Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Recombinant proteins of Plasmodium malariae merozoite surface protein 1 (PmMSP1): Testing immunogenicity in the BALB/c model and potential use as diagnostic tool

  • Yelina B. Elizardez ,

    Contributed equally to this work with: Yelina B. Elizardez, Wesley L. Fotoran

    Roles Formal analysis, Writing – original draft, Writing – review & editing

    Affiliation Núcleo de Estudos em Malária, Superintendência de Controle de Endemias/Instituto de Medicina Tropical, Universidade de São Paulo, São Paulo, Brazil

  • Wesley L. Fotoran ,

    Contributed equally to this work with: Yelina B. Elizardez, Wesley L. Fotoran

    Roles Conceptualization, Formal analysis, Writing – review & editing

    Affiliation Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil

  • Andrés J. Galisteo Junior,

    Roles Conceptualization, Formal analysis, Resources, Writing – review & editing

    Affiliation Laboratório de Protozoologia, Instituto de Medicina Tropical, Universidade de São Paulo, São Paulo, Brazil

  • Izilda Curado,

    Roles Conceptualization, Formal analysis, Resources, Writing – review & editing

    Affiliation Laboratório de Imunoepidemiologia, Superintendência de Controle de Endemias, São Paulo, Brazil

  • Norival Kesper Junior,

    Roles Resources, Writing – review & editing

    Affiliation Laboratório de Protozoologia, Instituto de Medicina Tropical, Universidade de São Paulo, São Paulo, Brazil

  • Eliana F. Monteiro,

    Roles Formal analysis, Writing – review & editing

    Affiliation Núcleo de Estudos em Malária, Superintendência de Controle de Endemias/Instituto de Medicina Tropical, Universidade de São Paulo, São Paulo, Brazil

  • Irineu Romero Neto,

    Roles Formal analysis, Writing – review & editing

    Affiliation Laboratório de Protozoologia, Instituto de Medicina Tropical, Universidade de São Paulo, São Paulo, Brazil

  • Gerhard Wunderlich ,

    Roles Conceptualization, Data curation, Funding acquisition, Resources, Supervision, Writing – original draft, Writing – review & editing

    ‡ These authors also contributed equally to this work.

    Affiliation Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil

  • Karin Kirchgatter

    Roles Conceptualization, Data curation, Funding acquisition, Resources, Supervision, Writing – original draft, Writing – review & editing

    karink@usp.br

    ‡ These authors also contributed equally to this work.

    Affiliation Núcleo de Estudos em Malária, Superintendência de Controle de Endemias/Instituto de Medicina Tropical, Universidade de São Paulo, São Paulo, Brazil

Abstract

Background

Plasmodium malariae is the third most prevalent human malaria-causing species and has a patchy, but ample distribution in the world. Humans can host the parasite for years without presenting significant symptoms, turning its diagnosis and control into a difficult task. Here, we investigated the immunogenicity of recombinant proteins of P. malariae MSP1.

Methods

Five regions of PmMSP1 were expressed in Escherichia coli as GST-fusion proteins and immunized in BALB/c mice. The specificity, subtyping, and affinity of raised antibodies were evaluated by enzyme-linked immunosorbent assays. Cellular immune responses were analyzed by lymphoproliferation assays and cytokine levels produced by splenocytes were detected by cytometry.

Results

We found that N-terminal, central regions, and PmMSP119 are strongly immunogenic in mice. After three doses, the induced immune responses remained high for 70 days. While antibodies induced after immunization with N-terminal and central regions showed similar affinities to the target antigens, affinities of IgG against PmMSP119 were higher. All proteins induced similar antibody subclass patterns (predominantly IgG1, IgG2a, and IgG2b), characterizing a mixed Th1/Th2 response. Further, autologous stimulation of splenocytes from immunized mice led to the secretion of IL2 and IL4, independently of the antigen used. Importantly, IgG from P. malariae-exposed individuals reacted against PmMSP1 recombinant proteins with a high specificity. On the other hand, sera from P. vivax or P. falciparum-infected individuals did not react at all against recombinant PmMSP1 proteins.

Conclusion

Recombinant PmMSP1 proteins are very useful diagnostic markers of P. malariae in epidemiological studies or in the differential diagnosis of malaria caused by this species. Immunization with recombinant PmMSP1 proteins resulted in a significant humoral immune response, which may turn them potential component candidates for a vaccine against P. malariae.

Introduction

Plasmodium malariae was the first malaria parasite observed in 1881 by Charles Laveran, Nobel Prize winner for discovering the cause of malaria [1]. P. malariae is a cosmopolitan parasite that does not develop below 15°C and is described in tropical and subtropical regions of sub-Saharan Africa, South America, much of Southeast Asia, Indonesia, and in many islands of the western Pacific. In general, the distribution coincides with that of P. falciparum. In South America, P. malariae is associated with zoonotic infections, and is easily confounded with P. brasilianum, a parasite that infects twelve different genera of New World primates [1]. These and humans have high levels of seropositivity for P. malariae and P. brasilianum antigens in endemic areas [2].

Parasitemias in P. malariae infections are usually low compared to those in patients infected with P. falciparum or P. vivax perhaps due to its longer developmental cycle (72 h for P. malariae versus 48 h for P. vivax and P. falciparum), lower number of merozoites produced per erythrocyte cycle, and its preference for developing in older erythrocytes [2]. The combination of these factors may be a trigger for the earlier development of an immune response by the host [2]. P. malariae may persist asymptomatically in the infected individual for long periods [3], but it may also result in sequelae after decades without being perceived by the carrier [4]. Unlike P. vivax and P. ovale infections, there is no evidence of latent liver stage forms (hypnozoites) [2].

Previous studies point to P. malariae as one of the oldest parasites that cause malaria. The adaptive success is characteristic of the slow biological cycle that favors its survival for long periods in the host organism without being noticed [1]. Although rarely associated with severe malaria, there are cases of nephrotic syndrome related to P. malariae (rev. in [2]) and cases of death in immunocompromised patients [5,6]. Also, treatment failures [7,8] and acute renal injury [9] are attributable to infection caused by P. malariae. It is important to note that this silent survival for long periods in the organism makes this parasite the most important in transfusion malaria [2,5,10,11,12]. Additionally, is difficult to detect and, consequently, to control. In this sense, an effective vaccine would be a valuable addition to the tools available for malaria control.

Merozoite surface protein 1 (MSP1) is one of the major candidates for an antimalarial vaccine because it is expressed abundantly on the merozoite surface and capable of activating the host protective immunity [13]. Studies using MSP1 in different endemic regions have shown that this protein possesses highly immunogenic fragments that generate an immune response, which frequently associates to protection in natural infections (rev. in [14]). While many studies that deal with MSP1 from P. vivax and P. falciparum have demonstrated a strong immune response in mice and monkeys [15,16], P. malariae MSP1 has not been examined so far.

