. In blood-stage parasites, the parasite can be protected within the PV in erythrocytes, which are cells devoid of proteins, lipid biosynthesis, and intracellular compartments [9]. Plasmodium parasites internalize host cell haemoglobin, which is degraded in a specialized compartment, the FV [10]. Some characteristics of the FV, such as a low pH and the presence of proteolytic enzymes, may lead to its classification as a lysosome-like organelle [11]. This highly specialized organelle is present only in Plasmodium blood stages; in other words, it is absent from the mosquito and liver stages and is not found in other apicomplexan parasites. In addition to PfMSP8, the C terminus of another well-known GPI-anchored protein, PfMSP1, also constitutes the FV [12, 13]. The findings described above suggest that PfMSP8 plays a distinctive role in FV of infected erythrocytes.
However, as a homologue of PfMSP8, little is known about PvMSP8 as potential targets of protective immunity. Only one previous study showed the recognition of recombinant PvMSP8 with Plasmodium vivax-infected patients’ sera [14]. To propose PvMSP8 as vaccine candidate against blood stage P. vivax parasite, in this study, a high-level and long-lived immune response was observed against PvMSP8 in vivax malaria patients and a high immunogenicity was detected in rPvMSP8-immunized mice. Dominant epitopes were also mapped in the C terminus of PvMSP8 and its subcellular localization in the blood stage was shown to be at the FV.

Methods

Expression and purification of recombinant PvMSP8

Gene sequence of Pvmsp8 was obtained from the PlasmoDB website (http://​plasmodb.​org; accession no. PVX_097625). Protein domains were further predicted using the Simple Modular Architecture Research Tool (SMART) (http://​smart.​embl-heidelberg.​de/​) [15, 16]. Recombinant PvMSP8 (rPvMSP8) with a truncated signal peptide (SP) and GPI anchor was expressed and purified using a wheat-germ cell-free (WGCF) expression system [17]. Briefly, the specific primers: PvMSP8F, 5′-GGGCGGATATCTCGAGGGAAACGTTAGCCCACCC-3′; and PvMSP8R, 5′-GCGGTACCCGGGATCCTTAGCAGTATATTCCGTCTCCCTCA-3′ were used for DNA amplification. Then, the PvMSP8 DNA was cloned into the pEU-E01-His-TEV-MCS vector (CellFree Sciences, Matsuyama, Japan). The rPvMSP8 protein was expressed using a WGCF expression system and purified using a Ni–NTA agarose column (Qiagen, Hilden, Germany), as described elsewhere [17]. The production of rPvMSP8 protein was separated using 12% SDS–PAGE and detected via Western Blot using an anti-Penta-His antibody (Qiagen).

Immunization of mice with rPvMSP8

Female BALB/c mice, at 6–8 weeks of age (DaehanBiolink Co., Eumsung, Korea) were immunized with 20 μg of rPvMSP8 and phosphate buffered saline (PBS, pH 7.4) with complete Freund’s adjuvant (Sigma-Aldrich, St. Louis, MO, USA) using intraperitoneal route of administration. Three and 6 weeks after immunization, the equal volume of antigen with incomplete Freund’s adjuvant (Sigma-Aldrich) was boosted. Mouse blood samples were taken after the final booster injection, 2 weeks later. The antisera against PvMSP1 was also produced following the same procedure as PvMSP8 [17]. Animal experimental protocols were approved by the Kangwon National University, and the experiments were performed according to the Ethical Guidelines for Animal Experiments of Ehime University and Kangwon National University.

Indirect immunofluorescence assay (IFA)

The schizont-stage-rich parasites of P. vivax were collected from malaria patients in Thailand, as described previously [18]. Briefly, the slides were blocked with PBS containing 5% nonfat milk, incubated with rabbit anti-MSP1-19 (1:200 dilution) [18] and mouse anti-MSP8 (1:100 dilution) as primary antibodies, followed by incubation with Alexa Flour 546-conjugated goat anti-rabbit IgG or Alexa Flour 488-conjugated goat anti-mouse IgG antibody (Invitrogen, Carlsbad, CA, USA) and nuclear staining with DAPI (Invitrogen). Then, the slides with ProLong Gold antifade reagent (Invitrogen) were mounted. The parasites were observed under oil immersion using a confocal laser scanning FV200 microscope (Olympus, Tokyo, Japan).

