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

Molecular Analysis of Non-Specific Protection against Murine Malaria Induced by BCG Vaccination

  • Marcela Parra,

    Affiliation Center for Biologics Evaluation and Review, USFDA, Bethesda, Maryland, United States of America

  • Xia Liu,

    Affiliation Center for Biologics Evaluation and Review, USFDA, Bethesda, Maryland, United States of America

  • Steven C. Derrick,

    Affiliation Center for Biologics Evaluation and Review, USFDA, Bethesda, Maryland, United States of America

  • Amy Yang,

    Affiliation Center for Biologics Evaluation and Review, USFDA, Bethesda, Maryland, United States of America

  • Jinhua Tian,

    Affiliation Center for Biologics Evaluation and Review, USFDA, Bethesda, Maryland, United States of America

  • Kristopher Kolibab,

    Affiliation Center for Biologics Evaluation and Review, USFDA, Bethesda, Maryland, United States of America

  • Sanjai Kumar,

    Affiliation Center for Biologics Evaluation and Review, USFDA, Bethesda, Maryland, United States of America

  • Sheldon L. Morris

    sheldon.morris@fda.hhs.gov

    Affiliation Center for Biologics Evaluation and Review, USFDA, Bethesda, Maryland, United States of America

Abstract

Although the effectiveness of BCG vaccination in preventing adult pulmonary tuberculosis (TB) has been highly variable, epidemiologic studies have suggested that BCG provides other general health benefits to vaccinees including reducing the impact of asthma, leprosy, and possibly malaria. To further evaluate whether BCG immunization protects against malarial parasitemia and to define molecular correlates of this non-specific immunity, mice were vaccinated with BCG and then challenged 2 months later with asexual blood stage Plasmodium yoelii 17XNL (PyNL) parasites. Following challenge with PyNL, significant decreases in parasitemia were observed in BCG vaccinated mice relative to naïve controls. To identify immune molecules that may be associated with the BCG-induced protection, gene expression was evaluated by RT-PCR in i) naïve controls, ii) BCG-vaccinated mice, iii) PyNL infected mice and iv) BCG vaccinated/PyNL infected mice at 0, 1, 5, and 9 days after the P. yoelii infection. The expression results showed that i) BCG immunization induces the expression of at least 18 genes including the anti-microbial molecules lactoferrin, eosinophil peroxidase, eosinophil major basic protein and the cathelicidin-related antimicrobial peptide (CRAMP); ii) an active PyNL infection suppresses the expression of important immune response molecules; and iii) the extent of PyNL-induced suppression of specific genes is reduced in BCG-vaccinated/PyNL infected mice. To validate the gene expression data, we demonstrated that pre-treatment of malaria parasites with lactoferrin or the cathelicidin LL-37 peptide decreases the level of PyNL parasitemias in mice. Overall, our study suggests that BCG vaccination induces the expression of non-specific immune molecules including antimicrobial peptides which may provide an overall benefit to vaccinees by limiting infections of unrelated pathogens such as Plasmodium parasites.

Introduction

Mycobacterium bovis BCG has been used globally as a vaccine against tuberculosis (TB) for more than eight decades. Although the effectiveness of BCG vaccination in preventing pulmonary TB is uncertain because efficacy estimates from controlled clinical trials have varied between 0–80%, BCG has consistently been shown to protect against severe disseminated forms of TB in infants [1], [2]. Interestingly, despite its questionable effectiveness in preventing TB, early observations suggested that immunization with BCG conferred an overall beneficial impact on childhood survival [3]. In more recent years, several studies have supported these initial findings by showing that BCG vaccination imparts wide ranging beneficial health-related effects that are not directly related to its anti-tuberculosis activity. For instance, epidemiologic data has shown that BCG vaccination was associated with a 45% decrease in infant mortality in Guinea-Bissau and Benin [4], [5]. This non-specific effect of BCG immunization transcended its impact on reducing disseminated childhood TB. Additionally, case control studies in Brazil demonstrated that BCG immunization reduced the risk of pneumonia-related deaths by 50% [6]. A meta-analysis of 23 studies showed that exposure to BCG in early life was also associated with a protective effect against the development of asthma [7]. For several decades, intravesical BCG therapy has been the treatment of choice for several forms of bladder cancer because of the potent anti-tumor activity of BCG [8].

The antimicrobial activity against non-TB pathogens seen after BCG immunization may contribute to the non-specific beneficial public health impact of BCG vaccine. Experiments in multiple animal models have shown that BCG immunization confers partial protection against unrelated pathogens including Babesia microti, Plasmodium berghei, Toxoplasma gondhi, and Trypanosoma cruzi [9], [10], [11], [12]. In humans, clinical trial data has indicated that BCG vaccination protects against paucibacillary as well as multibacillary leprosy; the magnitude of the anti-leprosy protection has been estimated to be 41% [13]. The increased risk of M. avium infections seen in the Czech Republic when BCG immunization of newborns was discontinued suggested that BCG may also provide protection against disease caused by M. avium complex bacilli [14]. Furthermore, an observational study in Guinea Bissau concluded that the presence of a BCG scar in children significantly reduced the risk of death from malaria [5].

