Next Article in Journal
Regenerative Inflammation: Lessons from Drosophila Intestinal Epithelium in Health and Disease
Next Article in Special Issue
Fasciola hepatica: Histology of the Reproductive Organs and Differential Effects of Triclabendazole on Drug-Sensitive and Drug-Resistant Fluke Isolates and on Flukes from Selected Field Cases
Previous Article in Journal / Special Issue
Antimicrobial Human β-Defensins in the Colon and Their Role in Infectious and Non-Infectious Diseases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Use of a Th1 Stimulator Adjuvant for Vaccination against Neospora caninum Infection in the Pregnant Mouse Model

1
Institute of Parasitology, Vetsuisse Faculty, University of Berne, Länggass-Strasse 122, CH-3012 Berne, Switzerland
2
Institute of Infectious Diseases, University of Berne, Friedbühlstrasse 51, CH-3010 Berne, Switzerland
3
Biology Division, Spiez Laboratory, Swiss Federal Office for Civil Protection, CH-3700 Spiez, Switzerland
*
Authors to whom correspondence should be addressed.
Pathogens 2013, 2(2), 193-208; https://doi.org/10.3390/pathogens2020193
Submission received: 23 January 2013 / Revised: 13 February 2013 / Accepted: 19 March 2013 / Published: 27 March 2013
(This article belongs to the Special Issue Host-Parasite Interactions)

Abstract

:
Vertical transmission from an infected cow to its fetus accounts for the vast majority of new Neospora caninum infections in cattle. A vaccine composed of a chimeric antigen named recNcMIC3-1-R, based on predicted immunogenic domains of the two microneme proteins NcMIC1 and NcMIC3, the rhoptry protein NcROP2, and emulsified in saponin adjuvants, significantly reduced the cerebral infection in non-pregnant BALB/c mice. Protection was associated with a mixed Th1/Th2-type cytokine response. However, the same vaccine formulation elicited a Th2-type immune response in pregnant mice and did not prevent vertical transmission or disease, neither in dams nor in offspring mice. In this study, an alternative vaccine formulation containing recNcMIC3-1-R emulsified in Freund’s incomplete adjuvant, a stimulator of the cellular immunity, was investigated. No protection against vertical transmission and cerebral infection in the pregnant mice and a very limited protective effect in the non-pregnant mice were observed. The vaccine induced a Th1-type immune response characterized by high IgG2a titres and strong IFN-γ expression, which appeared detrimental to pregnancy.

Graphical Abstract

1. Introduction

Neospora caninum is an intracellular apicomplexan parasite, which is reported as a major cause of bovine abortion worldwide, and thus, neosporosis represents an important veterinary disease of high economic significance [1,2,3]. Vaccination against bovine neosporosis is a rational approach that appears to be the most cost-effective method for the control of the disease [4,5]. In order to confer optimal protection against neosporosis, a vaccine should be able to prevent both, the horizontal transmission from contaminated food or water and vertical transmission from an infected cow to its fetus [6,7]. Different strategies for the development of such a vaccine have been investigated [8,9,10,11]. The only commercialized vaccine, based on tachyzoite extract [12] did not achieve the required protection against vertical transmission [13] and was recently withdrawn from the market. However, more recent studies employing live vaccines showed promising results in cattle [14] and mice (reviewed in [10,15]). Our investigations focus on the use of recombinant proteins for vaccination against experimental challenge in the mouse model. Indeed, the mouse model provides valuable proof-of-concept data [16,17,18,19]. Studies employing different recombinant protein vaccine candidates have shown promising results in terms of limiting the parasite load and reducing the effects of the disease (reviewed in [15]). For instance, vaccination of mice with recNcMIC1 [20], recNcMIC3 [21] and recNcROP2 [22] conferred protection against cerebral infection and against vertical transmission when combined in a single dose vaccine [23]. Vaccination with a recombinant chimeric antigen composed of putative immunogenic domains of the three proteins (recNcMIC3-1-R) predicted in silico and emulsified in saponin adjuvants conferred a high level of protection against cerebral infection in non-pregnant mice [24], but did not prevent vertical transmission, nor cerebral infection in pregnant mice [25]. Comparison of antibody responses and cytokine data in pregnant and non-pregnant mouse models showed that a mixed Th1-/Th2-type immune response was associated with the protection observed in the non-pregnant mouse model, but a Th2-biased immune response was observed in the pregnant mice [24]. The use of saponin, a stimulator of humoral immunity, in the pregnant mouse model was thus thought to be detrimental to the Th1/Th2 balance that is obviously required for protection [25]. Thus, the use of an adjuvant that stimulates the cellular immunity, in order to counterbalance the inherent Th2-type biased immune response observed in the pregnant mice, may lead to protection. It was shown earlier that immunization of mice with antigens emulsified in complete or incomplete Freund’s adjuvant induced a Th1-type immune response by stimulating IFN-γ production [26,27]. In this paper, we report on the use of recNcMIC3-1-R emulsified in Freund’s incomplete adjuvant (FIA) for vaccination in the pregnant mouse model.