Study sites and sample collections

One hundred and twelve serum samples from vivax-infected patients (mean age 25.5 years; range 18–42 years) from endemic countries and 80 samples from healthy individuals, assessed as being negative using microscopy, were collected in the Republic of Korea. The Myanmar samples (n = 56) were collected in 2012 from patients in Shwe Kyin area of Myanmar, and the Thai samples (n = 56) were collected from symptomatic, smear-positive patients from the Mae Sod district of western Thailand. For the 1-month follow-up study, 25 patients from the Shwe Kyin area were assessed at 7, 14, 21, and 28 days after treatment. The serum samples from healthy individuals with a vivax malaria history were collected from Chinese residents who had an episode of vivax malaria infection in malaria endemic areas of Anhui Province, China, in 2012, and who did not have a reinfection episode of vivax malaria in the preceding 5 (n = 30), 12 (n = 30), or 30 (n = 30) years (Table 1).
Table 1
Characteristics of study Plasmodium vivax samples from endemic areas of Korea, Myanmar, Thailand and China
Characteristic
Value
Acute vivax patients
1 month
Recovery subjects (China)
Healthy subjects (Korea)
Korea
Myanmar
Thailand
follow-up
5 year
12 year
30 year
Total (n)
112
56
56
25
30
30
30
80
Age (year)
 Mean (SD)
25.5 (9.50)
23.3 (8.50)
30.1 (12.46)
21.6 (7.85)
52.5 (22.52)
49.3 (16.47)
62 (13.0)
9.5 (3.30)
 Range
4–79
8–52
16–70
14–45
7–85
19–78
40–84
6–13
Parasitaemia (%)
 Mean (SD)
0.128 (0.11)
0.082 (0.11)
0.130 (0.12)
     
 Range
0.012–0.45
0.009–0.801
0.006–0.400
     
For the cellular immunity study, 10 mL of heparinized blood from P. vivax subjects (n = 15) who had recovered from P. vivax infection after 8–10 weeks were collected at malaria clinics in Tha Sae, Chumphon Province, which is located in the southern peninsular region of Thailand. Moreover, heparinized blood from healthy individuals (n = 15) who had no history of exposure to malaria were collected. The samples were used for peripheral blood mononuclear cell (PBMC) preparation.

Humoral immune responses and IgG isotyping

Amine (NH2-)-coated slides were prepared as previously described [19]. Serum samples from 112 cases of vivax malaria and 80 healthy individuals were used for humoral immune response analysis via well-type amine arrays. The chips were probed, scanned, and analysed as described previously [19]. The prevalence of an IgG isotype specific to PvMSP8 in the sera of 50 vivax patients and 10 healthy individuals was evaluated. Briefly, rPvMSP8 (50 μg/mL) was used for coating, followed by blocking and addition of human sera. Then these coated proteins were incubated with each IgG isotype for isotyping assay. The reaction was detected and analysed as described above.

Epitope mapping using a peptide array

An array of 22 peptides, 18-mer each (Additional file 1: Table S1) overlapping by nine amino acids with >90% purity and spanning the conserved C terminus of PvMSP8 Sal-1 sequences was custom synthesized, purified, and used for epitope mapping (Peptron Co., Ltd., Daejeon, Korea). In the process of coating step, 1 μL of peptides (10–40 μg/mL) in 200 mM 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide (EDC, Thermo Fisher Scientific Inc., Rockford, IL, USA) and 50 mM N-hydroxysuccinimide (NHS, Thermo Fisher Scientific Inc.) with coupling buffer were coated on amine-coated slide, respectively. After blocking, rPvMSP8-immunized mouse sera, pre-immunized mouse sera at 1:200 dilution, pooled sera from 10 high IgG titers of vivax-infected patients at 1:50 dilution, or 10 vivax-unexposed human sera at 1:50 dilution was added, respectively. Alexa Fluor 546 goat anti-mouse IgG (50 μg/mL, Invitrogen) or Alexa Fluor 546 goat anti-human IgG (10 μg/mL, Invitrogen) antibodies were used for detection of binding activity.

Naturally acquired cellular immunity in P. vivax-exposed individuals

Lymphocyte stimulation assay was carried out for measuring of cellular immunity in 15 recovered P. vivax subjects. Briefly, 2.5 × 105 PBMCs/well were stimulated with 10 μg/mL of rPvMSP8, 2% v/v of PHA, or complete RPMI 1640 medium only. After 96 h of stimulation, cytokine levels in the lymphocyte culture supernatant were measured using a BD OptEIA kit (BD Biosciences, San Jose, CA, USA). The phenotypes of T cells that responded to rPvMSP8 were analysed by intracellular cytokine staining after in vitro stimulation with 10 μg/mL of rPvMSP8, 20 ng/mL of PMA/ionomycin, or medium alone. Cells were stained with monoclonal antibodies (mAbs) against surface determinants of CD3, CD4, and CD8 and intracellular cytokine markers of anti-IFN-γ and anti-IL-10 antibodies, according to the manufacturer’s instructions.

Cellular immunity in PvMSP8-immunized mice

A splenocyte proliferation assay was performed using splenocytes removed from mice that had been immunized with rPvMSP8. The cells were stimulated with rPvMSP8 (2.5 μg/mL), Con A (5 μg/mL), LPS (10 μg/mL), or medium alone. Culture supernatants were collected after 72 h of incubation and assayed using a BD CBA Flex Set kit (BD Biosciences), according to the manufacturer’s instructions.

Statistical analysis

The correlation between the antibody reactivity of different concentrations of the recombinant proteins and duplicate spots of protein arrays was observed using GraphPad Prism software, version 5.0 (GraphPad, San Diego, CA, USA) and PASW Statistics 18.0 (SPSS Inc., Chicago, IL, USA). Sensitivity and specificity were measured by the percentage of patients who had a positive test result and the percentage of healthy individuals who had a negative test result, respectively. The significance of differences in mean fluorescence intensity (MFI) values between every two groups were performed with the Mann–Whitney U test.