The immune mechanisms associated with the non-specific beneficial effect of BCG immunization are unknown. It has been shown that BCG immunization creates a Th1-like immune environment where cytokines such as IFN-γ and IL-12 and related chemokines including Cxcl9 and Cxcl10 are over-expressed [15], [16]. This strong induction of Th1-type immune responses after BCG immunization may explain its impact on asthma; when Th2-type immune responses characteristic in patients with asthma are decreased, the development of atopic disorders are reduced. Importantly, BCG infection has also been shown to up-regulate the expression of antimicrobial molecules such as the cathelicidin-like peptides [17]. These immune molecules play an important role in innate host defenses via the direct killing of a wide variety of microbes and in adaptive immune responses through immune regulation [18], [19]. Often the synergism between cathelicidins and other defensin-like compounds can greatly amplify the host antimicrobial response. The capacity of BCG to stimulate innate immune mechanisms including the expression of broad spectrum antimicrobial molecules may contribute to its ability to limit infections by unrelated pathogens.

In this study, we assessed the impact BCG immunization on the progression of mouse malaria infections and evaluated the molecular factors that may contribute toward BCG-induced immunity against malaria. Similar to tuberculosis, malaria remains a significant global public health challenge with the total burden of disease estimated to be approximately 250 million clinical cases and about one million deaths, mainly in young sub-Saharan African children [20]. Clearly, because many children who are vaccinated with BCG reside in areas with high rates of malaria, it is important to investigate whether BCG induces non-specific protection against malaria. To characterize the impact that BCG immunization has on the course of malaria infections, we evaluated whether BCG vaccination protected against PyNL infections of mice. We showed that immunization with BCG partially protects against the malaria infection. In addition, we demonstrated that immunization with BCG induces the expression (even in malaria-infected mice) of at least 18 host genes including 4 genes encoding antimicrobial peptides. Finally, we showed in vivo that treatment with two of these antimicrobial peptides, lactoferrin and a cathelicidin-type peptide, significantly reduced the levels of parasitemia in PyNL-infected mice.

Materials and Methods

Murine Vaccination and P. yoelii17XNL Challenge Experiments

Pathogen-free C57BL/6 mice were obtained from the Jackson Laboratories (Bar Harbor, ME). For the malaria infection studies, five to ten mice per group were evaluated. First, the mice were vaccinated once subcutaneously with BCG Pasteur (106 bacteria in 0.1 ml of phosphate-buffered saline). At 2 months after the BCG vaccination, the mice were challenged intraperitoneally (IP) with 106 PyNL blood stage parasites. To prepare the malaria parasites, frozen stocks of PyNL-infected erythrocytes were thawed and used to infect IP three donor C57BL/6 female mice. The percent parasitemias were monitored every three days using blood smears. When 8 to 12% parasitemias were detected, blood was collected by cardiac puncture, diluted in PBS and used to infect experimental animals with 1×106 PyNL parasites in 200 µl of PBS. The percent parasitemia (parasitized RBCs/total RBCs × 100) after infection was determined by examining Giesma-stained thin blood smears.

The role of CD4 and CD8 T cell subsets in the BCG vaccine induced protective responses was assessed using cell depletion procedures. For these studies, BCG vaccinated mice were treated with 0.5 mg of a αCD4 antibody (clone GK1.5) or a αCD8 antibody (clone 2.43) at 4 and 2 days before the PyNL infection. Subsequent flow cytometry analysis showed that >99% of CD4 and CD8 T cells had been depleted by the antibody treatment.

To investigate the potential impact of the over-expressed antimicrobial peptides on the P. yoelii erythrocytic infection, 106 PyNL blood stage parasites were incubated for 2 hours with 50 µg/ml of lactoferrin or 1 hour with 50 µg/ml of the cathelicidin LL-37 peptide and then injected IP into naïve mice. One day later, 50 µg of lactoferrin or LL-37 was injected IP into the infected mice. The percent parasitemia was again determined by examining Giesma-stained thin blood smears.

Analysis of Gene Expression in Spleen Cells by RT-PCR

For the in vivo gene expression analysis, at 2 months post BCG vaccination and at Day 1, Day 5 and Day 9 following PyNL infections, the spleens were removed from mice in each of the experimental groups (naïve, BCG, PyNL and BCG plus PyNL infected mice) and the recovered cells were stored in RNA later (Qiagen, Valencia, CA). For the PyNL “in vitro” recall response studies, spleen cells from naïve and BCG vaccinated mice were plated at a concentration of 2.5×106 cells/well and incubated with 106 PyNL freshly isolated parasites. After an overnight incubation, the cells were harvested and stored in RNA later.