2. Results and Discussion

2.1. Cerebral Infection in Pregnant Mice

None of the formulations used produced visible lesions at the inoculation site or detectable adverse effects. All mice from the non-infected group survived without any clinical sign until the end of the experiment, and they tested all negative for N. caninum DNA in the brain (Table 1). All pregnant mice from the infected groups presented clinical signs of neosporosis and had to be killed before the end of the experiment, except for three out of eight mice in the group vaccinated with N. caninum crude extract (Table 1).
Table 1. Neosporosis in experimentally infected mice.
Table 1. Neosporosis in experimentally infected mice.
Experimental groupsNo. of miceNo. of sick mice (%)aMortality (%)bNc DNA in brain (%)cMean parasite load (±SD)d
PNPPNPPNPPNPPNP
Non-infected control9110 (0)0 (0)0 (0)0 (0)0 (0)0 (0)0 (±0)0 (±0)
FIA control13713 (100)7 (100)13 (100)5 (71.4)13 (100)7 (100)1446 (±184)1054 (±204)
MIC3-1-R in FIA8128 (100)9 (75)8 (100)7 (58.3)8 (100)12 (100)1342 (±187)851 (±364)
Crude extract in FIA8118 (100)8 (72.7)5 (62.5)6 (54.5)8 (100)11 (100)1179 (±400)748 (±443)
a Number of mice presenting at least one clinical sign of neosporosis (percentage); b Number of mice killed upon severe disease signs (percentage); c Number of dams tested positive for N; caninum DNA in the brain (percentage); d Mean parasite load per 80ng DNA in the positive mice (standard deviation); P = pregnant mice; NP = non-pregnant mice.
The survival of the mice in the crude extract group was slightly higher than in the other infected groups, and the survival probability was significantly increased compared to each of the other infected groups (P < 0.01, Cox regression analysis) (Figure 1A). However, all mice from the infected groups tested positive for N. caninum DNA in the brain (Table 1, Figure 1B), and no significant difference between the parasite loads was observed between the groups. After delivery, the weight of the non-infected mice was significantly higher than the weight of all infected mice until the end of the experiment (P < 0.01, Mann-Whitney U-test) (Figure 1C). Significant differences between the crude extract group and the other infected groups were only observed at particular time points during the experiment (data not shown).
Figure 1. Effects of vaccination in pregnant mice. (A) Kaplan-Meier survival curves of mice; n indicates the number of mice per group; * indicates a significant difference compared to the FIA group (P < 0.01). (B) Cerebral parasite burden assessed by quantitative real-time PCR. The y-axis represents the number of parasites detected in 20ng of DNA extracted from brain tissue. No significant difference between the infected groups was observed. (C) Mean weight of the surviving mice per group.
Figure 1. Effects of vaccination in pregnant mice. (A) Kaplan-Meier survival curves of mice; n indicates the number of mice per group; * indicates a significant difference compared to the FIA group (P < 0.01). (B) Cerebral parasite burden assessed by quantitative real-time PCR. The y-axis represents the number of parasites detected in 20ng of DNA extracted from brain tissue. No significant difference between the infected groups was observed. (C) Mean weight of the surviving mice per group.
Pathogens 02 00193 g001

2.2. Cerebral Infection in Non-pregnant Mice

Non-pregnant mice presenting clinical signs of neosporosis were observed in all infected groups. However, a minority of mice from each vaccinated group did not present clinical signs and almost half of the mice from each of these groups survived until the end of the experiment (Table 1, Figure 2A). The mortality rate of the mice in the two vaccinated groups was lower than in the FIA group (Table 1), although the survival curves were not statistically different from the adjuvant control group (Log-rank test) (Figure 2A). All mice from the infected groups were nonetheless positive for N. caninum DNA in the brain (Table 1), but the parasite load in each infected group was significantly lower than in the respective group in the pregnant mice (P < 0.01 for the FIA and MIC3-1-R groups; P < 0.05 for the crude extract group, Mann-Whitney U-test) (Figure 2B). However, no significant difference between the vaccinated groups and the adjuvant control group was observed. No significant difference between the mean weights of the survivors at the end of the experiment was observed (Figure 2C).
Figure 2. Effect of vaccination in non-pregnant mice. (A) Kaplan-Meier survival curves of mice; n indicates the number of mice per group; * indicates a significant difference compared to the FIA group (P < 0.01). (B) Cerebral parasite burden assessed by quantitative real-time PCR. The y-axis represents the number of parasites detected in 20 ng of DNA extracted from brain tissue. No significant difference between the infected groups was observed. (C) Mean weight of the surviving mice per group.
Figure 2. Effect of vaccination in non-pregnant mice. (A) Kaplan-Meier survival curves of mice; n indicates the number of mice per group; * indicates a significant difference compared to the FIA group (P < 0.01). (B) Cerebral parasite burden assessed by quantitative real-time PCR. The y-axis represents the number of parasites detected in 20 ng of DNA extracted from brain tissue. No significant difference between the infected groups was observed. (C) Mean weight of the surviving mice per group.
Pathogens 02 00193 g002

2.3. Vertical Transmission

No significant difference in the fertility rate was observed between the groups. However, the litter size in the non-infected group was significantly bigger than in all infected groups (P < 0.05, Mann-Whitney U-test), and the neonatal mortality was significantly lower in the non-infected group (P < 0.05, Mann-Whitney U-test) (Table 2).
Table 2. Effects of N. caninum infection of pregnant mice on offspring survival in the different experimental groups.
Table 2. Effects of N. caninum infection of pregnant mice on offspring survival in the different experimental groups.
Experimental groupsFertility (%)aNo. of pupsbAverage litter size (+/- SD)Neonatal mortality (%)cPostnatal mortality (%)d
Non-infected control9/20 (45)606.7 (1.7)1 (1.7)0/59 (0)
FIA control13/20 (65)644.9 (1.6)13 (20.3)51/51 (100)
MIC3-1-R in FIA8/20 (40)374.6 (1.4)10 (27.0)27/27 (100)
Crude extract in FIA8/19 (42.1)374.6 (1.7)7 (18.9)28/30 (93.3)
a Number of pregnant mice / number of mice in the group (%); b Total number of pups born (alive or dead) from the whole group; c Number of pups born dead or that died within the 3 first days post partum (percentage); d Number of pups dead from day 4 to 31 p.p. / no. of pups alive at day 4 p.p. (percentage).
All pups from the non-infected group that were alive on day 4 post-partum survived until the end of the experiment (Figure 3A), and tested negative for N. caninum DNA in the brain (Table 3). The survival of the pups from all infected groups was similar except for a minority of pups that survived longer in the crude extract group, although the survival curves were not significantly different (Log rank test) (Figure 3A). The number of dead neonatal mice testing positive for N. caninum DNA in the brain in the crude extract group was significantly lower than in the FIA group (P < 0.01, Chi-square test) (Table 3). All pups from each infected group died or had to be killed before the end of the experiment upon severe symptoms or euthanasia of the dam, except for two pups in the crude extract group.
All pups tested positive for N. caninum DNA in the brain, and no significant difference in the parasite load of the infected pups was observed (Kaplan-Meier multiple comparison test) (Figure 3B).
Table 3. Detection of N. caninum infection in offspring mice of the different experimental groups.
Table 3. Detection of N. caninum infection in offspring mice of the different experimental groups.
Experimental groupsNc DNA positive pupsMean parasite load (±SD)b
Neonatal deathsPostnatal deathsSurvivorsTotal
Non-infected control0/1 (0)0/0 (0)0/59 (0)0/60 (0)0 (±0)
FIA control8/9 (88.9)48/48 (100)0/0 (0)56/57 (98.2)1277 (±457)
MIC3-1-R in FIA4/7 (57.1)25/25 (100)0/0 (0)29/32 (90.6)1262 (±409)
Crude extract in FIA1/7 (14.3)25/25 (100)2/2 (100)28/34 (82.4)1276 (±192)
Figure 3. Effects of vaccination in offspring. (A) Kaplan-Meier survival curves of the pups; n indicates the overall number of pups per group; * indicates a statistically significant difference compared to the FIA group (P < 0.01). (B) Cerebral parasite burden assessed by quantitative real-time PCR in the pups. No significant difference between the infected groups was observed. The y-axis represents the number of parasites detected in 20 ng of DNA extracted from brain tissue.
Figure 3. Effects of vaccination in offspring. (A) Kaplan-Meier survival curves of the pups; n indicates the overall number of pups per group; * indicates a statistically significant difference compared to the FIA group (P < 0.01). (B) Cerebral parasite burden assessed by quantitative real-time PCR in the pups. No significant difference between the infected groups was observed. The y-axis represents the number of parasites detected in 20 ng of DNA extracted from brain tissue.
Pathogens 02 00193 g003

