Skip to main content
Infection and Immunity logoLink to Infection and Immunity
. 2000 Oct;68(10):5839–5845. doi: 10.1128/iai.68.10.5839-5845.2000

Genetic Immunization of BALB/c mice with a Plasmid Bearing the Gene Coding for a Hybrid Merozoite Surface Protein 1-Hepatitis B Virus Surface Protein Fusion Protects Mice against Lethal Plasmodium chabaudi chabaudi PC1 Infection

Gerhard Wunderlich 1,*, Ivan C Moura 1, Hernando A del Portillo 1
Editor: W A Petri Jr1
PMCID: PMC101545  PMID: 10992493

Abstract

The genetic immunization of rodents with a plasmid coding for a Plasmodium chabaudi merozoite surface protein 1 (C terminus)-hepatitis B virus surface fusion protein (pPcMSP119-HBs) provided protection of mice against subsequent lethal challenge with P. chabaudi chabaudi PC1-infected red blood cells. The percentage of survivor mice was higher in DNA-immunized mice than in animals immunized with a recombinant rPcMSP119– glutathione S-transferase fusion protein administered in Freund adjuvant. In all mice immunized with the pPcMSP119-HBs, a Th1-specific response, including the production of anti-MSP119-specific immunoglobulins predominantly of the immunoglobulin G2a subtype and reacting almost exclusively against discontinuous epitopes, was elicited. The coinjection of Th1-type cytokine-expressing plasmids (gamma interferon, interleukin-2, and granulocyte-macrophage colony-stimulating factor) mostly abolished protection and boosting of MSP119-specific antibodies. The inclusion of a lymph node-targeting signal did not significantly increase protection. These data provide further evidence that MSP119-HBs DNA constructs might be useful as components of a genetic vaccine against the asexual blood stages of Plasmodium.


The merozoite surface protein 1 (MSP1) of Plasmodium spp. is synthesized during schizogony as a large 190- to 250-kDa polypeptide which is later processed into four of the major merozoite surface proteins. Upon release of the merozoite, the N-terminal parts of the proteins are subsequently cleaved off so that only a short 19-kDa C-terminal fragment (MSP119) remains anchored on the mature merozoite's surface at the time of red blood cell invasion (3). While extensive sequence heterogeneity has been described for most parts of the protein, the C-terminal portion shows a highly analogous structure, including two epidermal growth factor-like domains in all Plasmodium species (13).

Therefore, MSP119 is considered an important candidate in developing a subunit vaccine against the asexual blood stages of malaria. Thus, in rodent models, immunization with affinity-purified and recombinant MSP1 from Plasmodium yoelii yielded protection, as did the passive transfer of monoclonal antibodies and immune serum against PyMSP-1 (9). In monkeys, immunization trials with proteic MSP119 also elicited protection and determined that the conformation of the recombinant protein was crucial for protection. Indeed, while the baculovirus-produced MSP119 protected monkeys against challenge with Plasmodium falciparum (8), the Escherichia coli-derived MSP119 fused to glutathione S-transferase (GST) did not (6). More recent studies in Macaca sinica with Plasmodium cynomolgi, also revealed that immunization of monkeys with baculovirus-derived recombinant P. cynomolgi-MSP119 or MSP142 led to a high degree of protection against infection, which was mostly antibody dependent (20).

An alternative to the use of recombinant proteins and toxic adjuvants is immunization with genetic vaccines (27). For MSP1, it was recently shown that mice immunized with MSP1-coding plasmids showed decreased peak parasitemias after challenge with a nonlethal P. yoelii strain (2). Moreover, genetic vaccines containing CpG motifs (15, 22) produce in mice a Th1-like response considered essential for the control of a primary blood stage infection (23). Recently, genetic immunizations, including multiple genes expressed during different stages of the human malarias caused by P. falciparum and P. vivax, were tested (reviewed in reference 12).

In this study, we tested the protective potential of a plasmid bearing the gene coding for a fusion protein of MSP119 of P. chabaudi and the small hepatitis B virus surface protein (HBs). A similar construct for the P. vivax MSP119 had been previously shown to be highly antigenic (10) and to form hybrid viral particles (28). Moreover, since an effective immune response against blood stage infection of mice with P. chabaudi has a Th1 profile (24), we also coinjected plasmid vectors coding for Th1-associated cytokines, namely, murine interleukin-2 (IL-2), gamma interferon (IFN-γ), and granulocyte-macrophage colony-stimulating factor (GM-CSF). Additionally, we tested a plasmid coding for a selectin-MSP119-HBs hybrid which was supposed to target the MSP119-HBs hybrid to lymph nodes (4) and therefore enhance the immune reaction.

MATERIALS AND METHODS

Construction of plasmid vectors used in genetic immunization trials.