The RNeasy mini kit (Qiagen) was used to isolate the total RNA from the cell suspensions. Equal amounts of RNA from these samples (typically about 1 µg) were reverse transcribed to cDNA using Ambion – RETROscript kit (Ambion, Austin, TX). The GAPDH control was used to determine the quality of the cDNA conversion of each sample by PCR. The cytokine/chemokine/innate transcriptional responses were determined using the cDNA as the template for RT2 profiler PCR array system (SABiosciences, Frederick, MD) and an Applied Biosystems ViiA 7 real time PCR detection system (Applied Biosystems, Foster City, CA). The mRNA expression levels for each gene were then normalized according to the manufacturer’s instructions (ViiA 7 real time PCR array system user manual). The relative gene expression values in cell cultures were determined by dividing the gene expression levels in experimental samples by the expression values in naïve controls [15]. This value represents the mean increase or decrease of mRNA expression compared to naïve controls [17].

Statistical Analysis

The Graph Pad Prism 5 program was used to analyze the data for these experiments (Graph Pad Software, San Diego, CA). The BCG vaccination protection data, the RT-PCR results, and the lactoferrin and LL-37 treatment data were evaluated using the unpaired t test analysis of the Graph Pad Prism software.

Results

BCG Vaccination Provides Partial Protection against a P. yoelii 17XNL Infection

To evaluate whether BCG vaccination induces protection against murine PyNL infection, mice were immunized with 106 CFU of BCG Pasteur and then infected with 106 erythrocytic PyNL parasites at 2 months after the vaccination. Earlier studies had shown that maximal BCG-induced protective responses in mice are detected at 2 months after immunization (S. Derrick, unpublished data). A typical parasitemia curve for the non-lethal PyNL strain is shown in Figure 1. After an erythrocytic infection with the PyNL strain, the peak of parasitemia is generally observed at 12–14 days post-infection and the parasites are generally cleared at about 3–4 weeks after the infection. As shown in Figure 1, significant protection against the malaria infection was observed at 2 months post-BCG immunization (p<0.05). During this experiment, decreases in parasitemia were detected in BCG vaccinated mice at days 14 (20.5%- naïve vs. 6.0% - BCG, 71% reduction) and 16 (13.4% - naïve vs. 0.5% - BCG, 96% reduction) after the PyNL challenge. Interestingly, significant BCG-induced protection after malarial challenge was not seen when the mice were infected with PyNL parasites at 2 weeks following BCG vaccination (M. Parra, data not shown).

thumbnail
Figure 1. BCG vaccination confers partial protection against P. yoelii 17XNL infections in mice.

Two months after immunization with BCG, vaccinated (closed squares) and naïve mice (closed circles) were challenged with an erythrocytic stage P. yoelii infection. The post-infection parasitemia levels were determined by blood smear microscopy. These data are representative results from 3 experiments. In each study, 10 mice were used for each experimental group. * Significant difference in parasitemia levels (p<0.05) as determined by unpaired t test analysis.

https://doi.org/10.1371/journal.pone.0066115.g001

Since cellular responses are usually a critical component of the anti-malarial protective responses, we evaluated whether CD4 and CD8 T cells were responsible for the BCG-induced protection. For these studies, mice were vaccinated with BCG and then depleted of CD4 or CD8 T cells by injection of cell specific antibodies prior to the PyNL challenge. The results shown in Figure 2 indicate that both CD4 and CD8 T cells contribute to the clearance of P. yoelii infections in C57BL/6 mice. In the absence of CD4 T cells, both naïve and BCG-vaccinated mice fail to control and clear the PyNL infection; extremely high levels of parasitemia (>75%) were seen in CD4-depleted naïve and BCG-vaccinated mice at 26 days post-infection (Figure 2A). Since the naïve mice failed to control the PyNL infections, the impact of the CD4 depletion on BCG-induced protection is difficult to interpret. In contrast, the increases in parasitemia levels were less dramatic in mice depleted of CD8 T cells; clearance of the infection was observed at 4 weeks in CD8 depleted naïve controls and BCG-vaccinated mice (Figure 2B). However, depletion of CD8 T cells did eliminate the BCG-induced protection against the malaria challenge of C57BL/6 mice.

thumbnail
Figure 2. CD4 and CD8 T cells are required for the BCG vaccine induced protection against P. yoelii 17XNL infection.

BCG vaccinated (closed squares) and naïve mice (closed circles) were injected with antibodies to deplete CD4 (A) or CD8 (B) cells and then were infected with erythrocytic stage P. yoelii parasites. The post-infection parasitemia levels were determined by blood smear microscopy. Ten mice were used for each experimental group. *Significant differences in parasitemia levels (p<0.05) as determined by unpaired t test analysis.

https://doi.org/10.1371/journal.pone.0066115.g002

Gene Expression in Malaria-infected and BCG-vaccinated Mice

To investigate the mechanisms of BCG-induced protection, gene expression experiments were initiated using the SuperArray technology and RT-PCR protocols. In these studies, the in vivo expression of more than 300 immune-response related genes was assessed in splenocytes from malaria infected and control mice. Since malaria infections are known to be immunosuppressive, it was not unexpected that decreased expression of 10 genes was detected at days 5 and 9 following the malaria challenge in P. yoelii infected mice relative to non-infected naïve controls (Table 1). Down-regulation by 2–4 fold of genes encoding immune mediators such as IFN-γ, IL-1B, IL-1m, lactoferrin (Ltf) and nitrous oxide synthase 2 (Nos2) was consistently observed at 5 and 9 days post-infection. Interestingly, as the infection progressed, up-regulation of specific genes was also detected. In particular, the significant over-expression of the Csf1, Lefty1, Ccrl2, Gdf3, Il-10 and Ccl8 genes (1.8–5.8 fold increases) was seen at day 9 post-infection.

thumbnail
Table 1. Relative gene expression in mice infected with P. yoelii and naïve controls at days 1, 5 and 9 after the malaria infection.

https://doi.org/10.1371/journal.pone.0066115.t001

The impact of BCG vaccination on gene expression in non-infected and P. yoelli infected mice was evaluated by preparing cDNA from RNA extracted from splenocytes at days 0 (BCG vaccination control), 1, 5, and 9 days after the malaria challenge (Table 2). The expression of at least 15 genes was increased by BCG vaccination including three IL-1 related genes and several genes encoding chemokines. Importantly, the expression of four genes encoding antimicrobial peptides (Camp, Ltf, Pgr2, and Epx,) were increased 2.4 to 6.1 fold in BCG vaccinated mice at day 0. The Ltf gene encodes the iron-binding lactoferrin protein, the Camp gene encodes the multi-faceted cathelicidin-like CRAMP peptide while the two other genes encode the eosinophil proteins – major basic protein (Pgr2) and eosinophil peroxidase (Epx). The relative effect of BCG vaccination on the expression of these antimicrobial compounds continued as the active malaria infection progressed. By day 9 post-infection when the mean parasitemia levels were often about 10%, the over-expression of Camp, Ltf, Pgr2, and Epx genes remained significantly elevated compared to the PyNL infected non-vaccinated controls. Although the levels of expression for these molecules had dropped after the malaria infection 2–5 fold relative to the initial BCG-vaccinated controls, these genes were still up-regulated 1.6–3.2 fold in BCG immunized mice compared to the non-vaccinated animals.

thumbnail
Table 2. Relative gene expression in BCG vaccinated, P. yoelii infected mice compared to non–immunized controls.

https://doi.org/10.1371/journal.pone.0066115.t002

To further evaluate the impact of BCG immunization on the expression of genes encoding antimicrobial molecules, splenocytes were recovered from naïve and BCG immunized mice and then were cultured overnight with and without PyNL parasitized erythrocytes. As shown in Table 3, spleen cells from BCG-vaccinated animals expressed 3–4 fold higher levels of RNA from the Camp, Ltf, Pgr2, and Epx genes than naïve mice. After incubation with malaria parasitized erythrocytes, increased gene expression of 3.3–7.6 fold was detected in BCG vaccinated mice for the same genes (BCG-Py vs. naïve-Py). Interestingly, while significant reductions in Camp, Ltf, and Pgr2 gene expression (p<0.01) was seen in naïve cells incubated with Plasmodium infected erythrocytes relative to non-treated naïve cells (Naïve vs. Naïve-Py), the levels of gene expression were not significantly decreased for any of the tested genes in splenocytes from BCG vaccinated mice incubated with parasitized red cells.

thumbnail
Table 3. Gene expression from in vitro cultures of splenocytes recovered from naïve and BCG vaccinated mice and then stimulated with and without P. yoelii parasitized erythrocytes.

https://doi.org/10.1371/journal.pone.0066115.t003

Treatment with Lactoferrin or a Cathelicidin-type Peptide Reduces the Levels of P. yoelii Parasitemias

The RT-PCR studies demonstrated that the Ltf and Camp genes were up-regulated in BCG vaccinated mice. To assess whether the exposure to antimicrobial peptides impacts the levels of post-infection malaria parasitemias in mice, erythrocytic P. yoelii 17XNL parasites were incubated for 1–2 hours with either lactoferrin or the cathelicidin LL-37 peptide prior to an IP infection. One day later, the infected mice were also directly injected with lactoferrin or the cathelicidin LL-37 peptide. (The LL-37 peptide is the human ortholog of the mouse CRAMP peptide. LL-37 and CRAMP are encoded by similar genes, have similar structures and exhibit similar antimicrobial activity and tissue distribution [21], [22], [23]). As shown in Figure 3A, reductions in the malaria infection were observed after the lactoferrin treatment. In these mice, significant 30% and 95% reductions (p<0.05) of malaria parasitemias were seen at days 14 and 17, respectively, relative to control P. yoelii 17XNL infected mice. Furthermore, modest but significant decreases in malaria parasitemias were detected after treatment with 50 µg/ml of the cathelicidin LL-37 peptide (Figure 3B). At day 14 following the erythrocytic malaria infection, a significant 43% reduction in P.yoelii parasitemias (p = 0.03) was seen in mice treated with the LL-37 peptide compared to infection controls.

thumbnail
Figure 3. Anti-microbial peptide treatment decreases parasitemia levels in P. yoelii 17XNL infected mice.