2.4. Humoral Immune Response

Antibody-reactivity against crude N. caninum extract was assessed by ELISA. Similar patterns were observed in the pregnant (Figure 4A) and in the non-pregnant mice (Figure 4B). In the pre-immune sera, as well as in the pre-infection and post-infection sera of the non-infected group, specific IgG1 and IgG2a levels were close to zero. In the FIA group, both IgG1 and IgG2a levels remained low after vaccination and were only elevated after challenge infection. After vaccination with recNcMIC3-1-R and crude N. caninum lysate, elevated levels of specific IgG1 and IgG2a were observed. These levels were all significantly higher than the respective levels in the FIA group (P < 0.001, Mann-Whitney U-test). After infection, both IgG1 and IgG2a levels were increased in each vaccinated groups, and IgG1 levels in the MIC3-1-R and crude extract group were significantly higher than in the FIA group (P < 0.001, Mann-Whitney U-test), while IgG2a levels reached a similar level in each group. The ratio IgG1/IgG2a in the post-infection sera was significantly higher in the MIC3-1-R group and in the Crude extract group compared to the FIA group (P < 0.01 and P < 0.001 respectively, Mann-Whitney U-test). The ratios were similar in the pregnant (Figure 4C) and in the non-pregnant mice (Figure 4D). The ratio IgG1/IgG2a in the MIC3-1-R group was below 1 in the pregnant mice and over 1 in the non-pregnant mice, but the difference was not significant.
Figure 4. Humoral immune responses measured by ELISA employing mice sera taken after vaccination, 7 days after mating prior to challenge infection (BI) and after challenge, at time of euthanasia (PI). (A) and (B) shows IgG1 and IgG2a antibody responses against N. caninum crude extract for pregnant and non-pregnant mice respectively. Values are given as OD405nm ± SD. (C) and (D): ratios of IgG1/IgG2a post-infection for pregnant and non-pregnant mice respectively. Differences in antibody response were statistically significant: * P < 0.01, ** P < 0.001
Figure 4. Humoral immune responses measured by ELISA employing mice sera taken after vaccination, 7 days after mating prior to challenge infection (BI) and after challenge, at time of euthanasia (PI). (A) and (B) shows IgG1 and IgG2a antibody responses against N. caninum crude extract for pregnant and non-pregnant mice respectively. Values are given as OD405nm ± SD. (C) and (D): ratios of IgG1/IgG2a post-infection for pregnant and non-pregnant mice respectively. Differences in antibody response were statistically significant: * P < 0.01, ** P < 0.001
Pathogens 02 00193 g004

2.5. Cytokine Responses

IFN-γ, IL-4, IL-10 and IL-12(p70) levels in pre-immune, pre-infection and post-infection sera were measured in pools from each experimental group. Low levels of cytokines were detected in all pre-immune sera (data not shown), as well as in all sera from the non-infected group.
In the FIA group, all cytokine levels were low in the pre-infection sera. IFN-γ level was increased up to 10 fold in the post infection sera of the pregnant and non-pregnant mice and IL-12 was slightly increased in the non-pregnant mice (Figure 5A,B).
In the MIC3-1-R group, all cytokine levels in the pre-infection sera were elevated compared to non-infected and FIA immunized mice. While they were increased after challenge infection in the pregnant mice, they were all reduced in the non-pregnant ones. Post-infection IL-4, as well as IL-10 and IL-12 levels were much higher than in the FIA group, while IFN-γ was only slightly higher (Figure 5A,B).
Mice vaccinated with the crude tachyzoite lysate showed low levels of cytokines after vaccination and increased levels of all cytokines after challenge infection. The relative elevation of the cytokines post-infection was similar to that seen with the MIC3-1-R group.
However, the post-infection IFN-γ/IL-4 ratio of all vaccinated group was equal or higher than 1 in both the pregnant and non-pregnant mice (Figure 5C). Moreover, this ratio was higher in the FIA group than in the MIC3-1-R and crude extract groups.