A fragment coding for the small HBs was excised from the plasmid pSV33M* by restriction with BamHI and EcoRV and inserted into pVXORF1, resulting in pVXORF-S (10). The MSP119 coding fragment was amplified from genomic P. chabaudi chabaudi PC1 DNA by standard PCR procedures. Complete sequencing of the fragment revealed that the sequence was identical to the sequence of P. chabaudi chabaudi strain CB (GenBank accession no. L22984). The EcoRI/BglII-restricted fragment was cloned into pVXORF-S via BamHI and EcoRI sites. A plasmid solely coding for MSP119 was cloned by insertion of an EcoRI-restricted and 5′-phosphorylated PCR fragment using the oligonucleotides described in Table 1 in pVXORF1 via EcoRI and EcoRV sites. In parallel, the MSP119 coding fragment was also inserted in the vector pGEX3X (Pharmacia, Uppsala, Sweden). Genes coding for IFN-γ, IL-2, and the ectodomain of l-selectin were cloned by standard reverse transcription-PCR from total RNA of 108 concanavalin A-stimulated BALB/c mouse splenocytes; the primers are listed in Table 1. IL-2 and IFN-γ fragments were subcloned via EcoRI/BamHI digestion into pVXVR, a pVXORF1 variant lacking its tissue plasminogen activator secretion signal. The l-selectin fragment was subcloned via BamHI/BglII into pVXORF1- and pPcMSP119-HBs, resulting in pSel and pSel-PcMSP119-HBs, respectively. A functional plasmid coding for GM-CSF (29), termed pGMCSF, was kindly provided by M. M. Rodrigues (Escola Paulista de Medicina, São Paulo, Brazil). All plasmid constructs were checked for the correctness of their inserts by manual dideoxy sequencing (21). Plasmids were purified using the Qiagen Mega-Prep columns (Qiagen, Hilden, Germany), according to the manufacturer's instructions.

TABLE 1.

Oligonucleotides used in PCRs

Amplified gene Sequence
Sense Antisense
MSP119 5′-CGAATTCATGGATTTATTAGGTATAGGTTC-3′ 5′-CCAGATCTGAAGGACAAGCTTAGGAAG-3′
IL-2 5′-CCGGATCCATGTACAGCATGCAGC-3′ 5′-CGAATTCTTATTGAGGGCTTGTTG-3′
IFN-γ 5′-CCGATCCATGAACGCTACACACTG-3′ 5′-CGAATTCTCAGCTCCTTTTCC-3′
l-Selectin 5′-CAGGATCCTGGACTTACCATTATTCTG-3′ 5′-CCAGATCTGTTGTAGTCACCTTCTTTG-3′

Expression of recombinant constructs in COS7 cells.

Plasmids were transfected in COS7 cells by the standard DEAE-dextran method, and metabolically labeled proteins were immunoprecipitated as described elsewhere (5). Briefly, at 48 h posttransfection, 50% confluent COS7 cells in 3-cm-diameter dishes were pulsed with 80 μCi of Tran35S-label (ICN, Costa Mesa, Calif.) for 15 min. After a 24-h chase period, culture supernatants were removed and immunoprecipitated for 2 h at room temperature with protein A-Sepharose-bound polyclonal goat anti-HBs (Dako, Hamburg, Germany) or immunoglobulins derived from the acute sera of P. chabaudi-infected trial mice. Proteins were resolved by sodium dodecyl sulfate-polyacrylamide gel electrophoresis on a standard 12% polyacrylamide gel, fixed, stained with fluorographic reagent (Amplify; Amersham), dried, and autoradiographed.

Production of recombinant P. chabaudi MSP119-GST and total schizont proteins.

Expression and purification of rPcMSP119-GST or rGST from pGEX3X-PcMSP119 or pGEX3X-transformed E. coli DH5α was done as described previously (17). Before use, the recombinant proteins were dialyzed extensively against phosphate-buffered saline (PBS). P. chabaudi proteins were obtained by lysis of IRBC containing schizont stage parasites with PBS–0.1% saponin for 5 min at room temperature. After three washing steps (ice-cold PBS–0.05% saponin), parasites were lysed in PBS–0.5% Triton X-100 for 10 min on ice in the presence of 1 mM phenylmethylsulfonyl fluoride. After pelleting of the insoluble debris, the supernatant was stored in liquid N2 until use.

ELISA, Western blot, and isotyping.

Proteins were analyzed by enzyme-linked immunosorbent assay (ELISA) and Western blot assays as described elsewhere (17). Isotyping of immunoglobulin G (IgG) was performed with sera at dilutions which resulted in an optical density at 450 nm (OD450) of approximately 0.5 and using the Bio-Rad antigen-dependent isotyping kit (Bio-Rad, Hercules, Calif.) according to the manufacturer's instructions.

Immunization and challenge regimens.