In order to investigate the immunogenicity of the P. malariae MSP1 polypeptide, we evaluated the humoral and cellular immune responses of BALB/c mice immunized with five recombinant proteins, representing different portions of the PmMSP1 protein.

Materials and methods

PmMSP-1 recombinant proteins

The PmMSP1 fragments were obtained from sample I11, isolated in 2002 from an infected patient in Iporanga, a municipality located in Atlantic Forest Region of the State of São Paulo, Brazil [17]. Five fragments were selected by representing conserved and polymorphic regions of the N-terminal, C terminal and the central portion of the protein (Fig 1A) (modified from [18]). The positioning of these fragments in relation to the PfMSP1 blocks, where sequences are divided into conserved blocks (white boxes), semi-conserved blocks (hatched boxes), and variable blocks (black boxes) is shown in Fig 1B (modified from [19]). Primer sequences and amplification conditions used were previously described [18]. The purified fragments were ligated into the pGEM-T Easy vector (Promega) according to the manufacturer’s instructions and subsequently subcloned into the pGEX-3Y vector (pGEX-3X altered by fill-in modification of the BamHI site). Recombinant plasmids were checked for their integrity by semiautomatic sequencing using the Big Dye Terminator v3.0 Cycle Sequencing Kit in an ABI Genetic Analyzer 3550 (ABI, USA). The sequences were deposited in GenBank (F1, #KR072279; F2, #KY189271; F3, #KY189272; F4, #KY189273; PmMSP119, #KY189274). The recombinant PvMSP119 and PfMSP119 proteins corresponding to the amino acid sequence of the 19-kDa fragment of MSP1 of the isolates Belém and S-20, respectively, were used as controls [20,21]. Then, the expression and purification of all recombinant proteins were performed as previously described [21].

thumbnail
Fig 1. Comparison of MSP1 amino acid sequences of 16 species of Plasmodium.

The location, size, and variability of each fragment of PmMSP1 (F1, F2, F3, F4 and PmMSP119) used in this work are represented (modified from [18]) (A). Positioning of A in relation to the PfMSP1 blocks, where sequences are divided into conserved blocks (white boxes), semi-conserved blocks (hatched boxes), and variable blocks (black boxes) (modified from [19] (B).

https://doi.org/10.1371/journal.pone.0219629.g001

Immunization of mice with the recombinant proteins

For all experiments, 6 groups (F1, F2, F3, F4, PmMSP119 and GST as control) of four female 6–8 weeks old BALB/c mice were used. The immunization was performed via the intraperitoneal (i.p.) route with 25 μg of recombinant protein in the presence of Freund’s incomplete adjuvant (Sigma) and 500 ng monophosphoryl lipid A adjuvant (MPLA) (Avanti)/dose. Three doses were administered at 15-day intervals. Blood samples were collected by a puncture of the submandibular artery of the immunized animals. Blood samples were collected before the first immunization and at the 14th, 28th, 45th and 70th day after the first immunization (Fig 2A). The blood was incubated at 37°C for 1 h, then at 4°C for 1 h and centrifuged at 12,000 rpm for 10 minutes at 4°C. Resulting serum was separated in new tubes and stored at -20°C.

thumbnail
Fig 2. Schematic representation of collections of blood, immunizations, and tests performed.

The blood samples were collected before each immunization. On the 70th day, the animals were sacrificed and the spleens removed for the CBA and lymphoproliferation assays (A). Immune response of IgG antibodies in mice immunized with recombinant proteins produced in E. coli. Groups of four BALB/c mice were immunized intraperitoneally with 25 μg of the indicated recombinant proteins emulsified in Freund adjuvant plus MPLA (B). Antibody titers were detected by ELISA in the 14th, 28th, 45th and 70th day after the first immunization. The results are expressed by geometric mean ± standard deviation. * (p <0.05), ** (p <0.001) and *** (p <0.0001).

https://doi.org/10.1371/journal.pone.0219629.g002

ELISA for detection of mouse antibodies against PmMSP1 recombinant proteins

Total IgG antibodies against F1, F2, F3, F4, PmMSP119 and GST in mice sera were detected by ELISA 14, 28, 45 and 70 days after the first immunization, as described previously [22]. Recombinant antigens were added to the plates at 250 ng/well and the secondary antibody (peroxidase-coupled goat anti-mouse IgG [KPL]) was used in a 1:5000 dilution. Each serum was analyzed in serial dilutions starting from 1:200. The titers were determined as the highest dilutions presenting optical density (OD) at 450 nm (OD450) values higher than 0.1. These values represent binding of IgG to the recombinant protein after subtraction of binding of the same serum to GST alone. The results are presented as the geometric mean ± standard deviation of four animals per group.

Detection of IgG subclass responses was performed as described above, except that the secondary antibodies were specific for mouse IgG1, IgG2a, IgG2b and IgG3 (all obtained from Bethyl Laboratories) diluted 1:5,000. The OD values of the sera against the fusion proteins were corrected by subtracting the OD values from the GST-coated wells.

The affinities of the antibodies were assessed by a thiocyanate elution-based ELISA [23]. The procedure was similar to that described for the standard ELISA with the inclusion of an extra step. After washing of plates following incubation of the pooled serum dilutions of the sample after the 3rd immunization (1:25,600), ammonium thiocyanate, diluted in PBS, was added to the wells in duplicate, in concentrations ranging from 0 to 8 M. The plates were allowed to stand for 15 min at room temperature before they were washed and incubated with the secondary antibody solution. Afterwards, the concentration of ammonium thiocyanate required to dissociate 50% of the bound antibody was determined. The percentage of binding was calculated as follows: OD492 in the presence of ammonium thiocyanate x100 / OD492nm in the absence of ammonium thiocyanate.

Determination of cytokine concentration in spleen cell supernatants

Cytokine levels secreted into the splenocyte culture supernatant stimulated for 72 h with the recombinant proteins (20 μg/mL) or ConA (2.5 μg /mL), were determined using a mouse Th1/Th2/Th17 cytometric bead array kit (CBA; BD Biosciences) on a LSR Fortessa BD flow cytometer in accordance with manufacturer’s instructions and data analysis was performed by FCAP Array v.3 software.