2.6. Discussion of Results

An efficient vaccine against bovine neosporosis has to prevent both horizontal and vertical transmission. Thus, studies in the animal model should focus on the outcomes of protection in both pregnant and non-pregnant animals. Among the vast number of investigations conducted in the mouse model, only few reported on protection against vertical transmission (reviewed in [15]). Indeed, as we previously showed, the same recombinant chimeric vaccine (recNcMIC3-1-R), which was protective in the non-pregnant mouse model [24] did not prevent the vertical transmission or the cerebral infection in pregnant dams [25]. The different outcomes of protection were associated with different cytokine profiles seen in pregnant and non-pregnant mice. A mixed Th1/Th2-type immune response was associated with protection in the non-pregnant mice while the lack of protection in the pregnant mice was associated with a Th2-type biased immune response [25]. As previously demonstrated, the choice of adjuvant is crucial to obtain the type of immune response sought [27,28]. In this study, we aimed to counterbalance the humoral immune response occurring during pregnancy by stimulating the cellular immune response with a Th-1 stimulator, namely FIA. Besides recNcMIC3-1-R, we included a crude antigen extract group due to its known partial efficacy against vertical transmission in mice [25,29] and cattle [13,30].
The vaccine composed of recNcMIC3-1-R induced a Th1-type biased immune response characterised by high IgG2a levels and an IgG1/IgG2a ratio close to 1 in both the pregnant and non-pregnant mice. High IFN-γ levels were observed in the post-infection sera. However, the IFN-γ/IL-4 ratio was higher in the non-pregnant mice than in the pregnant mice. Unfortunately, this vaccine formulation did not result in a balanced Th1/Th2 response, but in a strong Th-1-type immune response. This did not increase the protection against vertical transmission or cerebral infection in the pregnant mice and also decreased the protection against cerebral infection in the non-pregnant mice.
Interestingly, even if the protection against vertical transmission was lost when employing FIA as adjuvant, the protection achieved in the mice vaccinated with the crude extract group was still better than in the MIC3-1-R group. This response was associated with a mixed Th1-Th2-type immune response. The reduction of protection in this group compared to the formulation in saponin adjuvant may be explained by the lower IFN-γ expression, which appeared to be crucial for the control of the parasite replication in vitro [30] and in the mouse model [31].
Figure 5. Cytokines analysis in sera taken after vaccination, prior to challenge infection (BI) and after challenge at the time of euthanasia (PI). The sera of all mice per group were pooled and analysed in triplicates. (A) and (B) represents the concentrations of IFN-γ, IL-4, IL-10 and IL-12 in pg/mL measured in each group for the pregnant mice and non-pregnant mice respectively. Values below the detection limit were set to 1 and the arbitrary value recommended by the kit manufacturer – 1 was subtracted from measurements. Error bars represent the standard deviation of the triplicates. (C) Ratios of IFN-γ/IL-4 after challenge infection.
Figure 5. Cytokines analysis in sera taken after vaccination, prior to challenge infection (BI) and after challenge at the time of euthanasia (PI). The sera of all mice per group were pooled and analysed in triplicates. (A) and (B) represents the concentrations of IFN-γ, IL-4, IL-10 and IL-12 in pg/mL measured in each group for the pregnant mice and non-pregnant mice respectively. Values below the detection limit were set to 1 and the arbitrary value recommended by the kit manufacturer – 1 was subtracted from measurements. Error bars represent the standard deviation of the triplicates. (C) Ratios of IFN-γ/IL-4 after challenge infection.
Pathogens 02 00193 g005
The need of the appropriate Th1/Th2 balance for protection was already demonstrated in previous studies [32,33,34]. This is important considering that a Th2-type biased response was mainly associated with the vertical transmission [35,36] and exacerbation of the disease [37,38]. Indeed, it was recently demonstrated that N. caninum tachyzoites may infect dendritic cells and disseminate intracellularly across the placenta and the blood-brain barrier, thus, avoiding direct contact with serum antibodies [39]. On the other hand, a strong Th1-type immune response appeared to cause fetal death in cattle [40], and be detrimental in the mouse model [41,42,43,44]. Probably other important, but still under recognized, factors are also necessary for protection against neosporosis. Thus, new vaccination strategies should focus on wider immunological aspects, alternative ways of vaccine formulations and delivery. Indeed, Debache et al. [45] showed different outcomes of protection between intraperitoneal and intra-nasal vaccinations. Recently, Debache and Hemphill [46] also demonstrated high level protection against experimental challenge in mice by intra-cisternal vaccination. Despite the fact that the vaccine elicited high IgG2a and IFN-γ expression, which could be detrimental to the fetus, the outcome of the immune response may be different due to the immunomodulation that occurs during pregnancy.

3. Experimental Section

Unless otherwise stated, all cell culture reagents were supplied by Invitrogen (Paisly, UK) and chemicals were purchased from Sigma (St. Louis, MO, USA).

3.1. Culture and Purification of Parasites

Neospora caninum tachyzoites of the Nc-1 isolate [47] were maintained by serial passages in Vero cells using standard procedures [24]. Tachyzoites were harvested by repeatedly passaging infected cells through a 25 G-needle, and liberated parasites were purified on Sephadex G25 PD-10 columns (GE Healthcare, Buckinghamshire, England) [48]. Parasites were counted in a Neubauer chamber using trypan blue stain exclusion. For challenge infections, parasite numbers were adjusted to 2 × 107/mL in RPMI 1640 medium and tachyzoites were used immediately for infection experiments (100 µL/mouse).

3.2. Preparation of N. caninum Crude Protein Extract

N. caninum tachyzoites were purified as described in the previous section and adjusted to 1 × 108 parasites/mL in sterile PBS. They were then disrupted by performing 4 freeze/thaw cycles using a dry-ice/methanol bath and a 37 °C water bath followed by sonication for 5 × 1 min at 57 W (Branson Sonifier cell disruptor, Skan AG, Allschwill, Switzerland). The parasites were observed microscopically to ensure that complete disruption had occurred, and the protein concentration was measured using the Protein Assay Dye Reagent Concentrate (Bio-Rad, Hercules, CA, USA) and acetylated BSA as a standard. Just prior to vaccination, the lysate was vortexed and sonicated until a homogenous suspension was obtained.

3.3. Preparation of recNcMIC3-1-R

The recombinant chimeric protein recNcMIC3-1-R was obtained as described in [24]. Briefly, in silico predicted putative immunogenic epitopes of NcMIC1, NcMIC3 and NcROP2 were amplified from N. caninum cDNA and assembled as a single gene. RecNcMIC3-1-R was expressed in One Shot® BL21 (DE3) Chemically Competent E. coli (Invitrogen) and purified by Ni2+-chelate chromatography under denaturing conditions. After dialysis against 1 g/L ammonium bicarbonate the protein was dried in Speedvac and resuspended in PBS. The protein concentration was measured using the Protein Assay Dye Reagent Concentrate (Bio-Rad, Hercules, CA, USA) and acetylated BSA as a standard.

3.4. Vaccination Trial, Clinical Monitoring and Sample Collection

Six weeks old BALB/c mice were purchased from Charles River Laboratories (Sulzfeld, Germany) and maintained under conventional day/night cycle housing conditions as required by the animal welfare legislation of the Swiss Veterinary Office. They were used for experimentation after two weeks of acclimatization. Females were randomly allocated into four groups of twenty animals each and one group of 19 mice. Each group was vaccinated three times by intraperitoneal (i.p.) injection at 2 weeks interval as follows: group 1 (non-infected) was injected with 100μl PBS; group 2 (FIA) was injected with 100 μL Freund’s incomplete adjuvant (adjuvant control); group 3 (MIC3-1-R) was injected with 10 μg recNcMIC3-1-R emulsified in FIA; group 4 (crude extract) was vaccinated with 10μg N. caninum lysate (equivalent to approximately 1 × 106 parasites). Just prior to use, freshly purified recNcMIC3-1-R, as well as the crude protein extract were emulsified at the appropriate concentrations in Freund’s incomplete adjuvant (100 μL/injection). Two weeks after the third immunization, synchronization of oestrus by the Whitten effect [49], followed by mating for three nights was performed. Seven days after mating (day 7–9 of pregnancy), all mice from groups 2–4 were challenged i.p. with 2 × 106 freshly purified Nc-1 tachyzoites in 100 μL RPMI 1640 medium. The mice from group 1 received 100 μL RPMI 1640 medium. The pregnant mice were then housed separately to rear their pups, and all animals were checked for clinical signs on a daily basis until day 43 post challenge. Data on the fertility rate (percentage of pregnant mice/group), litter size, neonatal mortality (pups born dead or that died within the three first days post-partum) and postnatal mortality (pups dead between day 4 and 31 post-partum) were collected. Dams’ weights were monitored by daily measurement from day 2 before challenge until the end of the experiment, and pups were weighed from day 14 post-partum onward. Mice exhibiting clinical signs of neosporosis (ruffled coat, apathy, neurological and walking disorders), were euthanized at the onset of these signs. On day 43 post challenge, all surviving dams and pups were sacrificed by CO2 euthanasia. Upon euthanasia, blood was drawn by cardiac puncture and sera were frozen at −80 °C. Brains were collected using individual sterile instruments and were immediately frozen at −20 °C.

3.5. Cerebral Parasite Burden

Neospora-specific quantitative real-time PCR to determine the number of parasites present in the cerebral tissue was performed as described by Müller et al. [50]. DNA extraction from brain tissue was performed as described [22], and DNA concentrations were adjusted to 20ng/μL with sterile DNase-free water. Quantitative real-time PCR was performed using the Light Cycler™ Instrument (Roche Diagnostic, Basel, Switzerland). The parasite counts were calculated by interpolation from a standard curve with DNA equivalents from 1000, 100 and 10 parasites included in each run [25].

3.6. Serology

Individual sera taken at three distinct time points in the dams (pre-immune, post-immunization prior to infection (day 9 after the last immunization boost), and post-infection (at the time of euthanasia) were analyzed for N. caninum specific IgG1 and IgG2a by ELISA coated with N. caninum lysate prepared as described in section 2.2. The ELISAs were performed with a 1:100 dilution of the sera [25]. Serum from a mouse experimentally infected with Nc-1 that had reacted positively against N. caninum lysate in ELISA and serum from a non-infected mouse were used as positive and negative control, respectively.

3.7. Serum Cytokine Levels

Serum levels of IFN-γ, IL-4, IL-10 and IL-12(p70) were measured by means of multiplex bead immunoassay using the MILLIPLEX®MAP Mouse Cytokine/Chemokine kit (Millipore Corp. Billerica, MA, USA). Undiluted post-immunization and post-infection sera were pooled per group and analyzed in triplicate according to the manufacturer’s protocol. Microtitre filter plates were run on Luminex instruments (Bio-Plex™ 200 system, Bio-Rad), and cytokine concentrations were calculated with the Bio-Plex Manager software [25]. When cytokine concentration was below the detection limit, an arbitrary value corresponding to the detection limit of undiluted samples (provided by the kit manufacturer) was used for analysis. Arbitrary values corresponded to: 1 pg/mL for IFN-γ; 1 pg/mL for IL-4; 1.5 pg/mL for IL-10; 6 pg/mL for IL-12.

3.8. Statistical Analysis

Dam survival and pup mortality were analyzed according to Kaplan-Meier, and the survival curves between groups were compared with the logrank test followed by regression coefficient analysis. Litter size, neonatal mortality, weight, cerebral parasite burden, as well as serological data were compared using Kruskal-Wallis one-way ANOVA followed by the Kruskal-Wallis multiple comparison Z-value test. The P-value between two significantly different groups was calculated by Mann-Whitney U test. The fertility rates, proportion of animals with clinical signs, and data on N. caninum DNA positive animals were organized in a contingency table and compared by a Chi-square test.