All animal experiments were conducted in accordance with local rules for animal housing. Female 6-week-old BALB/c mice received, at 3-week intervals, three intramuscular injections of 50 μl of plasmid solution (1 μg/μl) in PBS in each tibialis anterior muscle using a Becton Dickinson 0.5-ml insulin syringe armed with a 31.5-gauge needle. When animals were immunized with recombinant proteins, 50 μg per dose of purified PcMSP119-GST or GST emulsified in Freund complete (first dose) or Freund incomplete adjuvant (second and third doses) were injected intraperitoneally (i.p.). P. chabaudi chabaudi PC1 parasites were maintained by weekly reinfection using 105 infected red blood cells (IRBC). The challenge of animals was performed by i.p. injection of 2.5 × 104 P. chabaudi chabaudi PC1 IRBC, obtained from one infected animal with 20 to 30% parasitemia. Under these challenge conditions, all naive mice died by days 8 to 10. Serum samples were taken 3 days before challenge and from day 5 on after challenge by tail bleeding. Parasitemias were determined by counting 1,000 red blood cells per Giemsa-stained blood smear and day.

Statistical analyses.

A two-tailed Student's t test was used to evaluate the significance of differences between antibody titers determined on the day of infection for all mice that were still alive. The significance of the proportions of survivors in different groups of mice was calculated by applying the χ2 method, considering Fisher's exact P values and treating all negative-group mice as the expected outcome group (n = 72).

RESULTS

N-terminal fusion of PcMSP119 to HBs results in HBsAg-like particles.

In the first experiment, we sought to determine whether a fusion protein containing PcMSP119 and HBs was released from transfected COS7 cells. To do so, immunoprecipitations from supernatants of transfected COS7 cells with plasmids coding for HBs or PcMSP119-HBs were performed using either anti-HBs or anti-PcMSP119 specific antibodies. As predicted, anti-HBs antibodies immunoprecipitated wild-type HBs (24-kDa and glycosylated 27-kDa HBs-protein) from the supernatant (Fig. 1A, lane 2). Using this same antibody, cells transfected with pPcMSP119-HBs secreted a hybrid protein with an expected monomer size of ∼45 kDa (lane 4). In contrast, anti-PcMSP119 antibodies from a reconvalescent mouse immunoprecipitated only the hybrid protein (lanes 10), whereas they failed to precipitate wild-type HBs (lane 8). Significantly, when the supernatant fraction of pPcMSP119-HBs-transfected COS7 cells was fractionated on 10 to 40% CsCl gradients, fractions with 1.22 g/ml contained particle-like structures resembling those described earlier for the P. vivax MSP119-HBs particle (Fig. 1B and reference 28). Taken together, these data strongly indicate that PcMSP119-HBs is secreted from COS7 transfected cells in the form of a hybrid viral particle which exposes HBs and PcMSP119 epitopes on its surface.

FIG. 1.

FIG. 1

PcMSP119-HBs is secreted from transfected COS7 cells. (A) COS7 cells were transfected with plasmids coding for HBs (lanes 1, 2, 7, and 8) or PcMSP119-HBs (lanes 3, 4, 9, and 10) or mock transfected with the expression vector pVXORF1 (lanes 5 and 6). After pulse-labeling and a 24-h chase, lysates (L) and supernatants (S) of cells were immunoprecipitated with antiHBs (diluted 1:500) or hyperimmune serum of a reconvalescent mouse (diluted 1:2,000). (B) Particle-like structures containing MSP119-HBs proteins after fractionation of transfected COS7 cell supernatants by CsCl gradient, resolved by electron microscopy. Material from the 1.21-g/ml fraction is shown at ×100,000 magnification.

Immunization of mice with pPcMSP119-HBs or pSel-PcMSP119-HBs but not pPcMSP119 results in the generation of antibodies against PcMSP119.

We then tested which of our MSP119 constructs resulted in the highest antibody titers against PcMSP119 (Table 2). Injection of pPcMSP119-HBs results in the seroconversion of most mice after the first boost and antibody titers of 1:4,000 to 1:25,600 after the second boost, whereas injection of pPcMSP119 led to very low anti-MSP119 antibody titers, which remained near the detection limit (endpoint dilution titer of <1:200). The injection of pSel-PcMSP119-HBs in mice caused the production of anti- PcMSP119 specific antibodies after the first boost, but the titers were never higher than those obtained upon genetic immunization with pPcMSP119-HBs. When an IL-2 coding plasmid was coinjected with pPcMSP119-HBs, seroconversion was detected after the first boost, and the final titers were as high as 1:51,600. In contrast, coinjection of plasmids coding for IFN-γ or GM-CSF with PcMSP119-HBs delayed seroconversion, and the final titers were lower than in the immunizations with all other DNA constructs except pPcMSP119. Immunization of mice with a recombinant GST-PcMSP119 fusion protein resulted in the generation of high ELISA titers (1:512,000 to 1:1,000,000). In order to confirm the titers detected in the ELISA, we pooled the sera of each trial group and tested them in Western blots against native PcMSP1 from crude P. chabaudi schizont extracts. While the anti-PcMSP1 titers from sera of genetically immunized mice were similar in both systems, the protein-immunized mice showed dramatically lower titers in Western blots against native MSP1 (230-kDa band [Table 2]). Therefore, the sera of mice immunized with rPcMSP119-GST were only tested from here on as pools in Western blot assays.