P. yoelii erythrocytic-stage parasites were incubated with lactoferrin (LTF - A) or the cathelicidin LL-37 (B) peptide for 1–2 hours prior to the malaria infection as described in the Methods section. One day later, animals in the treatment groups were given 50 micrograms of LTF or the LL-37 peptide. The post-infection parasitemia levels were determined by blood smear microscopy. These data are representative results from 2 experiments. Ten mice were used for each study group. * Significant difference in parasitemia levels (p<0.05) as determined by unpaired t test analysis.

https://doi.org/10.1371/journal.pone.0066115.g003

Discussion

Immunization strategies developed against childhood diseases generally aim to decrease the burden of infection by the pathogen targeted by the specific vaccine. Interestingly, considerable recent evidence suggests that vaccination may also provide non-specific benefits to the host; these non-specific effects may have significant public health consequences [24]. For example, the reduction in mortality observed in infants immunized with measles vaccine cannot be solely explained by decreased rates of measles infections [25]. Moreover, the overall increased survival seen in children with BCG scars in West Africa likely did not entirely result from improved control of tuberculosis [5]. While the non-specific impact of some vaccines has been demonstrated, the immune mechanisms of protection have generally not been elucidated. For BCG vaccine, it is assumed that the widespread consequences of immunization are related to BCG’s capacity to generate a Th1-type immune environment. However, the precise Th1 responses associated with the non-specific protection have not been defined.

In a seminal early study, Clark et al. showed that BCG immunization protected mice against lethal malaria infections (11). Although the mechanisms of the non-specific protections were not identified, the authors speculated that BCG-induced the release of non-antibody mediators of immunity from T lymphocytes. We wanted to further expand these initial observations and explore the molecular mechanisms that may contribute to the non-specific protection. Using a mouse malaria model, we investigated the non-specific immune responses induced by BCG vaccination. Initially, we demonstrated that immunization with BCG induced significant protection against a PyNL erythrocytic infection at 2 months post-infection. At days 14 and 16 after the PyNL infection, the levels of parasitemia were reduced 71% and 93%, respectively, in BCG vaccinated mice compared to naïve controls. Although we and others have shown that live BCG can be recovered from mice up to 2 years after standard subcutaneous injection of mice, it is unclear whether the presence of live organisms are required for the general anti-microbial effects of BCG [26, 27, S. Derrick, unpublished results]. Long-term drug therapy studies are planned to evaluate whether live BCG bacilli are needed for the non-specific protection. To determine whether T cells elicited the protective responses, we used cellular depletion strategies to investigate whether CD4 T cells and CD8 T cells are mediators of the BCG-induced protection. The CD4 depletion experiments were confounded by the critical role of CD4 T cells in limiting the progression of PyNL infections in naïve C57BL/6 mice. However, the loss of BCG-induced protection following CD8 T cell depletions show that CD8 T cells contribute to the anti-malarial activity evoked by BCG vaccination.

Importantly, the results of gene expression studies have suggested potential immunomolecules induced by BCG vaccination that may mediate non-specific antimicrobial immunity. As demonstrated in previous studies, BCG is a potent immunostimulant which can modulate the expression of numerous host genes [15], [16], [28]. In our experiments, BCG vaccination induced a broad array of immune molecules including 4 antimicrobial compounds, three IL-1 related molecules, and several chemokines. Of importance, the enhanced relative expression of the four genes encoding antimicrobial compounds (Ltf -lactoferrin, Camp - CRAMP cathelicidin, Prg2– major basic eosinophil protein, and Epx- eosinophil peroxidase) were maintained for at least 9 days after the immunosuppressive malaria infection. Each of these antimicrobial compounds plays a multifunctional role in host innate immunity and provides a first line of defense against invading microbes. The cathelicidin-type CRAMP peptide has been shown to have potent antibacterial activity against a variety of organisms including M. tuberculosis, K. pneumoniae, S. aureus, and P. aeriginosa [29], [30], [31], [32]. Furthermore, cathelicidin-type peptides can promote lymphocyte recruitment, dendritic mediated T cell activation, and the induction of pro-inflammatory cytokines and chemokines [19], [33]. Lactoferrin can also inhibit the growth of bacteria, viruses, and parasites by multiple mechanisms including sequestering iron, destabilizing microbial membranes, and interfering with microbial adherence to host cells [34]. Additionally, lactoferrin possesses a wide spectrum of immune modulating activities such as the promotion of T and B cell maturation, the activation of T cells, and the induction of Th1 cytokine responses. Directly relevant to our studies, lactoferrin has been shown to reduce invasion of Plasmodium berghei sporozoites into CHO cells by blocking binding of the malaria parasites and inhibit the in vitro growth of P. falciparum [35], [36]. Finally, release of the eosinophil granule proteins (eosinophil peroxidase and major basic protein) is clearly an important defense mechanism against filarial and malarial parasitic infections. Exposure to the major basic protein and eosinophil peroxidase has been shown to be toxic for many pathogens including Schistosoma mansoni, Trichinella spiralis, Bruzea pahargi, as well as P. falciparum [37], [38]. Overall, these antimicrobial molecules provide a critical link between innate and adaptive immunity. By inducing expression of these antimicrobial compounds and enhancing recruitment of eosinophils, BCG vaccination may provide an immune environment which could limit infections of many invading pathogens before effective host adaptive responses are generated [39], [40]. The initial experiment to validate this hypothesis in our mouse model involved preincubation of P. yoelii parasites with lactoferrin prior to the malaria challenge and then one day later parenteral injection of lactoferrin. Using these procedures, we showed that the lactoferrin treatment significantly reduced the levels of malaria parasitemia relative to naïve controls. Similar treatment protocolsshowed that a cathelicidin-type peptide also modestly but significantly decreased malaria parasitemia levels after the peak of the P. yoelii infection. Interestingly, both of these antimicrobial molecules did not have substantial effect on the early stages of the infection but did impact the later parasitemia levels. The absence of an early impact on the malaria infection suggests that these compounds did not directly inactivate the parasite during the pre-incubation period but likely activated cellular immune mechanisms which resulted in,a delayed anti-parasite response. Similar temporal antimicrobial cellular responses are seen in BCG-vaccinated mice infected with a low aerosol dose of M. tuberculosis [41]. In these experiments, differences in the pulmonary growth of the tuberculous infection between naïve and vaccinated mice are not detected until 7–10 days post-challenge. A pivotal role of cellular immune responses in the anti-parasitic activity of these antimicrobial peptides is consistent with the ablation of BCG-induced protection observed after CD8 T cell depletion. Further studies are ongoing to identify the precise cellular mechanisms such as Th1 cytokine induction or cytotoxic T cell activation, which are responsible for the antimicrobial peptide mediated protection.

In the mouse malaria model, BCG vaccination also reduced the suppression of gene expression by the malaria infection. It has been well established that malaria infections cause significant depressions of the host immune responses to concurrent infections and heterologous vaccination. Although the mechanisms underlying the immune hyporesponsiveness are uncertain, the induction of regulatory T cells, the inhibition of dendritic cell antigen presentation, and/or the rapid induction of IL-10 and TGF-β may contribute to immune suppression observed during malaria infections [42], [43], [44]. In this study, the expression of 11 genes was reduced in naive mice during the active malaria infection including the Nos2, Camp, Ltf, and Pgr2 genes. In contrast, the relative expression of these immune mediators was not suppressed in malaria-infected BCG vaccinated mice compared to controls. The modulation of Nos2 gene activity by BCG immunization is of particular interest because of the widely recognized role of inducible nitric oxide synthases in immune defense against intracellular pathogens [45]. Our preliminary studies in Nos2 knockout mice (which show that the BCG anti-malarial activity is abrogated in the absence of Nos2) strongly support the involvement of nitric oxide in BCG-induced protection (M. Parra, data not shown).

Overall, we have shown that vaccination with BCG clearly induces expression of innate immune molecules including antimicrobial peptides which can provide protection against unrelated pathogens. Furthermore, we have demonstrated that the immune environment created by immunization with this potent immunostimulatory mycobacterial strain can diminish the suppressive immune effects seen after infection with malaria parasites. These data support the evidence which indicates that BCG immunization can provide overall health benefits to the vaccinated individual. Most importantly, the results of this study and related work emphasize that the removal of BCG from TB vaccination protocols may have a negative impact on public health and that public health officials should proceed with caution when contemplating the removal of BCG from new immunization protocols against TB. Furthermore, well designed clinical studies are warranted to study the impact of BCG immunization on malaria associated mortality and morbidity in low and high transmission areas.

Author Contributions

Conceived and designed the experiments: MP SM SK. Performed the experiments: MP XL SD AY JT KK. Analyzed the data: MP SD KK JT XL. Contributed reagents/materials/analysis tools: MP SD KK JT KL SK. Wrote the paper: MP SM.

References

  1. 1. Colditz GA, Brewer TF, Berkey CS, Wilson ME, Burdick E, et al. (1994) Efficacy of BCG vaccine in the prevention of tuberculosis. Meta-analysis of the published literature. JAMA 271: 698–702.
  2. 2. Trunz BB, Fine P, Dye C (2006) Effect of BCG vaccination on childhood tuberculous meningitis and miliary tuberculosis worldwide: a meta-analysis and assessment of cost-effectiveness. Lancet 367: 1173–1180.
  3. 3. Levine MI, Sackett M (1946) Results of BCG immunization in New York City. Am Rev Tuberc 53: 517–532.
  4. 4. Velema JP AE, Gandaho T, Hounye FH (1991) Childhood mortality among users and non-users of primary health care in a rural west African community. Int J Epidemiol 20: 474–479.
  5. 5. Roth A, Gustafson P, Nhaga A, Djana Q, Poulsen A, et al. (2005) BCG vaccination scar associated with better childhood survival in Guinea-Bissau. Int J Epidemiol 34: 540–547.
  6. 6. Post CL Victoria C, Valente JG, Leal Mdo C, Niobey FM, et al. (1992) Prognostic factors of hospital mortality from diarrhea or pneumonia in infants younger than 1 year old. A case-control study. Rev Saude Publica 26: 369–378.
  7. 7. El-Zein M, Parent M, Benedetti A, Rousseau MC (2010) Does BCG vaccination protect against the development of childhood asthma? A systematic review and meta-analysis of epidemiological studies. Int J Epidemiol 39: 469–486.
  8. 8. Williams SK, Hoenig DM, Ghavamian R, Soloway M (2010) Intravesical therapy for bladder cancer. Expert Opin Pharmacother 11: 947–958.
  9. 9. Ortiz-Ortiz L, Gonzalez-Mendoza A, Lamoyi E (1975) A vaccination procedure against Trypanosoma cruzi infection in mice by nonspecific immunization. J Immunol 114: 1424–1425.
  10. 10. Tabbara KJ, O’Connor GR, Nozik RA (1975) Effect of immunization with attenuated Mycobacterium bovis on experimental toxoplasmic retinochoroiditis. Am J Ophthalmol 79: 641–647.
  11. 11. Clark IA Allison AC, Cox FE (1976) Protection of mice against Babesia and Plasmodium with BCG. Nature 259: 309–311.
  12. 12. Smrkovski LL, Strickland G (1978) Rodent malaria: BCG-induced protection and immunosuppression. J Immunol 121: 1257–1261.
  13. 13. Merle CS, Cunha SS, Rodrigues LC (2010) BCG vaccination and leprosy protection: review of current evidence and status of BCG in leprosy control. Expert Rev Vaccines 9: 209–222.
  14. 14. Trnka L, Danková D, Svandová E (1994) Six years’ experience with the discontinuation of BCG vaccination. 4. Protective effect of BCG vaccination against the Mycobacterium avium intracellulare complex. Tuber Lung Dis 75: 348–352.
  15. 15. Goter-Robinson C, Derrick S, Yang AL, Jeon BY, Morris SL (2006) Protection against an aerogenic Mycobacterium tuberculosis infection in BCG-immunized and DNA-vaccinated mice is associated with early type I cytokine responses. Vaccine 24: 3522–3529.
  16. 16. Lim J, Derrick SC, Kolibab K, Yang AL, Porcelli S, et al. (2009) Early pulmonary cytokine and chemokine responses in mice immunized with three different vaccines against Mycobacterium tuberculosis determined by PCR array. Clin Vaccine Immunol 16: 122–126.
  17. 17. Méndez-Samperio P, Miranda E, Trejo A (2008) Expression and secretion of cathelicidin LL-37 in human epithelial cells after infection by Mycobacterium bovis Bacillus Calmette-Guérin. Clin Vaccine Immunol 15: 1450–1455.
  18. 18. Méndez-Samperio P (2010) The human cathelicidin hCAP18/LL-37: a multifunctional peptide involved in mycobacterial infections. Peptides 31: 1791–1798.
  19. 19. Kin NW, Chen Y, Stefanov EK, Gallo RL, Kearney JF (2011) Cathelin-related antimicrobial peptide differentially regulates T- and B-cell function. Eur J Immunol 41: 3006–3016.
  20. 20. Murray CJ, Rosenfeld LC, Lim SS, Andrews KG, Foreman KJ, et al. (2012) Global malaria mortality between 1980 and 2010: a systematic analysis. Lancet 379: 413–431.
  21. 21. Nizet V, Gallo RL (2003) Cathelicidins and innate defense against invasive bacterial infection. Scand J Infect Dis 35: 670–679.
  22. 22. Arvidsson CM, Karpman D (2012) The antimicrobial peptide cathelicidin protects mice against Escherichia coli O157:H7 mediated disease. PLoS One 7: e46476.
  23. 23. Rivas-Santiago B, Rivas-Santiago CE, Casteneda-Delgado JE, Leon-Contreras JC, Hancock REW, et al.. (2012) Acivity of LL-37, CRAMP and antimicrobial peptide-derived compounds E2, E6, and CP 26 against Mycobacterium tuberculosis. Int J Antimicrob Agents dx.doi.org/10.1016
  24. 24. Aaby P, Benn CS (2011) Non-specific and sex-differential effects of routine vaccines: what evidence is needed to take these effects into consideration in low-income countries? Hum Vaccin 7: 120–124.
  25. 25. Aaby P, Samb B, Simondon F, Seck AM, Knudsen K, et al. (1995) Non-specific beneficial effect of measles immunisation: analysis of mortality studies from developing countries. BMJ 311: 481–485.
  26. 26. Olsen AW, Brandt L, Agger EM, van Pinxteren LA, Andersen P (2004) The influence of remaining live BCG organisms in vaccinated mice on the maintenance of immunity to tuberculosis. Scand J Immunol 60: 273–277.
  27. 27. Leversen NA, Sviland L, Wiker HG, Mustafa T (2012) Long-term persistence of BCG Pasteur in lungs of C57BL/6 mice following intranasal infection. Scand J Immunol 75: 489–499.
  28. 28. Mollenkopf HJ, Hahnke K, Kaufmann SH (2006) Transcriptional responses in mouse lungs induced by vaccination with Mycobacterium bovis BCG and infection with Mycobacterium tuberculosis. Microbes Infect 8: 136–144.
  29. 29. Cirioni O, Ghiselli R, Tomasinsig L, Orlando F, Silvestri C, et al. (2008) Efficacy of LL-37 and granulocyte colony-stimulating factor in a neutropenic murine sepsis due to Pseudomonas aeruginosa. Shock 30: 443–448.
  30. 30. Dean SN, Bishop B, van Hoek ML (2011) Natural and synthetic cathelicidin peptides with anti-microbial and anti-biofilm activity against Staphylococcus aureus.. BMC Microbiol 11: 114.
  31. 31. Sonawane A, Santos JC, Mishra BB, Jena P, Progida C, et al. (2011) Cathelicidin is involved in the intracellular killing of mycobacteria in macrophages. Cell Microbiol 13: 1601–1617.
  32. 32. Kovach MA, Ballinger M, Newstead MW, Zeng X, Bhan U, et al. (2012) Cathelicidin-related antimicrobial Peptide is required for effective lung mucosal immunity in gram-negative bacterial pneumonia. J Immunol 189: 304–311.
  33. 33. Wuerth K, Hancock RE (2011) New insights into cathelicidin modulation of adaptive immunity. Eur J Immunol 41: 2817–2819.
  34. 34. Jenssen H, Hancock RE (2009) Antimicrobial properties of lactoferrin. Biochimie 91: 19–29.
  35. 35. Shakibaei M, Frevert U (1996) Dual interaction of the malaria circumsporozoite protein with the low density lipoprotein receptor-related protein (LRP) and heparan sulfate proteoglycans. J Exp Med 184: 1699–1711.
  36. 36. Fritsch G, Sawatzki G, Treumer J, Jung A, Spira DT (1987) Plasmodium falciparum: inhibition in vitro with lactoferrin, desferriferrithiocin, and desferricrocin. Exp Parasitol 63: 1–9.
  37. 37. Waters LS, Taverne J, Tai PC, Spry CJ, Targett GA, et al. (1987) Killing of Plasmodium falciparum by eosinophil secretory products. Infect Immun 55: 877–881.
  38. 38. Malik A, Batra J (2012) Antimicrobial activity of human eosinophil granule proteins: involvement in host defence against pathogens. Crit Rev Microbiol 38: 168–181.
  39. 39. Menezes-de-Lima-Júnior O, Werneck-Barroso E, Cordeiro RS, Henriques MG (1997) Effects of inhibitors of inflammatory mediators and cytokines on eosinophil and neutrophil accumulation induced by Mycobacterium bovis bacillus Calmette-Guérin in mouse pleurisy. J Leukoc Biol 62: 778–785.
  40. 40. D’Avila H, Almeida P, Roque NR, Castro-Faria-Neto HC, Bozza PT (2007) Toll-like receptor-2-mediated C-C chemokine receptor 3 and eotaxin-driven eosinophil influx induced by Mycobacterium bovis BCG pleurisy. Infect Immun 75: 1507–1511.
  41. 41. Goter-Robinson C, Derrick SC, Yang A, Jeon BY, Morris SL (2006) Protection against an aerogenic Mycobacterium tuberculosis infection in BCG-immunized and DNA-vaccinated mice is associated with early type I cytokine responses. Vaccine 24: 3522–3529.
  42. 42. Millington OR, Di Lorenzo C, Phillips RS, Garside P, Brewer JM (2006) Suppression of adaptive immunity to heterologous antigens during Plasmodium infection through hemozoin-induced failure of dendritic cell function. J Biol 5: 5.
  43. 43. Were T, Davenport GC, Yamo EO, Hittner JB, Awandare GA, et al. (2009) Naturally acquired hemozoin by monocytes promotes suppression of RANTES in children with malarial anemia through an IL-10-dependent mechanism. Microbes Infect 11: 811–819.
  44. 44. Zheng W, Wang QH, Feng H, Liu J, Meng HR, et al. (2009) CD4+CD25+Foxp3+ regulatory T cells prevent the development of Th1 immune response by inhibition of dendritic cell function during the early stage of Plasmodium yoelii infection in susceptible BALB/c mice. Folia Parasitol 56: 242–250.
  45. 45. Chakravortty D, Hensel M (2003) Inducible nitric oxide synthase and control of intracellular bacterial pathogens. Microbes Infect 5: 621–627.