4. Conclusions

In summary, the use of FIA combined with the recombinant antigen recNcMIC3-1-R for the vaccination of experimentally infected mice was not protective against vertical transmission or against cerebral infection in pregnant and non-pregnant mice. Although the immune responses observed were different from those obtained with the vaccines formulated in saponin adjuvant, the shift toward a Th-1-type immune response may have been too strong, and the sensitive Th1/Th2 balance needed was not reached. This study demonstrates once again the difficulty to achieve protection against experimental neosporosis during pregnancy, either through a Th1- or a Th2-type immune response. Further recombinant vaccine development should focus on the finding of the appropriate Th1/Th2 balance and other aspects of the immune response that were not or poorly investigated so far. The use of expression systems that allow to fuse antigens of interest to a suitable Toll-like receptor ligand, as described e.g., by Basto et al. [51] could provide the means of modulating the immune response in order to limit the consequences of N. caninum infection.

Acknowledgments

The authors wish to thank Kenneth McCullough for great support and helpful suggestions during the course of the project. J.P. Dubey (USDA, Beltsville, USA) is gratefully acknowledged for the kind gift of the N. caninum Nc-1 isolate. This work was financed through the National Science Foundations (grants no. 31003A_127374/1 and 138904).

Conflict of Interest

The authors declare no conflict of interest.

References

  1. Dubey, J.P.; Schares, G. Neosporosis in animals--the last five years. Vet. Parasitol. 2011, 180, 90–108. [Google Scholar] [CrossRef]
  2. Dubey, J.P.; Schares, G.; Ortega-Mora, L.M. Epidemiology and control of neosporosis and Neospora caninum. Clin. Microbiol. Rev. 2007, 20, 323–367. [Google Scholar] [CrossRef]
  3. Hemphill, A.; Gottstein, B. A European perspective on Neospora caninum. Int. J. Parasitol. 2000, 30, 877–924. [Google Scholar] [CrossRef]
  4. Reichel, M.P.; Ellis, J.T. If control of Neospora caninum infection is technically feasible does it make economic sense? Vet. Parasitol. 2006, 142, 23–34. [Google Scholar] [CrossRef]
  5. Reichel, M.P.; Ellis, J.T. Re-evaluating the economics of neosporosis control. Vet. Parasitol. 2008, 156, 361–362. [Google Scholar] [CrossRef]
  6. Davison, H.C.; Otter, A.; Trees, A.J. Estimation of vertical and horizontal transmission parameters of Neospora caninum infections in dairy cattle. Int. J. Parasitol. 1999, 29, 1683–1689. [Google Scholar] [CrossRef]
  7. Williams, D.J.; Hartley, C.S.; Bjorkman, C.; Trees, A.J. Endogenous and exogenous transplacental transmission of Neospora caninum - how the route of transmission impacts on epidemiology and control of disease. Parasitology 2009, 136, 1895–1900. [Google Scholar] [CrossRef]
  8. Innes, E.A.; Bartley, P.M.; Maley, S.W.; Wright, S.E.; Buxton, D. Comparative host-parasite relationships in ovine toxoplasmosis and bovine neosporosis and strategies for vaccination. Vaccine 2007, 25, 5495–5503. [Google Scholar] [CrossRef]
  9. Innes, E.A.; Vermeulen, A.N. Vaccination as a control strategy against the coccidial parasites Eimeria, Toxoplasma and Neospora. Parasitology 2006, 133 (Suppl), S145–S168. [Google Scholar]
  10. Reichel, M.P.; Ellis, J.T. Neospora caninum--how close are we to development of an efficacious vaccine that prevents abortion in cattle? Int. J. Parasitol. 2009, 39, 1173–1187. [Google Scholar] [CrossRef]
  11. Williams, D.J.; Trees, A.J. Protecting babies: Vaccine strategies to prevent foetopathy in Neospora caninum-infected cattle. Parasite Immunol. 2006, 28, 61–67. [Google Scholar] [CrossRef]
  12. Romero, J.J.; Pérez, E.; Frankena, K. Effect of a killed whole Neospora caninum tachyzoite vaccine on the crude abortion rate of Costa Rican dairy cows under field conditions. Vet. Parasitol. 2004, 123, 149–159. [Google Scholar] [CrossRef]
  13. Weston, J.F.; Heuer, C.; Williamson, N.B. Efficacy of a Neospora caninum killed tachyzoite vaccine in preventing abortion and vertical transmission in dairy cattle. Prev. Vet. Med. 2011, 103, 136–144. [Google Scholar]
  14. Williams, D.J.; Guy, C.S.; Smith, R.F.; Ellis, J.; Bjorkman, C.; Reichel, M.P.; Trees, A.J. Immunization of cattle with live tachyzoites of Neospora caninum confers protection against fetal death. Infect. Immun. 2007, 75, 1343–1348. [Google Scholar] [CrossRef]
  15. Monney, T.; Debache, K.; Hemphill, A. Vaccines against a major cause of abortion in cattle, Neospora caninum infection. Animals 2011, 1, 306–325. [Google Scholar] [CrossRef]
  16. Hemphill, A.; Vonlaufen, N.; Naguleswaran, A. Cellular and immunological basis of the host-parasite relationship during infection with Neospora caninum. Parasitology 2006, 133, 261–278. [Google Scholar] [CrossRef]
  17. Innes, E.A. The host-parasite relationship in pregnant cattle infected with Neospora caninum. Parasitology 2007, 134, 1903–1910. [Google Scholar] [CrossRef]
  18. Lopez-Perez, I.C.; Collantes-Fernandez, E.; Aguado-Martinez, A.; Rodriguez-Bertos, A.; Ortega-Mora, L.M. Influence of Neospora caninum infection in BALB/c mice during pregnancy in post-natal development. Vet. Parasitol. 2008, 155, 175–183. [Google Scholar] [CrossRef]
  19. Lopez-Perez, I.C.; Risco-Castillo, V.; Collantes-Fernandez, E.; Ortega-Mora, L.M. Comparative effect of Neospora caninum infection in BALB/c mice at three different gestation periods. J. Parasitol. 2006, 92, 1286–1291. [Google Scholar] [CrossRef]
  20. Alaeddine, F.; Keller, N.; Leepin, A.; Hemphill, A. Reduced infection and protection from clinical signs of cerebral neosporosis in C57BL/6 mice vaccinated with recombinant microneme antigen NcMIC1. J. Parasitol. 2005, 91, 657–665. [Google Scholar] [CrossRef]
  21. Cannas, A.; Naguleswaran, A.; Muller, N.; Gottstein, B.; Hemphill, A. Reduced cerebral infection of Neospora caninum-infected mice after vaccination with recombinant microneme protein NcMIC3 and ribi adjuvant. J. Parasitol. 2003, 89, 44–50. [Google Scholar] [CrossRef]
  22. Debache, K.; Guionaud, C.; Alaeddine, F.; Mevissen, M.; Hemphill, A. Vaccination of mice with recombinant NcROP2 antigen reduces mortality and cerebral infection in mice infected with Neospora caninum tachyzoites. Int. J. Parasitol. 2008, 38, 1455–1463. [Google Scholar] [CrossRef]
  23. Debache, K.; Alaeddine, F.; Guionaud, C.; Monney, T.; Muller, J.; Strohbusch, M.; Leib, S.L.; Grandgirard, D.; Hemphill, A. Vaccination with recombinant NcROP2 combined with recombinant NcMIC1 and NcMIC3 reduces cerebral infection and vertical transmission in mice experimentally infected with Neospora caninum tachyzoites. Int. J. Parasitol. 2009, 39, 1373–1384. [Google Scholar] [CrossRef]
  24. Monney, T.; Rütti, D.; Schorer, M.; Debache, K.; Grandgirard, D.; Leib, S.L.; Hemphill, A. RecNcMIC3-1-R is a microneme- and rhoptry-based chimeric antigen that protects against acute neosporosis and limits cerebral parasite load in the mouse model for Neospora caninum infection. Vaccine 2011, 29, 6967–6975. [Google Scholar] [CrossRef]
  25. Monney, T.; Debache, K.; Grandgirard, D.; Leib, S.L.; Hemphill, A. Vaccination with the recombinant chimeric antigen recNcMIC3-1-R induces a non-protective Th2-type immune response in the pregnant mouse model for N. caninum infection. Vaccine 2012, 30, 6588–6594. [Google Scholar] [CrossRef]
  26. Grun, J.L.; Maurer, P.H. Different T helper cell subsets elicited in mice utilizing two different adjuvant vehicles: the role of endogenous interleukin 1 in proliferative responses. Cell. Immunol. 1998, 121, 134–145. [Google Scholar] [CrossRef]
  27. Feng, X.; Zhang, N.; Tuo, W. Neospora caninum tachyzoite- and antigen-stimulated cytokine production by bone marrow-derived dendritic cells and spleen cells of naive BALB/c Mice. J. Parasitol 2010, 96, 717–723. [Google Scholar] [CrossRef]
  28. Rojo-Montejo, S.; Collantes-Fernandez, E.; Regidor-Cerrillo, J.; Rodriguez-Bertos, A.; Prenafeta, A.; Gomez-Bautista, M.; Ortega-Mora, L.M. Influence of adjuvant and antigen dose on protection induced by an inactivated whole vaccine against Neospora caninum infection in mice. Vet. Parasitol. 2011, 175, 220–229. [Google Scholar] [CrossRef]
  29. Liddell, S.; Jenkins, M.C.; Collica, C.M.; Dubey, J.P. Prevention of vertical transfer of Neospora caninum in BALB/c mice by vaccination. J. Parasitol. 1999, 85, 1072–1075. [Google Scholar] [CrossRef]