Results

Structure, expression, purification, and western blot analysis of rPvMSP8

The predicted molecular weight of PvMSP8 (PVX_097625) is 54.7 kDa, with 487 amino acids containing a signal peptide, asparagine-rich region in N-terminal region and two EGF domains and GPI-motif in C-terminal region (Fig. 1a). The recombinant protein encoding the truncated PvMSP8 (ΔSP/GPI) was successfully expressed and purified as soluble form using a WGCF expression system. The purity of the purified recombinant PvMSP8 proteins were assessed, and the protein migrated as a single band of 55 kDa from SDS-PAGE analysis (Fig. 1b). The corresponding immunoblots probed with an anti-His tag antibody, anti-rPvMSP8 immune mouse serum, or a mixture of vivax patient sera revealed a similar and specific pattern of migration for PvMSP8 (Fig. 1c), whereas preimmune mouse sera and healthy individuals serum samples in non-endemic areas were used as negative controls; recombinant Pv41 did not react with either reagent.

Subcellular localization of PvMSP8

To determine PvMSP8 localization, IFA was carried out using anti-PvMSP8 and anti-PvMSP1-19 immune sera. In blood-stage parasites of P. vivax, PvMSP8 was localized to the outer circle of pigments associated with the FV (Fig. 2, arrow heads). A comparison with PvMSP1-19 in different intraerythrocytic-stage parasites indicated that PvMSP8 localized on pigments densely from the ring to late trophozoite stage (Fig. 2a, b) and on scattered pigments from mature to late-schizont-stage parasites (Fig. 2c, d).

Humoral immune response analysis of PvMSP8 in vivax malaria patients

Antibody responses against rPvMSP8 in serum samples from 112 P. vivax-infected patients and 80 healthy individuals were determined. Seropositivity of anti-rPvMSP8 antibodies was 73.2% in sensitivity and 96.2% in specificity (Additional file 1: Table S2). These sera from P. vivax-exposed individuals exhibited a significantly higher MFI than did those from malaria-naïve subjects (Fig. 3a, P < 0.0001). Furthermore, PvMSP8 produced similar IgG titers among isolates of vivax malaria patients from Korea, Myanmar, and Thailand, as well as from malaria-naïve individuals (Fig. 3b). The sera of P. vivax-exposed individuals had a significantly higher MFI than sera from malaria-naïve subjects.

Prevalence of IgG subclass response against PvMSP8 among malaria patients and immune mice

Protective antibodies against blood-stage parasites have been shown to belong to cytophilic classes; hence, the IgG subclass distribution was analysed. The prevalence of specific IgG subclasses was extremely high: >80% of the individuals had IgG antibodies that reacted with rPvMSP8 (Fig. 3c). Antibody responses against rPvMSP8 in vivax malaria patients were predominantly non-cytophilic IgG2 responses, which indicated that the efficacy of their protection may be poor against blood-stage parasites of P. vivax.
The isotypic distribution of anti-rPvMSP8 antibodies from immunized mice was analysed, assuming that the cytophilic IgG2a and IgG2b mouse isotypes would correspond to a Th1 response, whereas the non-cytophilic IgG1 and IgG3 would correspond to a Th2 response (Fig. 3d). Cytophilic antibodies (IgG2a and IgG2b) against PvMSP8 were the major components of the antibody response, especially IgG2b, observed in immunized mice. Non-cytophilic IgG antibodies were predominant in patients however cytophilic IgG antibodies were predominant in immune mice.

Longitudinal analysis of the IgG immune response against PvMSP8

The immune reactivity of the antibody response from serum samples during the 1-month follow-up after treatment in 25 patients with acute vivax malaria was assessed. Antibody response to PvMSP8, when detected, generally increased between the day of presentation, peaking on day 7, and was followed by a slight decrease from days 7 to 28. However, no significant difference was observed regarding IgG titers within 1 month (Fig. 4a; Additional file 1: Table S3).
The longevity of anti-PvMSP8 antibody responses was also analysed using long-term after exposure to vivax parasites (5-year recovery, 12-year recovery, and 30-year recovery samples) from China. A significantly higher IgG reactivity was observed in 5- and 12-year recovery sera, although the IgG titer was lower than that observed for acute infection. Moreover, in most cases, IgG antibody levels in individuals who infected malaria 30 years previously had decreased to baseline levels (Fig. 4b; Additional file 1: Table S3). These results indicate that IgG antibody responses against PvMSP8 are stably sustained in this population.

Mapping of the dominant epitopes of the PvMSP8 C terminus

A peptide was considered part of the potential epitope that there was significantly greater antibody reactivity for the antibodies in the vivax patient group compared with the vivax-unexposed human group, or greater reactivity for antibodies in rPvMSP8-immunized mice compared with preimmunized mice. Based on the reactivity of the pooled sera from vivax-infected patients, four peptides (nos. 2–5) located in the N terminus were highly reactive (Fig. 5a); based on those of immune mouse sera, two peptides (nos. 3 and 5) located in the central region and one peptide (no. 13) located in the C-terminal part were also highly reacted (Fig. 5b). Intriguingly, the two groups of sera were highly reacted with two peptides (nos. 3 and 5) located in the central region of PvMSP8. As a negative control, antibodies from preimmunized mice or unexposed human individuals that reacted with individual peptides were defined (all intensity values <1500).