TABLE 2.

anti-PcMSP110 titers after three immunizations of mice with different constructsa

Immunogen Anti-PcMSP119 titer ([ELISA]) Anti-MSP1 titer (Western)
pVXORF-PcMSP119 <1:200 ND
pVXORF-PcMSP119-HBs 1:4,000–1:25,800 1:32,000
pVXORF-PcMSP119HBs plus pVXVR-IL2 1:4,000–1:51,600 1:32,000
pVXORF-PcMSP119-HBs plus pVXVR-IFNγ 1:1,600-1:6,400 1:4,000
pVXORF-PcMSP119-HBs plus pGMCSF 1:1,600-1:6,400 1:4,000
pSel-PcMSP119-HBs 1:1,600-1:9,600 1:8,000
rPcMSP119-GST >1:1,000,000 1:8,000
pVXORF1 (includes groups coinjected with cytokines or rGST [n = 72]) <1:200 ND
a

These values present titers of pooled sera of all animals per immunogen (n = 12). The end point in all ELISAs was determined as a double signal above the average background signal, obtained from the GST wells. The Western blot endpoint was defined as the dilution of serum that resulted in a visible signal on X-ray films. ND, not determined. 

BALB/c mice immunized with pPcMSP119-HBs or pSel- PcMSP119-HBs, but not pPcMSP119 or pPcMSP119-HBs, coinjected with cytokine genes are partially protected from death.

We then tested the protective effect of each construct. Groups of mice were immunized as described above, and after three doses mice were challenged with 2.5 × 104 P. chabaudi IRBC. As shown in Fig. 2, only in the pPcMSP119-HBs (Fig. 2A) and in the pSel-PcMSP119-HBs (Fig. 2B) were there significant numbers of survivors (P = 0.00002 [χ2 = 31.2] and P = 0.002 [χ2 = 18.2], respectively). Groups of mice immunized with pPcMSP119-HBs and pIL2 (Fig. 2C) showed few survivors (2 of 12 mice recovered from infection; P = 0.019, χ2 12.0). Coinjection of pPcMSP119-HBs plus pGM-CSF (Fig. 2D) or pPcMSP119-HBs plus pIFNγ (Fig. 2E), and each of their respective control plasmids (pVXORF1+ pIFNγ or pGM-CSF, respectively) showed no survivors (not shown). Surprisingly, the injection of rPcMSP119-GST (Fig. 2G) did not provide significant protection; indeed, only 1 mouse out of 12 in two independent trials survived a 25,000 IRBC challenge (P > 0.05). The protection results are summarized in Table 3.

FIG. 2.

FIG. 2

Course of infection in mice immunized with different plasmid constructs expressing PcMSP119. Groups of mice (n = 12) were immunized with different plasmids as described and infected i.p. with 25,000 IRBC. The parasitemia is indicated in a dotted line with squares, the percentage of survivors is marked as a line with triangles (left y axis), and the actual anti-PcMSP119 titer is indicated as a thick gray line with diamonds (right y axis). All values are geometric mean values, and were plotted against the day of infection. Mice were immunized with pVXORF-PcMSP119-HBs (A), pSel-PcMSP119-HBs (B), pVXORF-PcMSP119-HBs plus pIL2 (C), pVXORF-PcMSP119-HBs plus pGMCSF (D), pVXORF-PcMSP119-HBs plus pIFNγ (E), rPcMSP119-GST (G), or pVXORF1 (F) as described in Materials and Methods. All negative control mice injected with cytokine vectors and pVXORF1 or rGST, reacted essentially as shown in panel F.

TABLE 3.

Protection of mice immunized with different constructs

Immunogen construct No. of survivors/total no. after infection on:
Pa
Day 5 Day 12 Day 20
Control 72/72 0/72 0/12
pPcMSP119HBs plus pGMCSF 12/12 0/12 0/12
pPcMSP119-HBs plus pIFNγ 12/12 1/12 0/12
rMSP119-GST 12/12 3/12 1/12 >0.05
pPcMSP119-HBs plus pIL-2 12/12 3/12 2/12 0.019
pSel-MSP119-HBs 12/12 3/12 3/12 0.002
pPcMSP119 12/12 5/12 5/12 0.00002
a

Values were calculated by using the χ2 test applying Fisher exact values and comparing the control group values of day 20 versus the test group values on day 20. 