  30. Yamane, I.; Kitani, H.; Kokuho, T.; Shibahara, T.; Haritani, M.; Hamaoka, T.; Shimizu, S.; Koiwai, M.; Shimura, K.; Yokomizo, Y. The inhibitory effect of interferon gamma and tumor necrosis factor alpha on intracellular multiplication of Neospora caninum in primary bovine brain cells. J. Vet. Med. Sci. 2000, 62, 347–351. [Google Scholar] [CrossRef]
  31. Baszler, T.V.; Long, M.T.; McElwain, T.F.; Mathison, B.A. Interferon-gamma and interleukin-12 mediate protection to acute Neospora caninum infection in BALB/c mice. Int. J. Parasitol. 1999, 29, 1635–1646. [Google Scholar] [CrossRef]
  32. Bartley, P.M.; Wright, S.E.; Maley, S.W.; Buxton, D.; Nath, M.; Innes, E.A. The development of immune responses in BALB/c mice following inoculation with attenuated or virulent Neospora caninum tachyzoites. Parasite Immunol. 2009, 31, 392–401. [Google Scholar] [CrossRef]
  33. Debache, K.; Kropf, C.; Schutz, C.A.; Harwood, L.J.; Kauper, P.; Monney, T.; Rossi, N.; Laue, C.; McCullough, K.C.; Hemphill, A. Vaccination of mice with chitosan nanogel-associated recombinant NcPDI against challenge infection with Neospora caninum tachyzoites. Parasite Immunol. 2011, 33, 81–94. [Google Scholar] [CrossRef]
  34. Nishikawa, Y.; Inoue, N.; Makala, L.; Nagasawa, H. A role for balance of interferon-gamma and interleukin-4 production in protective immunity against Neospora caninum infection. Vet. Parasitol. 2003, 116, 175–184. [Google Scholar] [CrossRef]
  35. Kano, R.; Masukata, Y.; Omata, Y.; Kobayashi, Y.; Maeda, R.; Saito, A. Relationship between type 1/type 2 immune responses and occurrence of vertical transmission in BALB/c mice infected with Neospora caninum. Vet. Parasitol. 2005, 129, 159–164. [Google Scholar] [CrossRef]
  36. Quinn, H.E.; Miller, C.M.D.; Ellis, J.T. The cell-mediated immune response to Neospora caninum during pregnancy in the mouse is associated with a bias towards production of interleukin-4. Int. J. Parasitol. 2004, 34, 723–732. [Google Scholar] [CrossRef]
  37. Baszler, T.V.; McElwain, T.F.; Mathison, B.A. Immunization of BALB/c mice with killed Neospora caninum tachyzoite antigen induces a type 2 immune response and exacerbates encephalitis and neurological disease. Clin. Diagn. Lab. Immunol. 2000, 7, 893–898. [Google Scholar]
  38. Long, M.T.; Baszler, T.V.; Mathison, B.A. Comparison of intracerebral parasite load, lesion development, and systemic cytokines in mouse strains infected with Neospora caninum. J. Parasitol. 1998, 84, 316–320. [Google Scholar] [CrossRef]
  39. Collantes-Fernandez, E.; Arrighi, R.B.; Alvarez-Garcia, G.; Weidner, J.M.; Regidor-Cerrillo, J.; Boothroyd, J.C.; Ortega-Mora, L.M.; Barragan, A. Infected dendritic cells facilitate systemic dissemination and transplacental passage of the obligate intracellular parasite Neospora caninum in mice. PLoS One 2012, 7, e32123. [Google Scholar] [CrossRef]
  40. Rosbottom, A.; Gibney, E.H.; Guy, C.S.; Kipar, A.; Smith, R.F.; Kaiser, P.; Trees, A.J.; Williams, D.J. Upregulation of cytokines is detected in the placentas of cattle infected with Neospora caninum and is more marked early in gestation when fetal death is observed. Infect. Immun. 2008, 76, 2352–2361. [Google Scholar] [CrossRef]
  41. Aguado-Martinez, A.; Alvarez-Garcia, G.; Fernandez-Garcia, A.; Risco-Castillo, V.; Marugan-Hernandez, V.; Ortega-Mora, L.M. Failure of a vaccine using immunogenic recombinant proteins rNcSAG4 and rNcGRA7 against neosporosis in mice. Vaccine 2009, 27, 7331–7338. [Google Scholar] [CrossRef]
  42. Jimenez-Ruiz, E.; Alvarez-Garcia, G.; Aguado-Martinez, A.; Salman, H.; Irache, J.M.; Marugan-Hernandez, V.; Ortega-Mora, L.M. Low efficacy of NcGRA7, NcSAG4, NcBSR4 and NcSRS9 formulated in poly-varepsilon-caprolactone against Neospora caninum infection in mice. Vaccine 2012, 30, 4983–4992. [Google Scholar] [CrossRef]
  43. Ribeiro, D.P.; Freitas, M.M.; Cardoso, M.R.; Pajuaba, A.C.; Silva, N.M.; Mineo, T.W.; Silva, J.S.; Mineo, J.R.; Silva, D.A. CpG-ODN combined with Neospora caninum lysate, but not with excreted-secreted antigen, enhances protection against infection in mice. Vaccine 2009, 27, 2570–2579. [Google Scholar] [CrossRef]
  44. Rojo-Montejo, S.; Collantes-Fernandez, E.; Rodriguez-Marcos, S.; Perez-Zaballos, F.; Lopez-Perez, I.; Prenafeta, A.; Ortega-Mora, L.M. Comparative efficacy of immunization with inactivated whole tachyzoites versus a tachyzoite-bradyzoite mixture against neosporosis in mice. Parasitology 2011, 138, 1372–1383. [Google Scholar] [CrossRef]
  45. Debache, K.; Guionaud, C.; Alaeddine, F.; Hemphill, A. Intraperitoneal and intra-nasal vaccination of mice with three distinct recombinant Neospora caninum antigens results in differential effects with regard to protection against experimental challenge with Neospora caninum tachyzoites. Parasitology 2010, 137, 229–240. [Google Scholar] [CrossRef]
  46. Debache, K.; Hemphill, A. Intra-cisternal vaccination induces high-level protection against Neospora caninum infection in mice. Vaccine 2012, 30, 4209–4215. [Google Scholar] [CrossRef]
  47. Dubey, J.P.; Carpenter, J.L.; Speer, C.A.; Topper, M.J.; Uggla, A. Newly recognized fatal protozoan disease of dogs. J. Am. Vet. Med. Assoc. 1988, 192, 1269–1285. [Google Scholar]
  48. Hemphill, A.; Gottstein, B.; Kaufmann, H. Adhesion and invasion of bovine endothelial cells by Neospora caninum. Parasitology 1996, 112, 183–197. [Google Scholar] [CrossRef]
  49. Whitten, M.K. Effect of exteroceptive factors on the oestrous cycle of mice. Nature 1957, 180, 1436. [Google Scholar] [CrossRef]
  50. Müller, N.; Vonlaufen, N.; Gianinazzi, C.; Leib, S.L.; Hemphill, A. Application of real-time fluorescent PCR for quantitative assessment of Neospora caninum infections in organotypic slice cultures of rat central nervous system tissue. J. Clin. Microbiol. 2002, 40, 252–255. [Google Scholar] [CrossRef]
  51. Basto, A.; Piedade, J.; Ramalho, R.; Alves, S.; Soares, H.; Cornelis, P.; Martins, C.; Leitao, A. A new cloning system based on the OprI lipoprotein for the production of recombinant bacterial cell wall-derived immunogenic formulations. J. Biotechnol. 2012, 157, 50–63. [Google Scholar]

Share and Cite

MDPI and ACS Style

Monney, T.; Grandgirard, D.; Leib, S.L.; Hemphill, A. Use of a Th1 Stimulator Adjuvant for Vaccination against Neospora caninum Infection in the Pregnant Mouse Model. Pathogens 2013, 2, 193-208. https://doi.org/10.3390/pathogens2020193

AMA Style

Monney T, Grandgirard D, Leib SL, Hemphill A. Use of a Th1 Stimulator Adjuvant for Vaccination against Neospora caninum Infection in the Pregnant Mouse Model. Pathogens. 2013; 2(2):193-208. https://doi.org/10.3390/pathogens2020193

Chicago/Turabian Style

Monney, Thierry, Denis Grandgirard, Stephen L. Leib, and Andrew Hemphill. 2013. "Use of a Th1 Stimulator Adjuvant for Vaccination against Neospora caninum Infection in the Pregnant Mouse Model" Pathogens 2, no. 2: 193-208. https://doi.org/10.3390/pathogens2020193

Article Metrics

Back to TopTop