Cellular immune response against rPvMSP8

The determination of IFN-γ or IL-10 cytokine levels after in vitro rPvMSP8 stimulation showed that IFN-γ was significantly upregulated (fourfold) in comparison to that observed in the absence of antigen stimulation (rPvMSP8 = 188.10 pg/mL, no stimulation = 27.68 pg/mL, P < 0.05; Fig. 6a). Moreover, the levels of IL-10 in rPvMSP8 cultures were significantly higher (twofold) in comparison to that observed in the absence of stimulation (rPvMSP8 = 119.17 pg/mL, no stimulation = 65.44 pg/mL, P < 0.05; Fig. 6a). The PBMCs from P. vivax subjects produced higher levels of IFN-γ and IL-10 in response to PHA and rPvMSP8 than did those of the healthy controls. The phenotyping of IFN-γ- or IL-10-producing cells that responded to the PvMSP8 antigen showed that CD8+ T cells were IFN-γ-producing cells; they produced IFN-γ at levels three times higher than those observed in un-stimulation (rPvMSP8 = 0.059%, no stimulation = 0.022%; Fig. 6b). In contrast, IFN-γ levels were low in CD4+ T cells (rPvMSP8 = 0.010%, no stimulation = 0.012%; Fig. 6b). Neither CD4+ nor CD8+ T cells produced IL-10 cytokine in response to the rPvMSP8 antigen (Fig. 6b). The cytokine responses from rPvMSP8-immunized mouse splenocytes stimulated by various concentrations of the rPvMSP8 protein, ConA (positive control), LPS (positive control), and culture medium alone (negative control) (Additional file 1: Figure S1A), respectively, showed predominant IFN-γ and TNF secretion compared with IL-2, IL-10, and IL-4 levels (Additional file 1: Figure S1B).

Discussion

PvMSP8 is a conserved GPI-anchored antigen that has been explored as potential antigens for vivax malaria vaccine development [4, 19]. As potent immune responses in both humans and mouse models were induced, PvMSP8 might be the target of actively induced B- and T-cell immune responses, which underscores its vaccine candidate potential.
In previous study about PvMSP8 [14], it could not defined exact subcellular localizations of blood-stage parasites with anti-PvMSP8 peptide antibody. However, in this study, the exact location of PvMSP8 was demonstrated that its FV subcellular localization surrounding pigments from intraerythrocytic-stage parasites were confirmed; therefore, PvMSP8 was referred to as an FV membrane-associated protein. In P. falciparum parasites, PfMSP8 has been shown to appear to co-localize on plasma membranes with PfMSP1, and only on the surface of early-ring-stage parasites, and its C terminus was found in the FV of infected erythrocytes in schizont-stage parasites [8]. In P. yoelii parasites, PyMSP8 was detected on ring-stage parasites and expressed together with PyMSP1 on the surface of each merozoite of mature schizont-stage parasites. In addition, PyMSP8 was highly expressed on the FV of trophozoite- and schizont-stage parasites [20]. However, the IFA images, which were produced using SP- and GPI-truncated PvMSP8 antibodies, showed that PvMSP8 (Fig. 2) was mainly expressed around the FV. Although weak reactivity of PvMSP8 was detected at the merozoite surface in schizont-stage parasites, the localization of PvMSP8 was obviously different from that of PvMSP1, which suggests different roles for the two proteins.
In previous studies, non-cytophilic classes (IgG2) also predominated among the anti-malarial antibodies developed by unprotected subjects, whereas cytophilic subclasses (IgG1 and mainly IgG3) were the most abundant isotypes produced by malaria patients, who are protected from malarial parasites [21, 22]. Conversely, a recent study showed an association between parasite levels and antigen-specific IgG2 and resistance to P. falciparum infection, suggesting that IgG2 plays a noncytophilic isotype role and contributes to parasite clearance [23]. In addition, purified IgG2 antibodies have been shown to block the ability to inhibit parasitic growth in vitro study. IgG2 responses were also related with the higher risk for severity of malarial infection in Kenyan children [24]. In this study, the high IgG2 response against PvMSP8 antigen was found in vivax patients, suggesting that PvMSP8 induce antibody production in patients, which may be associated with resistance to vivax malaria.
Here, the authors also observed that long-term maintenance of IgG antibodies against PvMSP8 was detected in individuals from Anhui Province, China (where malaria is not endemic recently, Fig. 4). Because the half-life of the human IgG molecule is around 21 days [25], the long-term maintenance of IgG antibodies may contribute to ongoing secretion of antibodies from plasma cells or to memory B cell differentiation in response to inflammatory stimuli. To investigate the actual longevity of antibodies against the PvMSP8 protein, a future longitudinal field study is required.
For vaccine consideration, conserved regions should include immunodominant protective T- and/or B-cell epitopes [26]. It appears that the immunogenic peptides involved in T-cell response are located at the C terminus of PfMSP8 [6]. To examine further B-cell epitopes, the antigenicity of each peptide was also compared. Both vivax patient serum and PvMSP8-immunized mouse serum antibodies were highly reacted with two peptides located outside the C-terminal EGF-like domain (Fig. 5). These data suggest that rPvMSP8 is highly immunogenic for both B and T cells, and that the C terminus appears to contain the dominant B-cell epitopes of PvMSP8. However, it remained to be further studies of potential vaccine candidates from B- and T-cell epitopes of PvMSP8.
As cytokine play an important role in cell-mediated immunity, the study showed that PvMSP8 stimulated PBMCs to produce the IFN-γ and IL-10 cytokines (with the IFN-γ-type response being more pronounced), and that CD8+ T cells were a major type of IFN-γ-producing cells (Fig. 6a, b). This indicates the immunogenicity of PvMSP8 in the induction of a cellular response against P. vivax parasite, which has been shown to be associated with protection against P. falciparum among volunteers undergoing experimentally induced infection [27], as well as in naturally exposed human populations [28, 29]. CD8+ T cells exhibited a stronger participation in the response to PvMSP8 than CD4+ T cells, whereas other IFN-γ-producing cells, including γδ T cells, NKT cells, and NK cells, should be considered in future studies. The regulation of the immune response in PvMSP8-stimulated PBMC cultures may be involved in IL-10 secretion, as it occurred significantly in the supernatant of 96 h cultures, whereas no evidence of immediate production of IL-10 by CD4+ and CD8+ effector cells was found upon short-term in vitro PvMSP8 stimulation.