In order to determine if a certain anti-PcMSP119 titer correlated with survival, we also analyzed the sera of mice during infection starting from day 5 and plotted them against parasitemias (Fig. 2). All mice immunized with pPcMSP119 and pSel-PcMSP119-HBs began to show increased anti-PcMSP119 levels significantly from day 6 or 7 on, coinciding with the increasing parasitemias. In mice coimmunized with pIL2, pGMCSF, and most prominently pIFNγ, the boosting effect was delayed (pIL2) or almost abolished (pGMCSF and pIFNγ). In the case of coinjection of pIFNγ, the maximum titer of anti-PcMSP119 was never higher than 1:32,000 as determined by ELISA, whereas pPcMSP119-HBs-injected mice had titers of up to 1:2,000,000 (Fig. 2A). Nevertheless, no distinct limit could determined as a parameter of protection; some animals died with a final anti-PcMSP119-titer of 1:512,000, while others survived with titers never higher than 1:128,000 (pSel-PcMSP119-HBs group, Fig. 2B). The injection of mice with rPcMSP119-GST led to the development of maximum titers of 1:40,000 on day 8 of infection in all mice in the endpoint dilution Western blots. The only surviving mouse developed a titer of 1:512,000 in the endpoint dilution Western blots on day 20 of the trial.

When comparing the developed anti-MSP119 antibody titers on day 9 of infection, we found that the titers were significantly higher in pPcMSP119-HBs-immunized mice than in all other groups (Student's t test, two-tailed; P < 0.004) with exception of the selectin-MSP119-HBs groups (P = 0.06). The titers on day 9 in mice coinjected with pIL2 and pVXORFMSP119-HBs were significantly higher than in mice coinjected with pIFNγ or pGMCSF or the negative control mice (P < 0.0006), while the titers of mice coinjected with either pIFNγ or pGMCSF were not significantly different, and the titers on day 9 of pIFNγ mice were not even different from those of the negative control mice (P > 0.05). Coinjection of pGMCSF with pPcMSP119-HBs resulted in the lowest day 9 titers, which were significantly lower than the titers in the control mice (P = 0.022).

Only discontinuous epitopes are mainly recognized in protected animals.

In order to define whether discontinuous or linear PcMSP1 epitopes are preferentially recognized by the sera of protected mice, they were analyzed in immunoblots by using reducing and nonreducing SDS-PAGE conditions. As shown in Fig. 3, the sera of mice immunized with DNA vaccines recognized only unreduced MSP1, indicating that pPcMSP119-specific antibodies are elicited against discontinuous epitopes (signal for second boost). When the recombinant protein was injected, both forms of MSP1 were recognized (Fig. 3C). Importantly, before infection all immunized mice recognized several forms of MSP1 (nonreduced forms), representing its processed products, but not the 33-kDa product, which results from cleavage of the 42-kDa protein into the 19- and 33-kDa forms. During infection, however, antibodies were developed against other parts of the molecule, leading to a positive signal against reduced MSP1 (Fig. 3A and B, day 9, +DTT lanes). These linear epitopes seem to be localized exclusively in the N-terminal region of MSP1, since only the full-length form was recognized by the sera of DNA-immunized mice (Fig. 3A and B, signal for day 9, +DTT) and not, for example, the biggest cleavage product in the range of 130 to 150 kDa (compare with Fig. 3A and B, second boost, −DTT lanes).

FIG. 3.

FIG. 3

Genetic immunization elicits mostly anticonformational PcMSP119-specific epitopes. Whole extracts of P. chabaudi schizonts were electrophoresed with (+) or without (−) dithiothreitol (DTT), blotted onto nitrocellulose membranes and detected with the sera of individual mice of three selected groups of six mice each. Sera were taken before infection (2.boost) or on day 8 (d8) or day 9 (d9) after infection. (A) Sera of six mice immunized with pVXORF-PcMSP119-HBs were incubated in a dilution of 1:1,000 (2.boost) or 1:5,000 (d9). (B) Sera of six mice immunized with pSel-PcMSP119-HBs were incubated in a dilution of 1:1,000 (2.boost) or 1:5,000 (d9). (C) Sera of six mice immunized with rPcMSP119-GST were incubated in a dilution of 1:500 (2.boost) or 1:5,000 (d8). Lanes −, reaction of anti-mouse IgG peroxidase conjugate with extracts and recombinant proteins.

Genetic immunization with pVXORF-PcMSP119-HBs induces mostly subclass IgG2a anti-PcMSP119.

To determine if the previous induction of a specific PcMSP119 antibody subclass was responsible for the protection of mice, we determined the immunoglobulin subclasses in all vaccine trials (Fig. 4). No striking correlation could be found between survival and the appearance of a specific IgG subclass pattern, since all mice immunized with PcMSP119-HBs or coinjected with IL-2, IFN-γ, GM-CSF, and pSel-PcMSP119-HBs produced mostly IgG2a and IgG2b. In contrast, immunization with rPcMSP119-GST induced mainly a Th2-like response, as characterized by high titers of IgG1, IgG2a, and IgG2b and the almost complete absence of all other subclasses and subtypes.

FIG. 4.

FIG. 4

Anti-PcMSP119-specific immunoglobulin isotypes after immunization with different plasmids. Mice were immunized with plasmids coding for the indicated products, and immunoglobulin subclasses were determined before infection. The serum dilution of each individual serum was adjusted so that an OD450 of 0.5 was expected in the ELISA. Geometric mean OD450 values with standard deviations are shown.