Human serum samples

Serum samples from 15 healthy individuals and 21 individuals with unrelated diseases (3 schistosomiasis, 5 visceral leishmaniasis, 5 Chagas disease, 2 toxoplasmosis, 2 rheumatoid arthritis, 4 lupus) were obtained from the Biorepository of Samples, Laboratory of Protozoology, Tropical Medicine Institute, University of São Paulo. Serum samples from 16 individuals with malaria were obtained from the Biorepository of Samples of the Department of Parasitology, Biomedical Sciences Institute, University of São Paulo. Ethical clearance for the extension of the use of these sera was obtained from the Ethics Commission (Biomedical Sciences Institute, USP, CEPSH 20/2015, n° 1.003.485).

Immunoblotting assay for PmMSP1 detection

For this analysis, the recombinant proteins were separated by SDS-PAGE (12%) diluted (vol/vol) in SDS-sample buffer (60 mM Tris-HCl [pH 6.8], 5% 2-mercaptoethanol, 10% glycerol, and 0.01% bromophenol blue), and boiled for 5 min at 100°C. Samples were loaded on a 12% polyacrylamide minigel (Mini-Protean II; Bio-Rad). Antigens were separated by electrophoresis, transferred to nitrocellulose membranes (Hybond-C, GE Healthcare), and blocked with PBS containing 5% non-fat milk for 1 h at room temperature. Membranes were incubated with a serum pool of P. malariae patients (1:500) diluted in PBS with 1% milk. Bound antibodies were detected with horseradish peroxidase-labeled anti-human IgG (Sigma Co.). The color of detected bands was developed after reaction using a chemiluminescence detection assay (Kit Western Pico Super Signal, Pierce/Thermo Scientific).

Statistical analysis

Statistical analysis was performed using the GraphPad Prism program version 5.0 for Windows (GraphPad Software, USA). For the comparison of antibody titers of more than two groups at the same sampling time ANOVA analysis was used, and a posteriori multiple comparisons between the means of the groups were performed with Tukey’s Test. To determine the differences between the sampling times ANOVA double-ranking was used, followed by a Bonferroni test. The cutoffs were established by using the mean plus three standard deviations of signal measured with negative sera.

Ethics statement

The mice used in this study were provided by the Department of Parasitology, Institute of Biomedical Sciences. All the procedures were submitted to the Ethics Committee of the Institute of Medicine Tropical and were approved under the registration number of the Committee on Ethics in the Use of Animals (CEUA) (CPE-IMT / 000304A). All animals immunized were females aged from 4 to 6 weeks and were maintained in a 12 h light/12 h dark cycle with free access to water and standard chow under appropriate environmental and hygienic conditions. Serum was harvested by submandibular vein bleeding. After trials, mice were euthanized using CO2 asphyxiation as recommended [24]. Ethical clearance for the extension of use of sera from the plasma/sera collection at the Department of Parasitology was obtained before the experimental work (CEPSH protocol 20/2015 and extension No. 1.003.485).

Results

Immunogenicity of five different PmMSP1 recombinant proteins in mice

To characterize the immune responses generated by immunization with PmMSP1, immunogenicity studies in mice were conducted. Groups of 4 BALB/c mice were immunized with F1, F2, F3, F4, PmMSP119 and with GST alone as a control, and the resulting antibody responses were measured by ELISA using recombinant proteins. Results in Fig 2B (a-d) show that all the recombinant proteins were immunogenic.

The time course of the immunizations and antibody titers showed that the responses to F1, F2, F3, F4, PmMSP119 were boosted after the second immunization while specific antibody titers were similar (1: 809,000) after the third immunization and were maintained until the end of the experiment (day 70) (S1 Fig) (for the Prism files that generate the figures in the paper see Raw Data in S1 File).

The isotype distribution of IgG responses to F1, F2, F3, F4 and PmMSP119 showed that all recombinant proteins elicited similar patterns of IgG subclasses. Antibodies of all isotypes were detected with the levels of IgG1, IgG2a and IgG2b higher when compared to IgG3 (Fig 3). However, no significant differences were observed in the mean OD values between IgG1, IgG2a or IgG2b. The data presented here correspond to pooled sera collected at day 45.

thumbnail
Fig 3. Analysis of specific IgG subclasses.

F1, F2, F3, F4, PmMSP119 and GST, in the different groups of mice immunized with the protein fragments on the 45th day after immunization at a dilution of 1: 1200. Subclass levels IgG1, IgG2a, IgG2b and IgG3 in the sera of the animals were measured by ELISA. The results are expressed by geometric mean ± deviation pattern. * (p <0.05), ** (p <0.001) and *** (p <0.0001).

https://doi.org/10.1371/journal.pone.0219629.g003

The sera obtained were used to determine their affinity for corresponding recombinant proteins. These antibodies induced after immunization with the recombinant proteins F1, F2, F3 and F4 showed similar affinities since the concentrations of ammonium thiocyanate required for dissociating 50% of the antibodies were similar (1.279 M, 1.087 M, 1.216 M and 1.391 M, respectively). Interestingly, the observed affinity was higher (2.096 M) for PmMSP119 (Fig 4).

thumbnail
Fig 4. Determination of the affinity by ELISA of specific anti-F1, anti-F2, anti-F3, anti-F4, anti-PmMSP119 IgG antibodies based on the elution method with ammonium thiocyanate.

The pooled sera of 4 BALB/c mice immunized intraperitoneally each of the recombinant proteins were evaluated in the presence of increasing concentrations (M) of ammonium thiocyanate. The results are expressed as the average of duplicates.

https://doi.org/10.1371/journal.pone.0219629.g004

Lymphoproliferation assays

The proliferation of lymphocytes stimulated with each antigen was determined by flow cytometry, using CFSE as a proliferation indicator. Concanavalin A stimulation was used to demonstrate the viability of cells used in this experiment (positive control). In Fig 5A, representative results of the F1, F2, F3, F4, PmMSP119, GST and concanavalin A stimulation are shown. In Fig 5B, data were measured in triplicate and transformed into percentage of proliferating cells. As expected, a high percentage of proliferation was obtained with concanavalin A. Cells stimulated with F2, F3 and PmMSP119 showed a progressive decrease in fluorescence after 72 h, indicating proliferation. Almost no proliferation was observed for GST-, F1- and F4-peptide stimulation (Fig 5B).

thumbnail
Fig 5. GST-fused F2, F3, and PmMSP119 lead to lymphocyte proliferation in spleen cells from immunized mice.

Splenocytes of immunized BALB/c mice were stimulated with 20 μg/ml of each recombinant protein used in the immunizations. The cells were stimulated for 72 hours and the fluorescence emitted by CFSE was monitored using a flow cytometer. Representative result (A) of the F1, F2, F3, F4, PmMSP119, GST and concanavalin A. Data were measured in triplicate and transformed in percentage of proliferating cells (B).

https://doi.org/10.1371/journal.pone.0219629.g005

Stimulated lymphocytes secrete IL4

After antigenic stimulation, levels of cytokines secreted in the culture supernatants of the splenocytes were determined by CBA (Fig 6). The levels of Th1/Th2 cytokines detected did not show significant differences when compared between the groups, but we observed that the level of IL-4 in the groups F1 and PmMSP119 was higher than in the other groups (Fig 6). Due to the low levels of cytokines IFN-γ, IL-2, IL-6, IL-10 and TNF-α (Fig 6) found in most animals, differences in these results are not statistically significant.

thumbnail
Fig 6. Effect of recombinant proteins on the Th1/Th2 ratio.

Levels of cytokines IL-4, IFN-γ, IL-2, IL-6, IL-10 and TNF-α, in the supernatants of splenocytes, stimulated for 72 hours in the different groups of mice immunized.

https://doi.org/10.1371/journal.pone.0219629.g006

Patient sera from natural infections recognize PmMSP1 recombinant proteins

The identity and specificity of the recombinant proteins were analyzed by Western blot using a pool of sera from patients positive for P. malariae confirmed by PCR (Fig 7). The results showed a migration pattern specific and similar in the SDS-PAGE (Fig 7A). Recombinant proteins of interest, F1, F2, F3, F4, and PmMSP119, reacted with human serum samples of patients with malaria caused by P. malariae (Fig 7B). GST alone was not recognized by sera from P. malariae infected patients (Fig 7B).

thumbnail
Fig 7. SDS-PAGE electrophoresis and Western blot.

Recombinant proteins were subjected to 12% SDS-PAGE electrophoresis and stained with Coomassie Blue (A). MW: Molecular weight standard (kDa), F1, F2, F3, F4, PmMSP119 and GST; Western blot showing the recognition of proteins by a pool of sera from P. malariae patients (B). MW: Molecular weight standard (Thermo/Fermentas broad range prestained marker, weights in kDa), F1, F2, F3, F4, PmMSP119 and GST.

https://doi.org/10.1371/journal.pone.0219629.g007

PmMSP1 proteins are not cross-recognized by sera from P. vivax or P. falciparum infected patients, and vice versa

The reactivity of MSP119 proteins of P. malariae (PmMSP119), P. vivax (PvMSP119) and P. falciparum (PfMSP119) was evaluated by ELISA with five patient sera. As shown in Fig 8, when the ELISA plate was sensitized with PmMSP119, only the sera from patients I11, I16, I65 (infected with P. malariae) recognized the antigen (Fig 8, PmMSP119). When the ELISA plate was sensitized with PvMSP119 (Fig 8, PvMSP119) or PfMSP119 (Fig 8, PfMSP119), only the sera from patients with P. vivax (C_Pv) or P. falciparum infection (C_Pf) reacted with their corresponding antigens. Some recognition by P. malariae patient sera was also recorded, possibly due to previous, not reported, contact with P. vivax and P. falciparum.

thumbnail
Fig 8. Reactivity by ELISA of recombinant proteins.

PmMSP119, PvMSP119 and PfMSP119 proteins, with sera from P. malariae (I11, I16 and I65), P. vivax (C_Pv) or P. falciparum (C_Pf) positive patients.

https://doi.org/10.1371/journal.pone.0219629.g008

We also evaluated the reactivity of the recombinant proteins F1, F2, F3 and F4 with those five sera belonging to patients confirmed with P. malariae, P. vivax and P. falciparum by ELISA (Fig 9). Although our primary goal with this small study was to show the natural antibody response acquired that is induced against the different MSP1 fragments, we found a stronger recognition of F1 (Fig 9, F1) than that obtained for the F2, F3 and F4 recombinant proteins (Fig 9, F2, F3 and F4). It was also possible to observe a higher recognition of F2 by the homologous serum than by the heterologous sera (Fig 9, F2).

thumbnail
Fig 9. Reactivity by ELISA.

F1, F2, F3 and F4 recombinant proteins in sera from P. malariae (I11, I16 and I65), P. vivax (C_Pv) or P. falciparum (C_Pf) positive patients.

https://doi.org/10.1371/journal.pone.0219629.g009

MSP119 from P. malariae was recognized by sera from malaria patients caused by P. malariae (100% of responders, n = 16) (Fig 10). The corresponding IgGs were species-specific and no reaction was detected with 21 sera from individuals with unrelated diseases (specificity of 100%) (Fig 10). Cut-off points (OD 0.07) were set at three standard deviations above the mean OD450 of sera from 15 individuals, never exposed to malarial infection, from the city of São Paulo. The reactivity obtained with the sera of patients from other diseases was similar to that obtained with sera from healthy individuals. This proves that the herein produced PmMSP1 antigens are very useful when a specific humoral response against P. malariae is required.

thumbnail
Fig 10. Reactivity by ELISA of PmMSP119 by IgG antibodies in samples from sera from 16 individuals diagnosed with P. malariae or unrelated diseases.

The line represents the cutoff value (OD 0.07) established at three standard deviations above the mean OD450 of sera from 15 individuals, never exposed to malaria, from the city of São Paulo.

https://doi.org/10.1371/journal.pone.0219629.g010

Discussion

The development of effective methods for malaria diagnosis and control is still on the malaria research agenda. There is currently one vaccine (RTS,S/AS01) being applied in some African countries (https://www.who.int/news-room/detail/23-04-2019-malaria-vaccine-pilot-launched-in-malawi). Of the 25 malaria vaccine projects under development in the world, four are in stage IIb or III [25,26]. All but one candidate are targeted exclusively against P. falciparum, which is probably insufficient in areas where a significant proportion of patients suffers from P. vivax or mixed infections. Furthermore, there are three other species that infect humans that have to be considered: P. malariae, P. ovale, and P. knowlesi.

P. malariae has a much lower incidence than P. falciparum and P. vivax, but unlike P. ovale and P. knowlesi, it occurs in all endemic areas of malaria, coexisting with other species, and should, therefore, be considered in the control strategies. In fact, there are no reports to date that study the immune response to P. malariae vaccine candidate antigens. Although the P. malariae merozoite surface protein 1 was isolated and characterized nine years ago [27], this is the first report showing the immunogenicity of five recombinant forms of this protein in BALB/c mice and their recognition by sera from individuals living in malaria-endemic regions of Brazil. Different regions of PmMSP1, covering many parts of the protein, encompassing polymorphic and conserved regions, were fused to glutathione S-transferase (GST). Fusion of GST to the N-terminus of proteins facilitates their purification and often helps to maintain fused polypeptide soluble compared to a 6xHistidine-fusion. The expression system in E. coli was chosen because of its simplicity, cost-effectiveness and high efficiency in the expression of non-glycosylated proteins [28,29]. The recombinant proteins produced were used to immunize BALB/c mice using monophosphoryl lipid A adjuvant (MPLA) and Freund’s incomplete adjuvant. The specificity and subtyping of the antibodies and cellular immune responses were evaluated by ELISA and cytometry, respectively, using the recombinant proteins as antigens.

Immunogenicity of PmMSP1 recombinant proteins in mice

Regarding the response of antibodies against blood stage antigens, known for their importance in protecting against malaria [30], our results demonstrate that the recombinant proteins were immunogenic in mice. The titers of IgG antibodies elicited by F1, F2, F4, and PmMSP119 were significantly higher than the antibody titers found in the control group (GST). After the second dose, the most immunogenic proteins were the F1 (N-terminal) and PmMSP119 (C-terminal) regions. These regions were also found to be the most immunogenic in other species of Plasmodium, such as P. vivax, where, using recombinant plasmids for the immunization of mice, the most antigenic plasmids were those that encoded the N-terminal and C-terminal regions [20].

Antibodies to MSP1 may persist for up to 30 years in some patients with malaria [31]. Although several studies have shown that a high level of total IgG against total antigens of the P. falciparum blood stage is a bad predictor of protection [32], it is believed that different functions are performed by each subclass of IgG in the acquisition of immunity to the malaria parasites [33]. The development of vaccines against malaria in the blood stage focuses on the proteins expressed on the surface of the parasite or infected erythrocytes, which are accessible for antibodies. Since both the cellular and the humoral response are important against malaria parasites (rev. in [34]), parameters of both responses were monitored after immunization. An analysis of generated antibodies demonstrated that the four subclasses of IgG were detected on the 45th day after the first immunization of the mice immunized in all groups. The five recombinant proteins induced similar antibody patterns, being IgG1, IgG2a and IgG2b the most predominant. Previous studies have shown that immune responses induced by PvMSP119 antigens [35,36] or PfMSP119 [37,38,39] in mice confer high levels of IgG1 subclasses, followed by IgG2a and IgG2b. Others authors also found similar results with recombinant proteins derived from PvMSP9 [40]. It is important to note that IgG2a antibodies from mice are considered the most effective in complement activation and in the activation of antibody-dependent cellular cytotoxic mechanisms, in addition to modulating the parasitemia of P. yoelii [41,42]. In our study, the recombinant proteins in emulsion with the adjuvant induced a mixed Th1/Th2 response also found in other studies [43]. Different studies have also shown that, in P. yoelii, protection occurs in the presence of elevated levels of IgG1, IgG2a and IgG2b, with a mixed Th1/Th2 immune response [44,45,46,47].

Cytokines play a critical role in determining the subclasses of IgG. IL-4 is secreted by Th2 cells and is associated with an IgG1 response, an indicator of a predominantly antibody-mediated response. As demonstrated by our results, the spleen cells of the mice immunized and stimulated with F1 and PmMSP119 were those with the highest levels of IL-4. IL-10 cytokine is anti-inflammatory, secreted by activated Th2 cells, and is believed to limit harmful inflammatory responses during the blood stage of the parasite in mice [48]. IL-10 was found at a higher level in sera collected from mice immunized with F1 and with PmMSP119. The cytokines produced by each subset promote the polarization, where the cytokines produced by Th1 cells negatively regulate the Th2 response, and vice versa [49]. Therefore, stimulation of Th1/Th2 subsets by immunization with recombinant proteins is important, since homeostasis between Th1/Th2 cells may achieve a balanced regulation between the pro-inflammatory and anti-inflammatory actions in the immune response.

Many studies have shown that both cellular and humoral immunity are involved in protective immunity against malaria [39,50,51]. Both immune responses were triggered by the recombinant proteins derived from PmMSP1. These results suggest that these fragments of P. malariae may be more widely evaluated as potential vaccine candidates against blood stage parasites. Similar results have been reported in studies involving mice immunized with MSP142 recombinant of P. falciparum and P. vivax [36,37].

The affinity of an antibody for its corresponding antigen is an important determinant of the antibody’s biological efficacy [52,53]. For bacteria, it has been shown that affinity of antibodies is important for protection from disease after vaccination [54,55]. More recently, high-affinity antibodies to P. falciparum merozoite antigens were associated with protection from malaria [56]. Thus, we evaluated the sera from the immunized mice for their affinity against the recombinant proteins (F1, F2, F3, F4, and PmMSP119) in the presence of the chaotropic agent ammonium thiocyanate. Our results showed that the antibodies generated with immunization with PmMSP119 showed the highest affinity, requiring a higher molarity of ammonium thiocyanate to dissociate 50% of the antibodies. In the sera of the animals immunized with the F1, F2, F3 and F4 proteins, the effect of ammonium thiocyanate in the antigen-antibody interaction was very similar. Antibodies with high affinity for the target antigen have also been reported for PvMSP119 [57,58].

Antigenicity of PmMSP1 recombinant proteins in humans

PmMSP1 recombinant proteins were recognized by IgG antibodies from human sera from patients with malaria caused by P. malariae in western blot assays suggesting that the presence of the GST did not modify the epitopes of MSP1 recognized by human IgG. GST, originally from Schistosoma japonicum, was not recognized by these sera. Our experiments demonstrated that PmMSP119 was antigenic in natural infections, with species-specific recognition. Similar results were also found with human sera from Zimbabwe, Haiti, Cambodia and Mozambique, where species-specific reactivity and rare cross-reactivity of host serum to MSP119 antigens were demonstrated [59,60]. Mice immunized with PvMSP119 and PfMSP119 also showed no reactivity with the respective heterologous antigen [43]. The use of PmMSP119 for detection of antibodies in human malaria patients as well as its serological efficacy for detection of IgG and IgM among experimentally infected chimpanzees has also been demonstrated [61].

Additionally, in our analysis of the reactivity of the F1, F2, F3 and F4 recombinant proteins with sera from positive patients for P. malariae, P. vivax and P. falciparum, we observed that only sera from patients positive for P. malariae recognized the F1 and F2 proteins. This did not occur for F3 and F4, where the observed ODs were generally lower but sera positive for P. vivax and P. falciparum also recognized these proteins. Such cross-reactivity may be a consequence of the higher identity percentage that is observed between these regions and the heterologous proteins (from 50 to 60% at the amino acid level).

By ELISA, we also evaluated recognition of PmMSP119 by serum IgG from Brazilian individuals diagnosed with malaria due to P. malariae in parallel with sera from patients with other diseases, reaching 100% of sensitivity and specificity. In the particular case of malaria caused by P. vivax, studies on the naturally acquired humoral immune response against PvMSP1 showed that PvMSP119 is the most immunogenic portion of the molecule and is recognized by 90% of the patients in the region of the Brazilian Amazon with a specificity of 98.3% [62,63]. Studies have also shown that patients from endemic regions have circulating antibodies that specifically recognize the N-terminal region of PvMSP1 [64]. PfMSP119 is also strongly recognized by human sera from endemic regions of malaria [65].

This study represents the first step in assessing humoral and cellular immune responses in animal models, as well as the naturally acquired response to the PmMSP1 antigen. The different recombinant proteins produced here may be useful in sero-epidemiological studies, both in humans as well as non-human primates from different geographic regions, highlighting possible targets in the development of more sensitive and specific diagnostic tests and enabling an evaluation of the exposure to P. malariae or P. brasilianum in populations. Since the produced antigens are quite immunogenic, they may also prove useful for the rational design of vaccine formulations against P. malariae malaria. However, studies as a growth inhibition assay (GIA) and challenges by P. brasilianum of PmMSP1 protein immunized monkeys are still needed to know the true potential of these antigens as vaccine candidates.

Supporting information

S1 Fig. Analysis of the IgG antibody immune response in each collection, in the mice immunized with the recombinant proteins.

F1, F2, F3, F4, PmMSP119 and GST. Antibody titers were detected by ELISA at the 14th, 28th, 45th and 70th days after the first immunization. The results are expressed by geometric mean ± standard deviation pattern. * (p <0.05), ** (p <0.001) and *** (p <0.0001).

https://doi.org/10.1371/journal.pone.0219629.s001

(TIF)

S1 File. Zip file with Prism files that generate the figures in the paper.

https://doi.org/10.1371/journal.pone.0219629.s002

(ZIP)

Acknowledgments

We thank Almir Robson Ferreira for drawing the Fig 1. This research was funded by FAPESP (2016/04559-0) to KK. GW and KK are CNPq research fellows.

References

  1. 1. Coatney GR, Collins WE, Warren M, Contacos PG (1971) The Primates Malaria. Bethesda: National Institutes of Health.
  2. 2. Collins WE, Jeffery GM (2007) Plasmodium malariae: parasite and disease. Clin Microbiol Rev 20: 579–592. pmid:17934075
  3. 3. Antinori S, Galimberti L, Milazzo L, Corbellino M (2012) Biology of human malaria plasmodia including Plasmodium knowlesi. Mediterr J Hematol Infect Dis 4: e2012013 pmid:22550559
  4. 4. Vinetz JM, Li J, McCutchan TF, Kaslow DC (1998) Plasmodium malariae infection in an asymptomatic 74-year-old Greek woman with splenomegaly. N Engl J Med 338: 367–371. pmid:9449730
  5. 5. Kirchgatter K, Nogueira SL, Padilha A, Curado I, Boulos M, Di Santi SM (2005) Lethal malaria caused by Plasmodium malariae in an asplenic patient in Brazil. BMJ 331: 76b.
  6. 6. Langford S, Douglas NM, Lampah DA, Simpson JA, Kenangalem E, Sugiarto P, et al. (2015) Plasmodium malariae infection associated with a high burden of anemia: a hospital-based surveillance study. PLoS Negl Trop Dis 9: e0004195. pmid:26720002
  7. 7. Visser R, de Mast Q, Munnix I, van der Ven A, Dofferhoff T (2016) Failure of atovaquone-proguanil chemoprophylaxis and chloroquine treatment in Plasmodium malariae infection. Travel Med Infect Dis 14: 644–645. pmid:27546283
  8. 8. Rutledge GG, Marr I, Huang GKL, Auburn S, Marfurt J, Sanders M, et al. (2017) Genomic Characterization of Recrudescent Plasmodium malariae after Treatment with Artemether/Lumefantrine. Emerg Infect Dis 23: 1300–1307. pmid:28430103
  9. 9. Badiane AS, Diongue K, Diallo S, Ndongo AA, Diedhiou CK, Deme AB, et al. (2014) Acute kidney injury associated with Plasmodium malariae infection. Malar J 13: 226. pmid:24906879
  10. 10. Scuracchio P, Vieira SD, Dourado DA, Bueno LM, Colella R, Ramos-Sanchez EM, et al. (2011) Transfusion-transmitted malaria: case report of asymptomatic donor harboring Plasmodium malariae. Rev Inst Med Trop Sao Paulo 53: 55–59. pmid:21412621
  11. 11. Lima GF, Lucchi NW, Silva-Flannery L, Macedo-de-Oliveira A, Hristov AD, Inoue J, et al. (2016) Still Searching for a Suitable Molecular Test to Detect Hidden Plasmodium Infection: A Proposal for Blood Donor Screening in Brazil. PLoS One 11: e0150391. pmid:26959994
  12. 12. Lima GFMC, Arroyo Sanchez MC, Levi JE, Fujimori M, Da Cruz Caramelo L, Sanchez AR, et al. (2018) Asymptomatic infections in blood donors harbouring Plasmodium: an invisible risk detected by molecular and serological tools. Blood Transfus 16: 17–25. pmid:27893347
  13. 13. Holder AA (1988) The precursor to major merozoite surface antigens: structure and role in immunity. Prog Allergy 41: 72–97. pmid:3043424
  14. 14. Versiani F, Almeida M, Mariuba L, Orlandi P, Nogueira P (2013) N-terminal Plasmodium vivax merozoite surface protein-1, a potential subunit for malária vivax vaccine. Clin Dev Immunol 2013: 965841. pmid:24187566
  15. 15. Singh S, Miura K, Zhou H, Muratova O, Keegan B, Miles A, et al. (2006) Immunity to recombinant plasmodium falciparum merozoite surface protein 1 (MSP1): protection in Aotus nancymai monkeys strongly correlates with anti-MSP1 antibody titer and in vitro parasite-inhibitory activity. Infect Immun 74: 4573–4580. pmid:16861644
  16. 16. López C, Yepes-Pérez Y, Hincapié-Escobar N, Díaz-Arévalo D, Patarroyo MA (2017) What Is Known about the Immune Response Induced by Plasmodium vivax Malaria Vaccine Candidates? Front Immunol 8: 126. pmid:28243235
  17. 17. Curado I, Dos Santos Malafronte R, de Castro Duarte AM, Kirchgatter K, Branquinho MS, Bianchi Galati EA (2006) Malaria epidemiology in low-endemicity areas of the Atlantic Forest in the Vale do Ribeira, São Paulo, Brazil. Acta Trop 100: 54–62. pmid:17126279
  18. 18. Guimarães LO, Wunderlich G, Alves JM, Bueno MG, Röhe F, Catão-Dias JL, et al. (2015) Merozoite surface protein-1 genetic diversity in Plasmodium malariae and Plasmodium brasilianum from Brazil. BMC Infect Dis 15: 529. pmid:26572971
  19. 19. Tanabe K, Mackay M, Goman M, Scaife JG (1987) Allelic dimorphism in a surface antigen gene of the malaria parasite Plasmodium falciparum. J Mol Biol 195: 273–287. pmid:3079521
  20. 20. de Oliveira CI, Wunderlich G, Levitus G, Soares IS, Rodrigues MM, Tsuji M, et al. (1999) Antigenic properties of the merozoite surface protein 1 gene of Plasmodium vivax. Vaccine 17: 2959–2968. pmid:10462230
  21. 21. Medeiros MM, Fotoran WL, dalla Martha RC, Katsuragawa TH, Pereira da Silva LH, Wunderlich G (2013) Natural antibody response to Plasmodium falciparum merozoite antigens MSP5, MSP9 and EBA175 is associated to clinical protection in the Brazilian Amazon. BMC Infect Dis 13: 608. pmid:24373342
  22. 22. Cunha MG, Rodrigues MM, Soares IS (2001) Comparison of the immunogenic properties of recombinant proteins representing the Plasmodium vivax vaccine candidate MSP1(19) expressed in distinct bacterial vectors. Vaccine 20: 385–396. pmid:11672901
  23. 23. Macdonald RA, Hosking CS, Jones CL (1988) The measurement of relative antibody affinity by ELISA using thiocyanate elution. J Immunol Methods 106: 191–194. pmid:3339255
  24. 24. Boivin GP, Hickman DL, Creamer-Hente MA, Pritchett-Corning KR, Bratcher NA (2017) Review of CO₂ as a Euthanasia Agent for Laboratory Rats and Mice. J Am Assoc Lab Anim Sci 56: 491–499. pmid:28903819
  25. 25. Nikolaeva D, Draper SJ, Biswas S (2015) Toward the development of effective transmission-blocking vaccines for malaria. Expert Rev Vaccines 14: 653–680. pmid:25597923
  26. 26. WHO (2015) World Malaria Report 2015. Geneva: World Health Organization.
  27. 27. Birkenmeyer L, Muerhoff AS, Dawson GJ, Desai SM (2010) Isolation and characterization of the MSP1 genes from Plasmodium malariae and Plasmodium ovale. Am J Trop Med Hyg 82: 996–1003. pmid:20519591
  28. 28. Epp C, Kauth CW, Bujard H, Lutz R (2003) Expression and purification of Plasmodium falciparum MSP-1(42): A malaria vaccine candidate. J Chromatogr B Analyt Technol Biomed Life Sci 786: 61–72. pmid:12651002
  29. 29. Singh S, Kennedy MC, Long CA, Saul AJ, Miller LH, Stowers AW (2003) Biochemical and immunological characterization of bacterially expressed and refolded Plasmodium falciparum 42-kilodalton C-terminal merozoite surface protein 1. Infect Immun 71: 6766–6774. pmid:14638762
  30. 30. Daly TM, Long CA (1995) 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 155: 236–243. pmid:7602100
  31. 31. Lim KJ, Park JW, Yeom JS, Lee YH, Yoo SB, Oh JH, et al. (2004) Humoral responses against the C-terminal region of merozoite surface protein 1 can be remembered for more than 30 years in persons exposed to Plasmodium vivax. Parasitol Res 92: 384–389. pmid:14745545
  32. 32. Marsh K, Otoo L, Hayes RJ, Carson DC, Greenwood BM (1989) Antibodies to blood stage antigens of Plasmodium falciparum in rural Gambians and their relations to protection against infection. Trans R Soc Trop Med Hyg 83: 293–303. pmid:2694458
  33. 33. Ndungu FM, Bull PC, Ross A, Lowe BS, Kabiru E, Marsh K (2002) 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 24: 77–82. pmid:11874562
  34. 34. Marsh K, Kinyanjui S (2006) Immune effector mechanisms in malaria. Parasite Immunol 28: 51–60. pmid:16438676
  35. 35. Dutta S, Ware LA, Barbosa A, Ockenhouse CF, Lanar DE (2001) Purification, characterization, and immunogenicity of a disulfide cross-linked Plasmodium vivax vaccine candidate antigen, merozoite surface protein 1, expressed in Escherichia coli. Infect Immun 69: 5464–5470. pmid:11500418
  36. 36. Sachdeva S, Ahmad G, Malhotra P, Mukherjee P, Chauhan VS (2004). Comparison of immunogenicities of recombinant Plasmodium vivax merozoite surface protein 1 19- and 42-kilo Dalton fragments expressed in Escherichia coli. Infect Immun 72: 5775–5782. pmid:15385477
  37. 37. Sachdeva S, Mohmmed A, Dasaradhi PV, Crabb BS, Katyal A, Malhotra P, et al. (2006) Immunogenicity and protective efficacy of Escherichia coli expressed Plasmodium falciparum merozoite surface protein-1(42) using human compatible adjuvants. Vaccine 24: 2007–2016. pmid:16377036
  38. 38. Li G, Basagoudanavar SH, Gowda DC (2008) Effect of GPI anchor moiety on the immunogenicity of DNA plasmids encoding the 19-kDa C-terminal portion of Plasmodium falciparum MSP-1. Parasite Immunol 30: 315–322. pmid:18422871
  39. 39. Hui GS, Hashimoto CN (2008) Adjuvant formulations possess differing efficacy in the potentiating of antibody and cell mediated responses to a human malaria vaccine under selective immune genes knockout environment. Int Immunopharmacol 8: 1012–1022. pmid:18486913
  40. 40. Oliveira-Ferreira J, Vargas-Serrato E, Barnwell JW, Moreno A, Galinski MR (2004) Immunogenicity of Plasmodium vivax merozoite surface protein-9 recombinant proteins expressed in E. coli. Vaccine 22: 2023–2030. pmid:15121316
  41. 41. White WI, Evans CB, Taylor DW (1991) Antimalarial antibodies to immunoglobulin G2a isotype modulate parasitemias in mice infected with Plasmodium yoelii. Infect Immun 59: 3547–3554. pmid:1894361
  42. 42. Matsumoto S, Yukitake H, Kanbara H, Yamada H, Kitamura A, Yamada T (2001) Mycobaterium bovis bacillus calmette-guérin induces protective immunity against infection by Plasmodium yoelii at blood stage depending on shifting immunity toward TH1 type and inducing protective IgG2a after the parasite infection. Vaccine 19: 779–787.
  43. 43. Mehrizi AA, Zakeri S, Rafati S, Salmanian AH, Djadid ND (2011) Immune responses elicited by co-immunization of Plasmodium vivax and P. falciparum MSP-1 using prime-boost immunization strategies. Parasite Immunol 33: 594–608. pmid:21883290
  44. 44. Ahlborg N, Ling IT, Holder AA, Riley EM (2000) Linkage of exogenous T-cell epitopes to the 19-kilodalton region of Plasmodium yoelli merozoite surface protein 1 (MSP1-19) can enhance protective immunity against malaria and modulate the immunoglobulin subclass response to MSP119. Infect Immun 68: 2102–2109. pmid:10722607
  45. 45. Near KA, Stowers AW, Jankovic D, Kaslow DC (2002) Improved immunogenicity and efficacy of the recombinant 19-kilodalton merozoite surface protein 1 by the addition of oligodeoxynucleotide and aluminum hydroxide gel in a murine malaria vaccine model. Infect Immun 70: 692–701. pmid:11796601
  46. 46. Hirunpetcharat C, Wipasa J, Sakkhachornphop S, Nitkumhan T, Zheng YZ, Pichyangkul S, et al. (2003) CpG oligodeoxynucleotide enhances immunity against blood-stage malaria infection in mice parentally immunized with a yeast expressed 19 kDa carboxyl-terminal fragment of Plasmodium yoelii merozoite surface protein 1 (MSP119) formulated in oil-based Montanides. Vaccine 21: 2923–2932. pmid:12798636
  47. 47. Kumar S, Jones TR, Oakley MS, Zheng H, Kuppusamy SP, Taye A, et al. (2004) CpG oligodeoxynucleotide and montanide ISA 51 adjuvant combination enhanced the protective efficacy of a subunit malaria vaccine. Infect Immun 72: 949–957. pmid:14742540
  48. 48. Linke A, Kuhn R, Muller W, Honarvar N, Li C, Langhorne J (1996) Plasmodium chabaudi: differential susceptibility of gene-targeted mice deficient in IL-10 to an erythrocytic-stage infection. Exp Parasitol 84: 253–263. pmid:8932775
  49. 49. Kidd P (2003) Th1/Th2 balance: the hypothesis, its limitations, and implications for health and disease. Altern Med Rev 8: 223–246. pmid:12946237
  50. 50. Miller LH, Hoffman SL (1998) Research toward vaccines against malaria. Nat Med 4: 520–524. pmid:9585203
  51. 51. Smith NC, Fell A, Good MF (1998) The immune response to asexual blood stages of malaria parasites. Chem Immunol 70: 144–162. pmid:9509674
  52. 52. Steward MW, Steensgaard J (1983) Antibody Affinity: Thermodynamic Aspects and Biological Significance. Boca Raton: CRC Press.
  53. 53. Iber D (2001) Affinity Maturation in the Immune System. Oxford: Oxford University.
  54. 54. Goldblatt D, Vaz AR, Miller E (1998) Antibody avidity as a surrogate marker of successful priming by Haemophilus influenzae type b conjugate vaccines following infant immunization. J Infect Dis 177: 1112–1115. pmid:9534995
  55. 55. Breukels MA, Jol-van der Zijde E, van Tol MJ, Rijkers GT (2002) Concentration and avidity of anti-Haemophilus influenzae type b (Hib) antibodies in serum samples obtained from patients for whom Hib vaccination failed. Clin Infect Dis 34: 191–197. pmid:11740707
  56. 56. Reddy SB, Anders RF, Beeson JG, Färnert A, Kironde F, Berenzon SK et al. (2012) High affinity antibodies to Plasmodium falciparum merozoite antigens are associated with protection from malaria. PLoS One 7: e32242. pmid:22363818
  57. 57. Rosa DS, Tzelepis F, Cunha MG, Soares IS, Rodrigues MM (2004) The pan HLA DR-binding epitope improves adjuvant-assisted immunization with a recombinant protein containing a malaria vaccine candidate. Immunol Lett 92: 259–268. pmid:15081621
  58. 58. Bargieri DY, Rosa DS, Lasaro MA, Ferreira LC, Soares IS, Rodrigues MM (2007) Adjuvant requirement for successful immunization with recombinant derivatives of Plasmodium vivax merozoite surface protein-1 delivered via the intranasal route. Mem Inst Oswaldo Cruz 102: 313–317. pmid:17568936
  59. 59. Amanfo SA, Mduluza T, Midzi N, Cavanagh DR, Mutapi F (2016) Seroepidemiology of Plasmodium species infections in Zimbabwean population. Malar J 15: 267. pmid:27165412
  60. 60. Priest JW, Plucinski MM, Huber CS, Rogier E, Mao B, Gregory CJ, et al. (2018) Specificity of the IgG antibody response to Plasmodium falciparum, Plasmodium vivax, Plasmodium malariae, and Plasmodium ovale MSP1(19) subunit proteins in multiplexed serologic assays. Malar J 17: 417. pmid:30413163
  61. 61. Muerhoff AS, Birkenmeyer LG, Coffey R, Dille BJ, Barnwell JW, Collins WE, et al. (2010) Detection of Plasmodium falciparum, P. vivax, P. ovale, and P. malariae merozoite surface protein 1-p19 antibodies in human malaria patients and experimentally infected nonhuman primates. Clin Vaccine Immunol 17: 1631–1638. pmid:20702658
  62. 62. Soares IS, Levitus G, Souza JM, Del Portillo HA, Rodrigues MM (1997) Acquired immune responses to the N- and C-terminal regions of Plasmodium vivax merozoite surface protein 1 in individuals exposed to malaria. Infect Immun 65: 1606–1614. pmid:9125537
  63. 63. Rodrigues MH, Cunha MG, Machado RL, Ferreira OC, Rodrigues MM, Soares IS (2003) Serological detection of Plasmodium vivax malaria using recombinant proteins corresponding to the 19-kDa C-terminal region of the merozoite surface protein-1. Malar J 2: 39. pmid:14617378
  64. 64. Levitus G, Mertens F, Speranca MA, Camargo LM, Ferreira M, del Portillo HA (1994) 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 51: 68–76. pmid:8059917
  65. 65. Egan AF, Morris J, Barnish G, Allen S, Greenwood BM, Kaslow DC, et al. (1996) Clinical immunity to Plasmodium falciparum malaria is associated with serum antibodies to the 19-kDa C-terminal fragment of the merozoite surface antigen, PfMSP-1. J Infect Dis 173: 765–769. pmid:8627050