Conclusions

The study results demonstrated the presence of PvMSP8-antigen-induced humoral and cellular immune responses in P. vivax infection and represent an important advance in the understanding of blood-stage immunity to P. vivax, at least in part. Intriguingly, the manner via which a potent immune response, such as the one shown here, can be induced remains to be studied, together with the FV localization of PvMSP8. To confirm the protective ability of PvMSP8 antibodies, functional assays, such as a short-time growth inhibition assay, need to be developed in the future.

Authors’ contributions

YC, PC, SC, J-HH and E-TH conceived and designed the experiments; YC, WB, PC, SC, J-HH and E-TH performed the experiments and analysed the data; YC, BW, SC, PC, TT, E-TH wrote the paper; FL, JC, MHN, WSP, S-HH, K-SH, CSL and SJ support technical advice and materials for this study and review manuscript critically. All authors read and approved the final manuscript.

Competing interests

The authors declare that they have no competing interests.
Ethical clearance was approved by Ethical Committee of the Kangwon National University Hospital, Republic of Korea, the Department of Medical Research, Myanmar, the Mahidol University, Thailand and the Jiangsu Institute of Parasitic Diseases, China. Written informed consents were taken in all of the participants.

Funding

This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIP) (NRF-2014R1A2A1A11052079) and by Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Science, ICT & Future Planning (2015R1A4A1038666).

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Black CG, Wu T, Wang L, Hibbs AR, Coppel RL. Merozoite surface protein 8 of Plasmodium falciparum contains two epidermal growth factor-like domains. Mol Biochem Parasitol. 2001;114:217–26.CrossRefPubMed Black CG, Wu T, Wang L, Hibbs AR, Coppel RL. Merozoite surface protein 8 of Plasmodium falciparum contains two epidermal growth factor-like domains. Mol Biochem Parasitol. 2001;114:217–26.CrossRefPubMed
2.
Zurück zum Zitat Burns JM Jr, Belk CC, Dunn PD. A protective glycosylphosphatidylinositol-anchored membrane protein of Plasmodium yoelii trophozoites and merozoites contains two epidermal growth factor-like domains. Infect Immun. 2000;68:6189–95.CrossRefPubMedPubMedCentral Burns JM Jr, Belk CC, Dunn PD. A protective glycosylphosphatidylinositol-anchored membrane protein of Plasmodium yoelii trophozoites and merozoites contains two epidermal growth factor-like domains. Infect Immun. 2000;68:6189–95.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Carlton JM, Adams JH, Silva JC, Bidwell SL, Lorenzi H, Caler E, et al. Comparative genomics of the neglected human malaria parasite Plasmodium vivax. Nature. 2008;455:757–63.CrossRefPubMedPubMedCentral Carlton JM, Adams JH, Silva JC, Bidwell SL, Lorenzi H, Caler E, et al. Comparative genomics of the neglected human malaria parasite Plasmodium vivax. Nature. 2008;455:757–63.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Pacheco MA, Elango AP, Rahman AA, Fisher D, Collins WE, Barnwell JW, et al. Evidence of purifying selection on merozoite surface protein 8 (MSP8) and 10 (MSP10) in Plasmodium spp. Infect Genet Evol. 2012;12:978–86.CrossRefPubMedPubMedCentral Pacheco MA, Elango AP, Rahman AA, Fisher D, Collins WE, Barnwell JW, et al. Evidence of purifying selection on merozoite surface protein 8 (MSP8) and 10 (MSP10) in Plasmodium spp. Infect Genet Evol. 2012;12:978–86.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Alaro JR, Lynch MM, Burns JM Jr. Protective immune responses elicited by immunization with a chimeric blood-stage malaria vaccine persist but are not boosted by Plasmodium yoelii challenge infection. Vaccine. 2010;28:6876–84.CrossRefPubMedPubMedCentral Alaro JR, Lynch MM, Burns JM Jr. Protective immune responses elicited by immunization with a chimeric blood-stage malaria vaccine persist but are not boosted by Plasmodium yoelii challenge infection. Vaccine. 2010;28:6876–84.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Alaro JR, Angov E, Lopez AM, Zhou H, Long CA, Burns JM Jr. Evaluation of the immunogenicity and vaccine potential of recombinant Plasmodium falciparum merozoite surface protein 8. Infect Immun. 2012;80:2473–84.CrossRefPubMedPubMedCentral Alaro JR, Angov E, Lopez AM, Zhou H, Long CA, Burns JM Jr. Evaluation of the immunogenicity and vaccine potential of recombinant Plasmodium falciparum merozoite surface protein 8. Infect Immun. 2012;80:2473–84.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Black CG, Wu T, Wang L, Topolska AE, Coppel RL. MSP8 is a non-essential merozoite surface protein in Plasmodium falciparum. Mol Biochem Parasitol. 2005;144:27–35.CrossRefPubMed Black CG, Wu T, Wang L, Topolska AE, Coppel RL. MSP8 is a non-essential merozoite surface protein in Plasmodium falciparum. Mol Biochem Parasitol. 2005;144:27–35.CrossRefPubMed
8.
Zurück zum Zitat Drew DR, Sanders PR, Crabb BS. Plasmodium falciparum merozoite surface protein 8 is a ring-stage membrane protein that localizes to the parasitophorous vacuole of infected erythrocytes. Infect Immun. 2005;73:3912–22.CrossRefPubMedPubMedCentral Drew DR, Sanders PR, Crabb BS. Plasmodium falciparum merozoite surface protein 8 is a ring-stage membrane protein that localizes to the parasitophorous vacuole of infected erythrocytes. Infect Immun. 2005;73:3912–22.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Cowman AF, Crabb BS. Invasion of red blood cells by malaria parasites. Cell. 2006;124:755–66.CrossRefPubMed Cowman AF, Crabb BS. Invasion of red blood cells by malaria parasites. Cell. 2006;124:755–66.CrossRefPubMed
10.
Zurück zum Zitat Francis SE, Sullivan DJ Jr, Goldberg DE. Hemoglobin metabolism in the malaria parasite Plasmodium falciparum. Annu Rev Microbiol. 1997;51:97–123.CrossRefPubMed Francis SE, Sullivan DJ Jr, Goldberg DE. Hemoglobin metabolism in the malaria parasite Plasmodium falciparum. Annu Rev Microbiol. 1997;51:97–123.CrossRefPubMed
11.
Zurück zum Zitat Goldberg DE, Slater AF, Cerami A, Henderson GB. Hemoglobin degradation in the malaria parasite Plasmodium falciparum: an ordered process in a unique organelle. Proc Natl Acad Sci USA. 1990;87:2931–5.CrossRefPubMedPubMedCentral Goldberg DE, Slater AF, Cerami A, Henderson GB. Hemoglobin degradation in the malaria parasite Plasmodium falciparum: an ordered process in a unique organelle. Proc Natl Acad Sci USA. 1990;87:2931–5.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Dluzewski AR, Ling IT, Hopkins JM, Grainger M, Margos G, Mitchell GH, et al. Formation of the food vacuole in Plasmodium falciparum: a potential role for the 19 kDa fragment of merozoite surface protein 1 (MSP1(19)). PLoS ONE. 2008;3:e3085.CrossRefPubMedPubMedCentral Dluzewski AR, Ling IT, Hopkins JM, Grainger M, Margos G, Mitchell GH, et al. Formation of the food vacuole in Plasmodium falciparum: a potential role for the 19 kDa fragment of merozoite surface protein 1 (MSP1(19)). PLoS ONE. 2008;3:e3085.CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Moss DK, Remarque EJ, Faber BW, Cavanagh DR, Arnot DE, Thomas AW, et al. Plasmodium falciparum 19-kilodalton merozoite surface protein 1 (MSP1)-specific antibodies that interfere with parasite growth in vitro can inhibit MSP1 processing, merozoite invasion, and intracellular parasite development. Infect Immun. 2012;80:1280–7.CrossRefPubMedPubMedCentral Moss DK, Remarque EJ, Faber BW, Cavanagh DR, Arnot DE, Thomas AW, et al. Plasmodium falciparum 19-kilodalton merozoite surface protein 1 (MSP1)-specific antibodies that interfere with parasite growth in vitro can inhibit MSP1 processing, merozoite invasion, and intracellular parasite development. Infect Immun. 2012;80:1280–7.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Perez-Leal O, Sierra AY, Barrero CA, Moncada C, Martinez P, Cortes J, et al. Plasmodium vivax merozoite surface protein 8 cloning, expression, and characterisation. Biochem Biophys Res Commun. 2004;324:1393–9.CrossRefPubMed Perez-Leal O, Sierra AY, Barrero CA, Moncada C, Martinez P, Cortes J, et al. Plasmodium vivax merozoite surface protein 8 cloning, expression, and characterisation. Biochem Biophys Res Commun. 2004;324:1393–9.CrossRefPubMed
15.
Zurück zum Zitat Schultz J, Milpetz F, Bork P, Ponting CP. SMART, a simple modular architecture research tool: identification of signaling domains. Proc Natl Acad Sci USA. 1998;95:5857–64.CrossRefPubMedPubMedCentral Schultz J, Milpetz F, Bork P, Ponting CP. SMART, a simple modular architecture research tool: identification of signaling domains. Proc Natl Acad Sci USA. 1998;95:5857–64.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Letunic I, Doerks T, Bork P. SMART: recent updates, new developments and status in 2015. Nucleic Acids Res. 2015;43:D257–60.CrossRefPubMed Letunic I, Doerks T, Bork P. SMART: recent updates, new developments and status in 2015. Nucleic Acids Res. 2015;43:D257–60.CrossRefPubMed
17.
Zurück zum Zitat Tsuboi T, Takeo S, Sawasaki T, Torii M, Endo Y. An efficient approach to the production of vaccines against the malaria parasite. Methods Mol Biol. 2010;607:73–83.CrossRefPubMed Tsuboi T, Takeo S, Sawasaki T, Torii M, Endo Y. An efficient approach to the production of vaccines against the malaria parasite. Methods Mol Biol. 2010;607:73–83.CrossRefPubMed
18.
Zurück zum Zitat Cheng Y, Wang Y, Ito D, Kong DH, Ha KS, Chen JH, et al. The Plasmodium vivax merozoite surface protein 1 paralog is a novel erythrocyte-binding ligand of P. vivax. Infect Immun. 2013;81:1585–95.CrossRefPubMedPubMedCentral Cheng Y, Wang Y, Ito D, Kong DH, Ha KS, Chen JH, et al. The Plasmodium vivax merozoite surface protein 1 paralog is a novel erythrocyte-binding ligand of P. vivax. Infect Immun. 2013;81:1585–95.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Chen JH, Jung JW, Wang Y, Ha KS, Lu F, Lim CS, et al. Immunoproteomics profiling of blood stage Plasmodium vivax infection by high-throughput screening assays. J Proteom Res. 2010;9:6479–89.CrossRef Chen JH, Jung JW, Wang Y, Ha KS, Lu F, Lim CS, et al. Immunoproteomics profiling of blood stage Plasmodium vivax infection by high-throughput screening assays. J Proteom Res. 2010;9:6479–89.CrossRef
20.
Zurück zum Zitat Shi Q, Cernetich-Ott A, Lynch MM, Burns JM Jr. Expression, localization, and erythrocyte binding activity of Plasmodium yoelii merozoite surface protein-8. Mol Biochem Parasitol. 2006;149:231–41.CrossRefPubMed Shi Q, Cernetich-Ott A, Lynch MM, Burns JM Jr. Expression, localization, and erythrocyte binding activity of Plasmodium yoelii merozoite surface protein-8. Mol Biochem Parasitol. 2006;149:231–41.CrossRefPubMed
21.
Zurück zum Zitat Oeuvray C, Bouharoun-Tayoun H, Gras-Masse H, Bottius E, Kaidoh T, Aikawa M, et al. Merozoite surface protein-3: a malaria protein inducing antibodies that promote Plasmodium falciparum killing by cooperation with blood monocytes. Blood. 1994;84:1594–602.PubMed Oeuvray C, Bouharoun-Tayoun H, Gras-Masse H, Bottius E, Kaidoh T, Aikawa M, et al. Merozoite surface protein-3: a malaria protein inducing antibodies that promote Plasmodium falciparum killing by cooperation with blood monocytes. Blood. 1994;84:1594–602.PubMed
22.
Zurück zum Zitat Branch OH, Oloo AJ, Nahlen BL, Kaslow D, Lal AA. Anti-merozoite surface protein-1 19-kDa IgG in mother-infant pairs naturally exposed to Plasmodium falciparum: subclass analysis with age, exposure to asexual parasitemia, and protection against malaria. V. The Asembo Bay Cohort Project. J Infect Dis. 2000;181:1746–52.CrossRefPubMed Branch OH, Oloo AJ, Nahlen BL, Kaslow D, Lal AA. Anti-merozoite surface protein-1 19-kDa IgG in mother-infant pairs naturally exposed to Plasmodium falciparum: subclass analysis with age, exposure to asexual parasitemia, and protection against malaria. V. The Asembo Bay Cohort Project. J Infect Dis. 2000;181:1746–52.CrossRefPubMed
23.
Zurück zum Zitat Aucan C, Traore Y, Tall F, Nacro B, Traore-Leroux T, Fumoux F, et al. High immunoglobulin G2 (IgG2) and low IgG4 levels are associated with human resistance to Plasmodium falciparum malaria. Infect Immun. 2000;68:1252–8.CrossRefPubMedPubMedCentral Aucan C, Traore Y, Tall F, Nacro B, Traore-Leroux T, Fumoux F, et al. High immunoglobulin G2 (IgG2) and low IgG4 levels are associated with human resistance to Plasmodium falciparum malaria. Infect Immun. 2000;68:1252–8.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Ndungu FM, Bull PC, Ross A, Lowe BS, Kabiru E, Marsh K. Naturally acquired immunoglobulin (Ig)G subclass antibodies to crude asexual Plasmodium falciparum lysates: evidence for association with protection for IgG1 and disease for IgG2. Parasite Immunol. 2002;24:77–82.CrossRefPubMed Ndungu FM, Bull PC, Ross A, Lowe BS, Kabiru E, Marsh K. Naturally acquired immunoglobulin (Ig)G subclass antibodies to crude asexual Plasmodium falciparum lysates: evidence for association with protection for IgG1 and disease for IgG2. Parasite Immunol. 2002;24:77–82.CrossRefPubMed
25.
Zurück zum Zitat Hopkins RJ, Kramer WG, Blackwelder WC, Ashtekar M, Hague L, Winker-La Roche SD, et al. Safety and pharmacokinetic evaluation of intravenous vaccinia immune globulin in healthy volunteers. Clin Infect Dis. 2004;39:759–66.CrossRefPubMed Hopkins RJ, Kramer WG, Blackwelder WC, Ashtekar M, Hague L, Winker-La Roche SD, et al. Safety and pharmacokinetic evaluation of intravenous vaccinia immune globulin in healthy volunteers. Clin Infect Dis. 2004;39:759–66.CrossRefPubMed
26.
Zurück zum Zitat Dame JB, Williams JL, McCutchan TF, Weber JL, Wirtz RA, Hockmeyer WT, et al. Structure of the gene encoding the immunodominant surface antigen on the sporozoite of the human malaria parasite Plasmodium falciparum. Science. 1984;225:593–9.CrossRefPubMed Dame JB, Williams JL, McCutchan TF, Weber JL, Wirtz RA, Hockmeyer WT, et al. Structure of the gene encoding the immunodominant surface antigen on the sporozoite of the human malaria parasite Plasmodium falciparum. Science. 1984;225:593–9.CrossRefPubMed
27.
Zurück zum Zitat Teirlinck AC, McCall MB, Roestenberg M, Scholzen A, Woestenenk R, de Mast Q, et al. Longevity and composition of cellular immune responses following experimental Plasmodium falciparum malaria infection in humans. PLoS Pathog. 2011;7:e1002389.CrossRefPubMedPubMedCentral Teirlinck AC, McCall MB, Roestenberg M, Scholzen A, Woestenenk R, de Mast Q, et al. Longevity and composition of cellular immune responses following experimental Plasmodium falciparum malaria infection in humans. PLoS Pathog. 2011;7:e1002389.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Dodoo D, Omer FM, Todd J, Akanmori BD, Koram KA, Riley EM. Absolute levels and ratios of proinflammatory and anti-inflammatory cytokine production in vitro predict clinical immunity to Plasmodium falciparum malaria. J Infect Dis. 2002;185:971–9.CrossRefPubMed Dodoo D, Omer FM, Todd J, Akanmori BD, Koram KA, Riley EM. Absolute levels and ratios of proinflammatory and anti-inflammatory cytokine production in vitro predict clinical immunity to Plasmodium falciparum malaria. J Infect Dis. 2002;185:971–9.CrossRefPubMed
29.
Zurück zum Zitat D’Ombrain MC, Robinson LJ, Stanisic DI, Taraika J, Bernard N, Michon P, et al. Association of early interferon-gamma production with immunity to clinical malaria: a longitudinal study among Papua New Guinean children. Clin Infect Dis. 2008;47:1380–7.CrossRefPubMed D’Ombrain MC, Robinson LJ, Stanisic DI, Taraika J, Bernard N, Michon P, et al. Association of early interferon-gamma production with immunity to clinical malaria: a longitudinal study among Papua New Guinean children. Clin Infect Dis. 2008;47:1380–7.CrossRefPubMed
Metadaten
Titel
Naturally acquired humoral and cellular immune responses to Plasmodium vivax merozoite surface protein 8 in patients with P. vivax infection
verfasst von
Yang Cheng
Bo Wang
Siriruk Changrob
Jin-Hee Han
Jetsumon Sattabongkot
Kwon-Soo Ha
Patchanee Chootong
Feng Lu
Jun Cao
Myat Htut Nyunt
Won Sun Park
Seok-Ho Hong
Chae Seung Lim
Takafumi Tsuboi
Eun-Taek Han
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
Malaria Journal / Ausgabe 1/2017
Elektronische ISSN: 1475-2875
DOI
https://doi.org/10.1186/s12936-017-1837-5

Weitere Artikel der Ausgabe 1/2017

Malaria Journal 1/2017 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.