DISCUSSION

In this study, we demonstrate that the genetic immunization of mice with recombinant plasmids encoding the C terminus of the P. chabaudi merozoite surface protein 1, PcMSP119, is protective against death caused by experimental blood stage infections with the highly virulent strain P. chabaudi PC1. The most efficient construct used in our study encodes a fusion between PcMSP119 and the small hepatitis B virus surface antigen, PcMSP119-HBs, which forms particle-like structures upon synthesis in transfected cells. In an attempt to increase the protective capacity of PcMSP119-HBs, we tried the coinjection of different cytokines of the Th1 subset, namely, IFN-γ-, IL-2, and GM-CSF, and the inclusion of a lymph node-targeting signal in PcMSP119-HBs. None of them, however, increased the numbers of survivors after challenge compared to the use of PcMSP119-HBs alone. To the best of our knowledge, this report represents the first protective MSP119 DNA vaccine in the P. chabaudi-mouse model.

In earlier studies, others have shown that genetic immunizations of plasmids encoding MSP1 or portions of it, including MSP119, provided protection in BALB/c and C57BL/6 mice against infection with the nonlethal P. yoelii NL17 strain (2). Moreover, Kang and collaborators compared the protective capacity of a fusion between the MSP119 of P. yoelii and GST delivered as a DNA vaccine or as protein (14). They found no protection against lethal challenge with P. yoelii after immunization with the DNA construct. In our studies, DNA vaccination with the plasmid pPcMSP119-HBs provided a better protective effect than any other anti-blood stage malaria MSP1-based DNA vaccine so far reported (14). Survival occurred in 5 of 12 animals. Most likely, the HBs moiety plays a crucial role in this protection since we had previously demonstrated that HBs augmented the antigenicity of a plasmid encoding the MSP119 from P. vivax (10) and that it was able to form a viral hybrid particle (28). Moreover, a widely recognized T-helper-cell epitope is contained in the HBs domain (11).

In order to increase the degree of protection initially found for the PcMSP119-HBs construct, we opted for the coinjection of Th1-type response-associated cytokines, previously described as being essential in combating primary malaria infections in mice (16, 25). Specifically, IFN-γ was reported to help suppress malarial blood stage infection in the early phases (19), injection of recombinant IL-2 reduced parasitemias in murine malaria infections (18), and coinjection of GM-CSF-coding plasmids increased protection against sporozoite challenge, mediated by genetic immunization with a CS construct (26). None of the coinjected cytokine-coding plasmids (pIFNγ, pIL2, or pGMCSF) increased the level of protection during blood stage infection as opposed to the use of pPcMSP119-HBs alone. In fact, while the IL-2 coinjection resulted only in slightly increased prechallenge anti-MSP119 titers, all tested cytokines exhibit a more or less pronounced inhibitory effect on the boosting of anti-MSP119 titers. This suggests that these cytokines, in concert with the cytokine repertoire induced during malarial blood stage infection, inhibited (IL-2) or abrogated (GM-CSF or IFN-γ) antibody boosting. Together with recent reports on systemic side effects upon overexpression of these cytokines (see, for example, references 7, 30 and 31), the application of GM-CSF or IFN-γ in the development of malarial DNA vaccines may have to be reconsidered.

We also attempted to increase the protective efficacy of pPcMSP119-HBs by including a lymph node targeting signal. Previous reports have demonstrated that the fusion of l-selectin to an antigen led to an increase of two orders of magnitude in the antibody response against the antigen (4). Unfortunately, no significant increase in protection or in anti-MSP119 titers in comparison with those obtained with pPcMSP119-HBs could be observed; yet, the construct was still capable of inducing antibodies against discontinuous MSP119 epitopes and significant numbers of survivor mice were found. Taken together, the protection results demonstrated that pPcMSP119-HBs conferred the highest degree of protection, followed by pSel-MSP119-HBs, pPcMSP119-HBs+pIL2, and rPcMSP119-GST. In contrast, coinjection of pIFNγ and pGMCSF offered no protection, most likely due to the low antibody titers on the day of death (Fig. 2 and Table 3).

It has been well established that protection against asexual blood stages is antibody dependent, that discontinuous epitopes are the main target of protective antibodies, and that such antibodies are cytophilic (1). Significantly, the PcMSP119-HBs DNA construct induced IgG2a antibodies mainly against nonlinear epitopes, as demonstrated in immunoblots using reduced or nonreduced whole schizont antigen as a substrate. These results reference MSP119-HBs constructs for inclusion in future trials of anti-blood stage malaria DNA vaccines.

ACKNOWLEDGMENTS

G.W. was supported by a postdoctoral fellowship from Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP), grant 96/12286-9. I.C.M. is supported by a grant from CNPQ/CAPES.

REFERENCES

  • 1.Ahlborg N, Ling I T, Holder A A, Riley E M. Linkage of exogenous T-cell epitopes to the 19-kilodalton region of Plasmodium yoelii merozoite surface protein 1 (MSP119) can enhance protective immunity against malaria and modulate the immunoglobulin subclass response to MSP119. Infect Immun. 2000;68:2102–2109. doi: 10.1128/iai.68.4.2102-2109.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Becker S I, Wang R, Hedstrom R C, Aguiar J C, Jones T R, Hoffman S L, Gardner M J. Protection of mice against Plasmodium yoelii sporozoite challenge with P. yoelii merozoite surface protein 1 DNA vaccines. Infect Immun. 1998;66:3457–3461. doi: 10.1128/iai.66.7.3457-3461.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Blackman M J, Heidrich H G, Donachie S, McBride J S, Holder A A. A single fragment of a malaria merozoite surface protein remains on the parasite during red cell invasion and is the target of invasion-inhibiting antibodies. J Exp Med. 1990;172:379–382. doi: 10.1084/jem.172.1.379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Boyle J S, Brady J L, Lew A M. Enhanced responses to a DNA vaccine encoding a fusion antigen that is directed to sites of immune induction. Nature. 1998;392:408–411. doi: 10.1038/32932. [DOI] [PubMed] [Google Scholar]
  • 5.Bruss V, Thomssen R. Mapping a region of the large envelope protein required for hepatitis B virion maturation. J Virol. 1994;68:1643–1650. doi: 10.1128/jvi.68.3.1643-1650.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Burghaus P A, Wellde B T, Hall T, Richards R L, Egan A F, Riley E M, Ballou W R, Holder A A. Immunization of Aotus nancymai with recombinant C terminus of Plasmodium falciparum merozoite surface protein 1 in liposomes and alum adjuvant does not induce protection against a challenge infection. Infect Immun. 1996;64:3614–3619. doi: 10.1128/iai.64.9.3614-3619.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Car B D, Eng V M, Schnyder B, LeHir M, Shakhov A N, Woerly G, Huang S, Aguet M, Anderson T D, Ryffel B. Role of interferon-gamma in interleukin 12-induced pathology in mice. Am J Pathol. 1995;147:1693–1707. [PMC free article] [PubMed] [Google Scholar]
  • 8.Chang S P, Case S E, Gosnell W L, Hashimoto A, Kramer K J, Tam L Q, Hashiro C Q, Nikaido C M, Gibson H L, Lee Ng C T, Barr P J, Yokota B T, Hut G S. A recombinant baculovirus 42-kilodalton C-terminal fragment of Plasmodium falciparum merozoite surface protein 1 protects Aotus monkeys against malaria. Infect Immun. 1996;64:253–261. doi: 10.1128/iai.64.1.253-261.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Daly T M, Long C A. Humoral response to a carboxyl-terminal region of the merozoite surface protein-1 plays a predominant role in controlling blood-stage infection in rodent malaria. J Immunol. 1995;155:236–243. [PubMed] [Google Scholar]
  • 10.de Oliveira C I, Wunderlich G, Levitus G, Soares I S, Rodrigues M M, Tsuji M, Del Portillo H A. Antigenic properties of the merozoite surface protein 1 gene of Plasmodium vivax. Vaccine. 1999;17:2959–2968. doi: 10.1016/s0264-410x(99)00176-0. [DOI] [PubMed] [Google Scholar]
  • 11.Greenstein J L, Schad V C, Goodwin W H, Brauer A B, Bollinger B K, Chin R D, Kuo M C. A universal T cell epitope-containing peptide from hepatitis B surface antigen can enhance antibody specific for HIV gp120. J Immunol. 1992;148:3970–3977. [PubMed] [Google Scholar]
  • 12.Hoffman S L, Carucci D J, Rogers W O. Using DNA based vaccine technology and the Malaria Genome Project to overcome obstacles to Malaria vaccine development. In: Sherman I W, editor. Malaria: parasite biology, pathogenesis and protection. Washington, D.C.: ASM Press; 1998. pp. 73–91. [Google Scholar]
  • 13.Holder A A. The precursor to major merozoite surface antigens: structure and role in immunity. Prog Allergy. 1988;41:72–97. [PubMed] [Google Scholar]
  • 14.Kang Y, Calvo P A, Daly T M, Long C A. Comparison of humoral immune responses elicited by DNA and protein vaccines based on merozoite surface protein-1 from Plasmodium yoelii, a rodent malaria parasite. J Immunol. 1998;161:4211–4219. [PubMed] [Google Scholar]
  • 15.Krieg A M, Yi A K, Schorr J, Davis H L. The role of CpG dinucleotides in DNA vaccines. Trends Microbiol. 1998;6:23–27. doi: 10.1016/S0966-842X(97)01145-1. [DOI] [PubMed] [Google Scholar]
  • 16.Langhorne J, Gillard S, Simon B, Slade S, Eichmann K. Frequencies of CD4+ T cells reactive with Plasmodium chabaudi chabaudi: distinct response kinetics for cells with Th1 and Th2 characteristics during infection. Int Immunol. 1989;1:416–424. doi: 10.1093/intimm/1.4.416. [DOI] [PubMed] [Google Scholar]
  • 17.Levitus G, Mertens F, Speranca M A, Camargo L M, Ferreira M U, del Portillo H A. Characterization of naturally acquired human IgG responses against the N-terminal region of the merozoite surface protein 1 of Plasmodium vivax. Am J Trop Med Hyg. 1994;51:68–76. doi: 10.4269/ajtmh.1994.51.68. [DOI] [PubMed] [Google Scholar]
  • 18.Lucas B, Kasper L H, Smith K, Haque A. In vivo treatment with interleukin 2 reduces parasitemia and restores IFN-gamma gene expression and T-cell proliferation during acute murine malaria. C R Acad Sci III. 1996;319:705–710. [PubMed] [Google Scholar]
  • 19.Meding S J, Cheng S C, Simon-Haarhaus B, Langhorne J. Role of gamma interferon during infection with Plasmodium chabaudi chabaudi. Infect Immun. 1990;58:3671–3678. doi: 10.1128/iai.58.11.3671-3678.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Perera K L, Handunnetti S M, Holm I, Longacre S, Mendis K. Baculovirus merozoite surface protein 1 C-terminal recombinant antigens are highly protective in a natural primate model for human Plasmodium vivax malaria. Infect Immun. 1998;66:1500–1506. doi: 10.1128/iai.66.4.1500-1506.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Sanger F, Nicklen S, Coulson A R. DNA sequencing with chain-terminating inhibitors. Proc Natl Acad Sci USA. 1977;74:5463–5467. doi: 10.1073/pnas.74.12.5463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Sato Y, Roman M, Tighe H, Lee D, Corr M, Nguyen M D, Silverman G J, Lotz M, Carson D A, Raz E. Immunostimulatory DNA sequences necessary for effective intradermal gene immunization. Science. 1996;273:352–354. doi: 10.1126/science.273.5273.352. [DOI] [PubMed] [Google Scholar]
  • 23.Stevenson M M, Tam M F. Differential induction of helper T cell subsets during blood-stage Plasmodium chabaudi AS infection in resistant and susceptible mice. Clin Exp Immunol. 1993;92:77–83. doi: 10.1111/j.1365-2249.1993.tb05951.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Taylor-Robinson A W. Regulation of immunity to malaria: valuable lessons learned from murine models. Parasitol Today. 1995;11:334–342. doi: 10.1016/0169-4758(95)80186-3. [DOI] [PubMed] [Google Scholar]
  • 25.Taylor-Robinson A W, Phillips R S, Severn A, Moncada S, Liew F Y. The role of TH1 and TH2 cells in a rodent malaria infection. Science. 1993;260:1931–1934. doi: 10.1126/science.8100366. [DOI] [PubMed] [Google Scholar]
  • 26.Weiss W R, Ishii K J, Hedstrom R C, Sedegah M, Ichino M, Barnhart K, Klinman D M, Hoffman S L. A plasmid encoding murine granulocyte-macrophage colony-stimulating factor increases protection conferred by a malaria DNA vaccine. J Immunol. 1998;161:2325–2332. [PubMed] [Google Scholar]
  • 27.Wolff J A, Malone R W, Williams P, Chong W, Acsadi G, Jani A, Felgner P L. Direct gene transfer into mouse muscle in vivo. Science. 1990;247:1465–1468. doi: 10.1126/science.1690918. [DOI] [PubMed] [Google Scholar]
  • 28.Wunderlich G, del Portillo H A. Biochemical and immunological properties of a viral hybrid particle expressing the Plasmodium vivax merozoite surface protein 1 C-terminal region. Mol Med. 2000;6:238–245. [PMC free article] [PubMed] [Google Scholar]
  • 29.Xiang Z, Ertl H C. Manipulation of the immune response to a plasmid-encoded viral antigen by coinoculation with plasmids expressing cytokines. Immunity. 1995;2:129–135. doi: 10.1016/s1074-7613(95)80001-8. [DOI] [PubMed] [Google Scholar]
  • 30.Xing Z, Tremblay G M, Sime P J, Gauldie J. Overexpression of granulocyte-macrophage colony-stimulating factor induces pulmonary granulation tissue formation and fibrosis by induction of transforming growth factor-beta 1 and myofibroblast accumulation. Am J Pathol. 1997;150:59–66. [PMC free article] [PubMed] [Google Scholar]
  • 31.Yu J M, Emmons R V, Hanazono Y, Sellers S, Young N S, Dunbar C E. Expression of interferon-gamma by stromal cells inhibits murine long-term repopulating hematopoietic stem cell activity. Exp Hematol. 1999;27:895–903. doi: 10.1016/s0301-472x(99)00009-0. [DOI] [PubMed] [Google Scholar]

Articles from Infection and Immunity are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES