Skip to main content

Trichinella spiralis paramyosin activates mouse bone marrow-derived dendritic cells and induces regulatory T cells

Abstract

Background

Dendritic cells (DCs) are antigen-presenting cells that regulate T cell responses for many infectious diseases. The tissue-dwelling nematode Trichinella spiralis expresses paramyosin (TsPmy) not only as a structural protein but also as an immunomodulator to alleviate complement attack by binding to some host complement components. Whether TsPmy is involved in other immunomodulatory pathway and how TsPmy interacts with host DCs is still unknown.

Methods

Mouse bone marrow-derived DCs were incubated with recombinant TsPmy (rTsPmy) for activation. Maturation of DC was determined by the expression of surface markers CD40, CD80, CD86 and MHCII. The rTsPmy-pulsed DCs were co-incubated with T. spiralis-sensitized or naïve mouse CD4+ T cells to observe their activation on T cells and polarizing regulatory T cells using flow cytometry. Cytokines were measured by enzyme-linked immunosorbent assays (ELISA).

Results

TsPmy was able to activate mouse bone marrow-derived DCs to semi-mature status characterized by expressing surface CD40 and CD86, but not CD80 and MHCII. The semi-mature TsPmy-pulsed DCs were able to stimulate T. spiralis-sensitized CD4+ T cells to proliferate. Incubation of TsPmy-pulsed DCs with naïve CD4+ splenocytes polarized the latter to CD4+CD25+Foxp3+ regulatory T cells. However, mice immunized with rTsPmy only induce the CD4+CD25−Foxp3+ T cell population, associated with high level of IL-10, TGF-β and IL-17A.

Conclusions

During T. spiralis infection, TsPmy plays an important role in modulating the host immune system by stimulating DCs to differentiate the CD4+ T cells to regulatory T cells, in addition to binding to components of the host complement cascade, as survival strategies to live in host.

Background

Trichinellosis is a serious zoonotic parasitic disease caused by the infection of Trichinella spiralis through ingestion of meat contaminated with infective larvae. It is estimated that more than 11 million people could be infected with T. spiralis worldwide [1] and heavy infection can even causes death [2]. Trichinella spiralis is a tissue-dwelling parasitic nematode. During T. spiralis infection, the entire life-cycle is completed within the same host. After being ingested, the infective muscle larvae develop to adult worms in the host intestine. The newborn larvae are released from sexually-mature adult worms and soon migrate to skeletal muscles to form encysted muscle larvae that may live for several years and cause a chronic infection [3]. How the Trichinella parasite maintains the chronic infection within the host without being recognized and attacked by the host’s immune system remains unknown [4]. Understanding the mechanism underlying the immune evasion would greatly benefit the design of preventive/therapeutic vaccines or drugs to control the infection.

Paramyosin is not only a fibrillar protein exclusively found in invertebrates, but also a functional protein expressed on the surface of many helminths [5–7] that plays an important role as an immunomodulatory molecule to defend against host immune attack [8–10]. Paramyosin of T. spiralis (TsPmy) was cloned from the adult T. spiralis in a previous study [11]. Subsequent studies have identified that TsPmy binds to host complement components C8, C9 and C1q that interferes with the forming of complement membrane attack complex and protects parasite from being attacked by the host innate immune system [12–15]. Partial protective immunity against T. spiralis larval challenge was determined in BALB/c mice immunized with recombinant TsPmy (rTsPmy) [16] and protective epitopes [17–19] or through RNAi [20]. Except for interfering with host complement system, whether TsPmy is involved in other immunomodulatory function is unknown.

Dendritic cells (DCs) are antigen presenting cells that play a pivotal role in the control and modulation of immune responses by initiating T cell responses and producing cytokines and other molecules that regulate adaptive immunity [21]. How TsPmy interacts with DC and subsequently impacts DC activation and function during T. spiralis infection is not understood.

In this study, we investigated the roles of TsPmy on DCs maturation and subsequent T cell polarization. The study herein demonstrated that TsPmy could activate bone marrow-derived mouse DCs and consequently promote the differentiation of CD4+ T cells to regulatory T cells (Tregs). The induction of Tregs by TsPmy through activated DCs during T. spiralis infection may inhibit the host immune response and play an important role in the survival of T. spiralis in infected host.

Methods

Experimental animals

Specific pathogen-free 6–8 week-old female BALB/c mice were purchased from the Laboratory Animal Services Center of the Capital Medical University (Beijing, China) and housed under specific pathogen-free conditions with humidity and temperature controlled (temperature of 20 ± 2 °C; humidity of 70 ± 10 %). All animal protocols and husbandry were approved by Capital Medical University Institutional Animal Care and Use Committee (IACUC).

Parasites and experimental infection

The ISS 533 strain of T. spiralis was maintained in female ICR mice. Muscle larvae (ML) were received from the muscles of infected mice by previously described method of modified pepsin-hydrochloric acid digestion [17]. BALB/c mice were infected with 400 infective T. spiralis ML by oral gavage and immunized with recombinant TsPmy (rTsPmy) as described below.

Antigen preparation

The recombinant TsPmy with a His-tag at C-terminus was expressed in Baculovirus/insect cell Sf9 (Invitrogen, Carlsbad, CA, USA) and purified with Ni-affinity chromatography (Qiagen, Valencia, CA, USA). Lipopolysaccharide (LPS) (Sigma-Aldrich, St. Louis, MO, USA) was used as a positive control for immune response. Sf9 cell lysis proteins were used as non-relevant proteins control. All antigens were stored at -80 °C.

Generation of dendritic cells

DCs were generated from mouse bone marrow cells as described [22]. Briefly, bone marrow cells were obtained from BALB/c mice and cultivated in RPMI 1640 medium (Hyclone, Logan, UT, USA) supplemented with 10 % foetal bovine serum (FBS; Thermo Fisher, Life Technologies, Carlsbad, CA, USA) and penicillin/streptomycin at 37 °C, 5 % CO2 for 3 h. After removing the suspended cells, the remaining adherent cells were cultured in RPMI 1640 medium containing growth factors of 10 ng/ml recombinant GM-CSF and 2 ng/ml IL-4 (Prospec, Rehovot, Israel) and 10 % FBS for 6 days with replenishment of the same culture medium on Day 3 and Day 5. The immature DCs were harvested on Day 6 for further experiments.

In vitro DC activation

The immature DCs produced above were stimulated with rTsPmy (10 μg/ml), LPS (2 μg/ml) or PBS respectively in vitro for 48 h. The stimulated cells were stained with APC-conjugated monoclonal antibody (mAb) to CD11c, the major marker of mature DCs [23], and PE-conjugated mAbs to major histocompatibility complex II (MHCII), CD40, CD80 or CD86 respectively (BD Biosciences, San Jose, CA, USA). The cytokine levels in the culture supernatants were determined by the corresponding enzyme-linked immunosorbent assay (ELISA) kits according to the manufacturer’s instructions (Dakewe Biotech, Shenzhen, China).

Co-incubation of rTsPmy pulsed DCs with T. spiralis-sensitized CD4+ T cells

To determine whether rTsPmy-pulsed DCs could activate T. spiralis-sensitized CD4+ T cells, the DCs pulsed with rTsPmy for 72 h, and then were co-cultivated with T. spiralis-sensitized CD4+ T cells, the T. spiralis-sensitized CD4+ T cells were obtained from the spleens of BALB/c mice infected with 400 T. spiralis ML for 60 days using magnetic-activated cell sorting (MACS) with a mouse CD4+ T cell isolation kit (Miltenyi Biotec, Bergisch Gladbach, Germany). A total of 5 × 104 or 2.5 × 104 DCs were plated in each well of round-bottom 96-well plates and then co-cultivated with 5 × 105 T. spiralis-sensitized CD4+ T cells stained with 5-and 6-carboxyfluorescein diacetate succinimidyl ester (CFSE) (eBioscience, San Diego, CA, USA), in the presence of 5 μg/ml Concavalin-A (Con-A) (Sigma-Aldrich, St. Louis, MO, USA) which is a nonspecific stimulator for mouse T cells. Subsequently, the proliferation of T cells was measured by fluorescence-activated cell sorting (FACS).

To determine the cytokine production, 5 × 104 DCs were plated in each well of round-bottom 96-well plates and co-incubated with 5 × 105 T. spiralis-sensitized CD4+ T cells for 36 h, then supernatants were collected and cytokines measured by ELISA as described above.

rTsPmy pulsed DCs/naïve CD4+ T cells co-incubation

To assess the ability of rTsPmy-pulsed DCs on naïve T cell polarization, DCs were stimulated with rTsPmy for 72 h, and the naïve CD4+ T cells were isolated from the spleens of healthy BALB/c mice by MACS using mouse CD4+ T cell isolation kit. Total 5 × 104 DCs were plated in each well of round-bottom 96-well plates and co-incubated with 5 × 105 naïve CD4+ T cells. The co-incubated DCs/naïve T cells were stimulated with 5 μg/ml plate-bound anti-CD3/anti-CD28 (Peprotech, NJ, USA) which delivers signal one and a costimulatory signal two without leading to early cell death for proliferated cells [24]. The co-incubation was continued at 37 °C for 36 h and cells were recovered for detecting the percentage of CD4+CD25+Foxp3+ T cells. Meanwhile, the co-incubation supernatants were collected for detection of cytokines level by ELISA as described above.

T cell response primed by rTsPmy in vivo

BALB/c mice were divided into 3 groups with 4 mice each, and each group was immunized intraperitoneally with 100 μg of rTsPmy or the same amount of Sf9 insect cell protein as a non-relevant protein control twice at 2 weeks intervals. Another group of 4 mice were given PBS only. Fourteen days after the final immunization, all mice were sacrificed and the splenocytes were harvested for the analysis of cytokine production and the presence of Th17 cells and Tregs.

For FACS analysis of Th17 cells, the harvested splenocytes were stimulated with 25 ng/ml phorbol-12-myristate-13-acetate (PMA, Sigma-Aldrich, St. Louis, MO, USA), 1 μg/ml ionomycin (Sigma-Aldrich, St. Louis, MO, USA) and 0.66 μl/ml Golgistop™ (BD Biosciences, San Jose, CA, USA) for 6 h before cell staining with anti-IL-17A-PE-Cyanine7 (eBioscience, San Diego, CA, USA). The culture supernatants were recovered for measuring cytokine release as described above. For detection of Tregs, the harvested splenocytes were directly stained with Mouse Regulatory T Cell Staining Kit #1 according to the manufacturer’s instructions (eBioscience, San Diego, CA, USA).

Statistical analysis

GraphPad Prism version 6 software (San Diego, CA, USA) was used to analyze statistical data. The results are presented as mean ± standard deviation. Statistical significance was determined by one-way ANOVA with Dunnett or Tukey’s post-hoc analysis; P < 0.05 was considered statistically significant.

Results

Semi-maturation of DCs after rTsPmy stimulation

FACS data demonstrated that both rTsPmy and LPS (positive control) significantly upregulated the expression of CD40 and CD86 on stimulated CD11c+ DCs compared to PBS control (rTsPmy vs PBS: CD40, t D(6) = 2.963, P = 0.044; CD86, t D(6) = 3.106, P = 0.037; LPS vs PBS, CD40, t D(6) = 3.547, P = 0.021; CD86, t D(6) = 4.213, P = 0.01) (Fig. 1). However, rTsPmy and LPS did not stimulate the expression of CD80 and MHCII on CD11c+ DCs (CD80, F (2,6) = 1.209, P = 0.362; MHCII, F (2,6) = 0.6119, P = 0.573). These results indicate that rTsPmy was able to stimulate the BMDCs to semi-mature status.

Fig. 1
figure 1

Expression of surface markers on DCs pulsed with rTsPmy. DCs were pulsed with PBS, rTsPmy or LPS for 48 h. The surface markers CD40, CD80, CD86 and MHCII were sorted by FACS (a). The percentage of each surface marker expression on DCs stimulated with the indicated antigens (b). Data represent means ± standard deviations of the results from three individual experiments. *P < 0.05 compared to PBS control

Detection of cytokine production of DCs response to rTsPmy

To further investigate if rTsPmy stimulates DCs to secrete Th1, Th2, Th17 and regulatory cytokines, IL-1β, IL-5, IL-6, IL-10, IL-17A, IL-12p70, IFN-γ, TNF-α and TGF-β were detected in culture supernatants of antigen-stimulated DCs. Compared to the PBS control, IL-1β, IL-6, IL-12p70, IFN-γ, TNF-α and TGF-β were significantly elevated following rTsPmy stimulation (IL-1β, t D(6) = 24.95, P < 0.001; IL-6, t D(6) = 27.28, P < 0.001; IL-12p70, t D(6) = 15.02, P < 0.001; IFN-γ, t D(6) = 12.55, P < 0.001; TNF-α, t D(6) = 51.19, P < 0.001; TGF-β, t D(6) = 14.13, P < 0.001), indicating a mixed Th1/Th2/Treg responses. There was no change in the secretion of IL-5, IL-10 and IL-17A in the supernatants of rTsPmy-stimulated DCs (Fig. 2). As a positive control, LPS stimulated secretion of all detected cytokines.

Fig. 2
figure 2

Cytokine production by DCs stimulated with rTsPmy. DCs were cultured with rTsPmy, LPS or PBS for 48 h. Culture supernatants were then collected and the levels of different cytokines were determined by ELISA. Results are presented as the mean ± standard deviation from three individual experiments. *P < 0.05; ***P < 0.001 compared to PBS control

rTsPmy-pulsed DCs activate T. spiralis-sensitized CD4+ T cells

To assess whether rTsPmy-pulsed DCs could activate T. spiralis-sensitized CD4+ T cells, DCs pulsed with rTsPmy for 72 h were co-cultivated with T. spiralis-sensitized CD4+ T cells from T. spiralis-infected mice for 72 h in the presence of Con-A. FACS results revealed that T. spiralis-sensitized CD4+ T cells were significantly induced by rTsPmy-pulsed DCs with a significantly higher proliferation rate compared to PBS incubated DCs (20 folds: q (6) = 14.83, P < 0.001; 10 folds: q (6) = 20.9, P < 0.001) (Fig. 3a, b). LPS-pulsed DCs also boosted T cell proliferation at a lower rate than that induced by rTsPmy-pulsed DCs.

Fig. 3
figure 3

Proliferation and cytokines secretion of T. spiralis-sensitized CD4+ T cells co-incubated with rTsPmy-pulsed DCs. To assess whether rTsPmy-pulsed DCs enable to stimulate T. spiralis-sensitized CD4+ T cells, the CD4+ T cells from splenocytes of T. spiralis infected BALB/c mice were incubated with rTsPmy-, LPS- or PBS-treated DCs and the proliferation of co-incubated T cells were determined by CFSE labeling and FACS (a, b). The levels of IFN-γ, IL-4, IL-10, TGF-β and IL-17A in the culture supernatants were measured by ELISA (c). Data are presented as the mean ± standard deviation from three individual experiments. *P < 0.05; **P < 0.01; ***P < 0.001

Cytokine profiling demonstrated that T. spiralis-sensitized CD4+ T cells secreted higher level of IL-4, IL-10, TGF-β and IL-17A when being incubated with rTsPmy-pulsed DCs compared to cells incubated with LPS or PBS treated DCs (rTsPmy vs PBS: IL-4, t D(6) = 3.367, P = 0.027; IL-10, t D(6) = 9.988, P < 0.001; TGF-β, t D(6) = 10.92, P < 0.001; IL-17A, t D(6) = 13.38, P < 0.001) (Fig. 3c). However, rTsPmy-pulsed DCs did not stimulate the secretion of IFN-γ in T. spiralis-sensitized T cells (t D(6) = 1.227, P = 0.417). The cytokine profile results support that helminth infections generally polarize the T cell response towards Th2, while IL-10 and TGF-β might suppress Th1 response and therefore inhibit the production of IFN-γ. The results of proliferation and cytokine profiling revealed that TsPmy-pulsed DCs are able to activate T cells or boost the memory T cells with Th2 and Treg-related cytokine responses. LPS-stimulated DCs only induced some level of IFN-γ in CD4+ T. spiralis-sensitized T cells.

rTsPmy-pulsed DCs induces naïve T cells to polarize to Tregs

In order to determine whether rTsPmy-pulsed DCs induce naïve CD4+ T cell polarization, the rTsPmy-treated DCs were incubated with naïve T cells isolated from spleens of normal BALB/c mice for 36 h. The FACS results demonstrated that the CD4+CD25+Foxp3+ T cell population was significantly elevated in naïve T cells co-cultivated with rTsPmy-pulsed DCs compared to PBS-treated DCs (t D(6) = 4.333, P = 0.009) (Fig. 4a, b), while LPS-pulsed DCs did not obviously affected the population of CD4+CD25+Foxp3+ T cells (t D(6) = 0.8826, P = 0.608).

Fig. 4
figure 4

Induction of CD4+CD25+ Foxp3+ T cell population and Treg cytokines in naïve T cells when incubated with rTsPmy-pulsed DCs. The splenocytes isolated from naïve BALB/c mice were incubated with rTsPmy-, LPS- or PBS-treated DCs for 36 h, then labeled with anti-CD4-FITC and anti-CD25-APC and anti-Foxp3-PE for FACS plot analysis (a). The percentage of CD4+CD25+Foxp3+ cells in CD4+ T cell population was showed in b. c Cytokine profile of naïve CD4+ T cells incubated with rTsPmy-, LPS- or PBS-treated DCs. The co-incubation supernatants were recovered and measured for secretions of IFN-γ, IL-4, IL-10, TGF-β and IL-17A by ELISA. Data are presented as the mean ± standard deviation from three individual experiments. *P < 0.05; **P < 0.01; ***P < 0.001 compared to PBS control

In parallel, the culture supernatants were measured for IFN-γ,IL-4, IL-10, TGF-β and IL-17A secretion. The results revealed that rTsPmy-pulsed DCs not only induced naïve T cells to secrete cytokine IFN-γ (Th1), IL-4 (Th2) and IL-17A (Th17) (IFN-γ, t D(6) = 11.23, P < 0.001; IL-17A, t D(6) = 14.68, P < 0.001; IL-4, t D(6) = 6.626, P = 0.001), but also stimulated high levels of cytokines IL-10 and TGF-β secreted mostly by Tregs (IL-10, t D(6) = 32.71, P < 0.001; TGF-β, t D(6) = 4.211, P = 0.01) (Fig. 4c), which is consistent with the increase of Treg population observed by FACS. LPS-pulsed DCs also stimulated naïve T cells to secrete IFN-γ, IL-10 and IL-17A, but the level was not as high as that induced by rTsPmy-pulsed DCs.

rTsPmy induces Treg in immunized mice

To confirm if TsPmy enables to induce Treg in vivo, BALB/c mice were immunized with rTsPmy and the CD4+CD25+Foxp3+ T cells were sorted from splenocytes of immunized mice. The FACS results demonstrated that rTsPmy immunization did not increase the population of CD4+CD25+Foxp3+ T cells compared to PBS or non-relevant Sf9 protein control groups, however, CD4+CD25−Foxp3+ T cells were upregulated (rTsPmy vs PBS: t D(9) = 3.005, P = 0.027). Interestingly, the Th17 cells were also upregulated in rTsPmy immunized mice (rTsPmy vs PBS: t D(9) = 3.402, P = 0.014) (Fig. 5a, b). As we know Th17 cells are able to convert T cells into Tregs in mesenchymal stem cell-mediated allograft survival [25].

Fig. 5
figure 5

Tregs and Th17 cell differentiation and cytokines production in splenocytes of mice immunized with rTsPmy. Splenocytes were isolated from mice immunized with rTsPmy or Sf9 protein and sorted for CD4, CD25, Foxp3 and Th17 by FACS (a, b). The different cytokines in splenocyte culture were determined by ELISA (c). Data (means ± standard deviations) are representative of three independent experiments. *P < 0.05; **P <0.01; ***P <0.001 compared to PBS or Sf9 control as indicated

To further investigate the cytokine profile secreted by splenocytes of rTsPmy immunized mice, splenocytes from immunized mice were isolated and stimulated with PMA/ionomycin. The different cytokine level in the culture supernatants was detected by ELISA. Results showed IL-4, IL-10, TGF-β and IL-17A levels were significantly elevated in cultures of splenocytes from mice immunized with rTsPmy compared to PBS (IL-4, t D(9) = 7.482, P < 0.001; IL-10, t D(6) = 5.507, P = 0.003; TGF-β, t D(9) = 6.55, P < 0.001; IL-17A, t D(10) = 10.01, P < 0.001) or non-relevant protein (Sf9) injection control groups (IL-4, t D(9) = 6.696, P < 0.001; IL-10, t D(6) = 3.964, P = 0.013; TGF-β, t D(9) = 6.554, P < 0.001; IL-17A, t D(10) = 10.38, P < 0.001) (Fig. 5c). There was no change in IFN-γ level (F (2,7) = 2.162, P = 0.186). The secretions of regulatory cytokine IL-10, TGF-β are consistent with the differentiation of CD4+CD25−Foxp3+ Tregs.

Discussion

During pathogen infections, DCs play a critical role in the induction and orchestration of immune responses. The infection itself induces DC activity and maturation through various families of pattern recognition receptors (PRRs) such as Toll-like receptors [26, 27], and produces different cytokines to prime distinct types of adaptive immune responses [28]. Therefore, DC responses are crucial to control and eliminate the invading pathogens during infection [29]. However, the specific mechanism during interaction between DC and helminthic antigens, especially in the role of helminth immune evasion, are still not quite understood.

We have identified that TsPmy is a strong immunomodulator by interfering with host complement functions as a strategy to evade host innate immune attack in our previous studies [12–15]. In this study, we found that rTsPmy enabled to stimulate mouse bone marrow-derived DCs to express CD40, CD86, but not CD80 and MHCII, on the surface of CD11c+ DCs. rTsPmy-pusled DCs secreted high level of IL-1β, IL-6, IL-12p70, IFN-γ, TNF-α and TGF-β, but not for IL-5, IL-10 and IL-17A, indicating rTsPmy induces DCs to a semi-mature status in vitro to secrete a mix Th1/Th2/Treg response in cytokine expression. Our results are consistent with other studies that showed parasitic helminth antigens activated DCs to incomplete maturation [22, 30, 31].

When incubating with T. spiralis-infected mouse splenocytes, the rTsPmy-pulsed DCs stimulated the T. spiralis-infected mouse CD4+ cells to proliferate (Fig. 3a, b), but did not strongerly stimulate the naïve mouse CD4+ cells to proliferate (data not shown), indicating rTsPmy-activated DCs was able to present TsPmy antigen to T cells previously exposed to T. spiralis infection to activate the TsPmy memory cells. The cytokine profile also confirmed that rTsPmy-pulsed DCs stimulated T. spiralis-infected mouse CD4+ cells to secrete IL-4, IL-10, TGF-β and IL-17A, but not IFN-γ, consistent with the Th2-skewed immune response induced by helminth infections [32].

Evasion of host adaptive immunity is key strategy for the survival of parasites in the hostile environment within the host [33, 34]. Many studies have demonstrated both helminths and protozoans create more permissive environments for surviving in hosts by interfering with DCs activity [29, 35]. Helminths and their products have been shown to suppress immune response of the host by inducing a regulatory network. DCs play a crucial role in this regulatory network, as they can regulate T cell-mediated effector responses by generating anti-inflammatory cytokines that can lead to induction of regulatory T cells [36] and promote parasite immune escape by inhibiting parasite-specific immune responses [37]. In order to determine if TsPmy possesses the same immunomodulatory ability to induce tolerogenic DCs so as to stimulate the host T cell regulatory network, the rTsPmy was incubated with DCs and the rTsPmy-pulsed DCs were co-incubated with naïve mouse CD4+ T cells. The results showed that TsPmy-pulsed DCs enabled to induce CD4+CD25+Foxp3+ Treg cells in vitro associated with higher level of IL-10 and TGF-β, the cytokines mostly secreted by Tregs [38]. Our results confirmed that TsPmy was able to stimulate tolerogenic DCs that subsequently induce Treg cells to modulate host immune response, possibly through signal passage from DC to T cells or TsPmy-pulsed DC-secreted Th1/Th2/Treg cytokines (Fig. 2). Even though rTsPmy only induced bone marrow-derived DCs to a semi-mature status, it did not affect their abilities to induce Treg cells, confirming that semi-mature DCs also induce tolerance [39, 40]. Our results are consistent with other investigations that showed T. spiralis excretory-secretory antigen-stimulated dendritic cells alleviated experimental autoimmune encephalomyelitis or DSS-induced colitis through inducing Treg that increased the secretion of IL-4, IL-10 and TGF-β [4, 41, 42]. However, rTsPmy immunization only induced CD4+CD25−Foxp3+ T cells, not CD4+CD25+Foxp3+ T cells, in immunized mice. There could be an explanation that experiments represent the situation simplified in vitro, differ from originally existing in live infection. It is possible that rTsPmy stimulates the Tregs through multiple channels in vivo except for inducing tolerogenic DCs (such as CTLA-4, TGF-β, IL-10, and GITR). Naïve T cells can be converted to a Treg phenotype by culture with CTLA-4-Ig [43]. IL-6 can convert CD4+CD25+Foxp3+ Tregs but not CD4+CD25−Foxp3+ Tregs to Th17 cells [44]. The reasons why CD4+CD25−Foxp3+ Tregs, not CD4+CD25+Foxp3+ Tregs were induced with rTsPmy immunization need to be further studied. Nevertheless, Foxp3 expression, rather than CD25 expression is essential for Treg’s activity [45]. CD4+CD25−Foxp3+ T cells also showed suppressive activity [45, 46]. Actually, during T. spiralis chronic infection, CD4+CD25− effector T cells control inflammation, rather than CD4+CD25+ Tregs [47]. It was an interesting finding that rTsPmy immunization in vivo generated CD4+CD25−Foxp3+ Tregs that is different from in vitro stimulation of CD4+CD25+Foxp3+ Tregs via inducing tolerogenic DCs.

In addition, in this study we identified that rTsPmy-pulsed DCs induced T. spiralis-infected mouse CD4+ or naïve CD4+ T cells to produce high level of IL-17A. Mice immunized with rTsPmy in vivo also induced the generation of Th17 cells. Even though Th17 cells have been considered to be pro-inflammatory and induce autoimmunity [44], the generation of Th17 cells during Schistosoma japonicum infection in C57BL/6 mice has determined to induce suppressive immunity to schistosome infection [48]. Interestingly, some Foxp3+ Treg cells could convert to IL-17+ T cells upon co-culture with dendritic cells selectively activated by dectin-1, a C-type lectin receptor involved in fungal recognition [49]. The conversion of Treg cells into Th17 cells may help restrain infections with specific fungi or other pathogens [50]. The flexibility between induced regulatory T cells and Th17 cells may affect the differentiation of CD4+ T cells and therefore may alter the direction of immune response [44, 51]. However, the relationship between the Treg and Th17 responses in T. spiralis infection remains unclear.

Together with our previous studies, our results further suggest the immunomodulatory function of T. spiralis paramyosin, which interacts with dendritic cells and stimulates regulatory T cells and Th17 cells. The data further support that TsPmy plays an important role in the immunomodulation of host immune response as a survival strategy, and is therefore a good candidate for vaccine development against trichinellosis. It is also possible to use rTsPmy as a therapeutic reagent for autoimmune or allergic diseases by taking advantage of its stimulating regulatory network of the immune system.

Conclusions

Our results showed that TsPmy is able to activate mouse bone marrow-derived DCs to semi-mature status characterized by expressing CD40 and CD86, without CD80 and MHCII on the surface of CD11c+ DCs. The semi-matured TsPmy-pulsed DCs were able to stimulate T. spiralis-sensitized CD4+ T cells to proliferate associated with the secretion of IL-10 and TGF-β produced mostly by Treg cells. Incubation of TsPmy-pulsed DCs with naïve CD4+ splenocytes polarized the latter to CD4+CD25+Foxp3+ Tregs. However, mice immunized with rTsPmy only induce the CD4+CD25−Foxp3+ T cell population, associated with high level of IL-10 and TGF-β. rTsPmy also induced Th17 response, possibly converted from Foxp3+ Tregs. During T. spiralis infection, TsPmy plays an important role in modulating the host immune system by stimulating DCs to promote differentiation of regulatory T cells, in addition to binding to components of the host complement cascade, as survival strategies to live in host.

Abbreviations

BMDCs:

Mouse bone marrow-derived DC cells

CFSE:

5- and 6-carboxyfluorescein diacetate succinimidyl ester

Con-A:

Concavalin-A

DCs:

Dendritic cells

ELISA:

Enzyme-linked immunosorbent assay

FACS:

Fluorescence-activated cell sorting

IACUC:

Institutional Animal Care and Use Committee

LPS:

Lipopolysaccharide

mAb:

Monoclonal antibody

MACS:

Magnetic-activated cell sorting

MHCII:

Major histocompatibility complex II

ML:

Muscle larvae

PMA:

Phorbol-12-myristate-13-acetate

PRRs:

Pattern recognition receptors

rTsPmy:

Recombinant TsPmy

Tregs:

Regulatory T cells

TsPmy:

Trichinella spiralis paramyosin

References

  1. Dupouy-Camet J. Trichinellosis: a worldwide zoonosis. Vet Parasitol. 2000;93(3–4):191–200.

    Article  CAS  PubMed  Google Scholar 

  2. Wilson NO, Hall RL, Montgomery SP, Jones JL. Trichinellosis surveillance - United States, 2008-2012. MMWR Surveill Summ. 2015;64(1):1–8.

  3. Yang J, Pan W, Sun X, Zhao X, Yuan G, Sun Q, et al. Immunoproteomic profile of Trichinella spiralis adult worm proteins recognized by early infection sera. Parasit Vectors. 2015;8:20. doi:10.1186/s13071-015-0641-8.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Yang X, Yang Y, Wang Y, Zhan B, Gu Y, Cheng Y, et al. Excretory/secretory products from Trichinella spiralis adult worms ameliorate DSS-induced colitis in mice. PLoS One. 2014;9(5):e96454. doi:10.1371/journal.pone.0096454.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Cancela M, Carmona C, Rossi S, Frangione B, Goni F, Berasain P. Purification, characterization, and immunolocalization of paramyosin from the adult stage of Fasciola hepatica. Parasitol Res. 2004;92(6):441–8. doi:10.1007/s00436-003-1059-3.

    Article  PubMed  Google Scholar 

  6. Loukas A, Jones MK, King LT, Brindley PJ, McManus DP. Receptor for Fc on the surfaces of schistosomes. Infect Immun. 2001;69(6):3646–51. doi:10.1128/IAI.69.6.3646-3651.2001.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Muhlschlegel F, Sygulla L, Frosch P, Massetti P, Frosch M. Paramyosin of Echinococcus granulosus: cDNA sequence and characterization of a tegumental antigen. Parasitol Res. 1993;79(8):660–6.

    Article  CAS  PubMed  Google Scholar 

  8. Gazarian KG, Solis CF, Gazarian TG, Rowley M, Laclette JP. Synthetic peptide-targeted selection of phage display mimotopes highlights immunogenic features of alpha-helical vs non-helical epitopes of Taenia solium paramyosin: implications for parasite- and host-protective roles of the protein. Peptides. 2012;34(1):232–41. doi:10.1016/j.peptides.2011.10.003.

    Article  CAS  PubMed  Google Scholar 

  9. Deng J, Gold D, LoVerde PT, Fishelson Z. Inhibition of the complement membrane attack complex by Schistosoma mansoni paramyosin. Infect Immun. 2003;71(11):6402–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Wang X, Chen W, Lv X, Tian Y, Men J, Zhang X, et al. Identification and characterization of paramyosin from cyst wall of metacercariae implicated protective efficacy against Clonorchis sinensis infection. PLoS One. 2012;7(3):e33703. doi:10.1371/journal.pone.0033703.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Yang J, Yang Y, Gu Y, Li Q, Wei J, Wang S, et al. Identification and characterization of a full-length cDNA encoding paramyosin of Trichinella spiralis. Biochem Biophys Res Commun. 2008;365(3):528–33. doi:10.1016/j.bbrc.2007.11.012.

    Article  CAS  PubMed  Google Scholar 

  12. Zhang Z, Yang J, Wei J, Yang Y, Chen X, Zhao X, et al. Trichinella spiralis paramyosin binds to C8 and C9 and protects the tissue-dwelling nematode from being attacked by host complement. PLoS Negl Trop Dis. 2011;5(7):e1225. doi:10.1371/journal.pntd.0001225.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Zhao X, Hao Y, Yang J, Gu Y, Zhu X. Mapping of the complement C9 binding domain on Trichinella spiralis paramyosin. Parasit Vectors. 2014;7:80. doi:10.1186/1756-3305-7-80.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Hao Y, Zhao X, Yang J, Gu Y, Sun R, Zhu X. Monoclonal antibody targeting complement C9 binding domain of Trichinella spiralis paramyosin impairs the viability of Trichinella infective larvae in the presence of complement. Parasit Vectors. 2014;7:313. doi:10.1186/1756-3305-7-313.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Sun R, Zhao X, Wang Z, Yang J, Zhao L, Zhan B, et al. Trichinella spiralis paramyosin binds human complement C1q and inhibits classical complement Activation. PLoS Negl Trop Dis. 2015;9(12):e0004310. doi:10.1371/journal.pntd.0004310.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Yang J, Gu Y, Yang Y, Wei J, Wang S, Cui S, et al. Trichinella spiralis: immune response and protective immunity elicited by recombinant paramyosin formulated with different adjuvants. Exp Parasitol. 2010;124(4):403–8. doi:10.1016/j.exppara.2009.12.010.

    Article  CAS  PubMed  Google Scholar 

  17. Gu Y, Wei J, Yang J, Huang J, Yang X, Zhu X. Protective immunity against Trichinella spiralis infection induced by a multi-epitope vaccine in a murine model. PLoS One. 2013;8(10):e77238. doi:10.1371/journal.pone.0077238.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Wei J, Gu Y, Yang J, Yang Y, Wang S, Cui S, et al. Identification and characterization of protective epitope of Trichinella spiralis paramyosin. Vaccine. 2011;29(17):3162–8. doi:10.1016/j.vaccine.2011.02.072.

    Article  CAS  PubMed  Google Scholar 

  19. Gu Y, Huang J, Wang X, Wang L, Yang J, Zhan B, et al. Identification and characterization of CD4+ T cell epitopes present in Trichinella spiralis paramyosin. Vet Parasitol. 2016; doi: 10.1016/j.vetpar.2016.06.022.

  20. Chen X, Yang Y, Yang J, Zhang Z, Zhu X. RNAi-mediated silencing of paramyosin expression in Trichinella spiralis results in impaired viability of the parasite. PLoS One. 2012;7(11):e49913. doi:10.1371/journal.pone.0049913.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Aranzamendi C, Fransen F, Langelaar M, Franssen F, van der Ley P, van Putten JP, et al. Trichinella spiralis-secreted products modulate DC functionality and expand regulatory T cells in vitro. Parasite Immunol. 2012;34(4):210–23. doi:10.1111/j.1365-3024.2012.01353.x.

    Article  CAS  PubMed  Google Scholar 

  22. Ilic N, Worthington JJ, Gruden-Movsesijan A, Travis MA, Sofronic-Milosavljevic L, Grencis RK. Trichinella spiralis antigens prime mixed Th1/Th2 response but do not induce de novo generation of Foxp3+ T cells in vitro. Parasite Immunol. 2011;33(10):572–82. doi:10.1111/j.1365-3024.2011.01322.x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Daro E, Pulendran B, Brasel K, Teepe M, Pettit D, Lynch DH, et al. Polyethylene glycol-modified GM-CSF expands CD11b(high)CD11c(high) but notCD11b(low)CD11c(high) murine dendritic cells in vivo: a comparative analysis with Flt3 ligand. J Immunol. 2000;165(1):49–58.

    Article  CAS  PubMed  Google Scholar 

  24. Li Y, Kurlander RJ. Comparison of anti-CD3 and anti-CD28-coated beads with soluble anti-CD3 for expanding human T cells: differing impact on CD8 T cell phenotype and responsiveness to restimulation. J Transl Med. 2010;8:104. doi:10.1186/1479-5876-8-104.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Obermajer N, Popp FC, Soeder Y, Haarer J, Geissler EK, Schlitt HJ, et al. Conversion of Th17 into IL-17A(neg) regulatory T cells: a novel mechanism in prolonged allograft survival promoted by mesenchymal stem cell-supported minimized immunosuppressive therapy. J Immunol. 2014;193(10):4988–99. doi:10.4049/jimmunol.1401776.

    Article  CAS  PubMed  Google Scholar 

  26. Terrazas CA, Alcantara-Hernandez M, Bonifaz L, Terrazas LI, Satoskar AR. Helminth-excreted/secreted products are recognized by multiple receptors on DCs to block the TLR response and bias Th2 polarization in a cRAF dependent pathway. FASEB J. 2013;27(11):4547–60. doi:10.1096/fj.13-228932.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Hu W, Jain A, Gao Y, Dozmorov IM, Mandraju R, Wakeland EK, et al. Differential outcome of TRIF-mediated signaling in TLR4 and TLR3 induced DC maturation. Proc Natl Acad Sci U S A. 2015;112(45):13994–9. doi:10.1073/pnas.1510760112.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Reis e Sousa C, Diebold SD, Edwards AD, Rogers N, Schulz O, Sporri R. Regulation of dendritic cell function by microbial stimuli. Pathol Biol (Paris). 2003;51(2):67–8.

    Article  CAS  Google Scholar 

  29. Terrazas CA, Terrazas LI, Gomez-Garcia L. Modulation of dendritic cell responses by parasites: a common strategy to survive. J Biomed Biotechnol. 2010;2010:357106.

  30. van Riet E, Hartgers FC, Yazdanbakhsh M. Chronic helminth infections induce immunomodulation: consequences and mechanisms. Immunobiology. 2007;212(6):475–90.

  31. Ilic N, Colic M, Gruden-movsesijan A, Majstorovic I, Vasilev S, Sofronic-Milosavljevic L. Characterization of rat bone marrow dendritic cells initially primed by Trichinella spiralis antigens. Parasite Immunol. 2008;30(9):491–5.

  32. McKee AS, Pearce EJ. CD25 + CD4+ cells contribute to Th2 polarization during helminth infection by suppressing Th1 response development. J Immunol. 2004;173(2):1224–31.

    Article  CAS  PubMed  Google Scholar 

  33. Schmid-Hempel P. Parasite immune evasion: a momentous molecular war. Trends Ecol Evol. 2008;23(6):318–26.

  34. Layland LE, Ajendra J, Ritter M, Wiszniewsky A, Hoerauf A, Hubner MP. Development of patent Litomosoides sigmodontis infections in semi-susceptible C57BL/6 mice in the absence of adaptive immune responses. Parasit Vectors. 2015;8:396.

  35. Jangpatarapongsa K, Chootong P, Sattabongkot J, Chotivanich K, Sirichaisinthop J, Tungpradabkul S, et al. Plasmodium vivax parasites alter the balance of myeloid and plasmacytoid dendritic cells and the induction of regulatory T cells. Eur J Immunol. 2008;38(10):2697–705.

  36. Maldonado RA, von Andrian UH. How tolerogenic dendritic cells induce regulatory T cells. Adv Immunol. 2010;108:111–65.

  37. Wilson MS, Taylor MD, Balic A, Finney CA, Lamb JR, Maizels RM. Suppression of allergic airway inflammation by helminth-induced regulatory T cells. J Exp Med. 2005;202(9):1199–212.

  38. Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of CD4 + CD25+ regulatory T cells. Nat Immunol. 2003;4(4):330–6.

  39. Lutz MB, Schuler G. Immature, semi-mature and fully mature dendritic cells: which signals induce tolerance or immunity? Trends Immunol. 2002;23(9):445–9.

    Article  CAS  PubMed  Google Scholar 

  40. Lutz MB, Kurts C. Induction of peripheral CD4+ T-cell tolerance and CD8+ T-cell cross-tolerance by dendritic cells. Eur J Immunol. 2009;39(9):2325–30. 

  41. Sofronic-Milosavljevic LJ, Radovic I, Ilic N, Majstorovic I, Cvetkovic J, Gruden-Movsesijan A. Application of dendritic cells stimulated with Trichinella spiralis  excretory-secretory antigens alleviates experimental autoimmune encephalomyelitis. Med Microbiol Immunol. 2013;202(3):239–49.

  42. Gruden-Movsesijan A, Ilic N, Mostarica-Stojkovic M, Stosic-Grujicic S, Milic M, Sofronic-Milosavljevic L. Mechanisms of modulation of experimental autoimmune encephalomyelitis by chronic Trichinella spiralis infection in Dark Agouti rats. Parasite Immunol. 2010;32(6):450–9.

  43. Razmara M, Hilliard B, Ziarani AK, Chen YH, Tykocinski ML. CTLA-4 x Ig converts naive CD4+ CD25- T cells into CD4+ CD25+ regulatory T cells. Int Immunol. 2008;20(4):471–83.

  44. Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, Oukka M, et al. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature. 2006;441(7090):235–8.

  45. Fontenot JD, Rasmussen JP, Williams LM, Dooley JL, Farr AG, Rudensky AY. Regulatory T cell lineage specification by the forkhead transcription factor foxp3. Immunity. 2005;22(3):329–41.

  46. Ono M, Shimizu J, Miyachi Y, Sakaguchi S. Control of autoimmune myocarditis and multiorgan inflammation by glucocorticoid-induced TNF receptor family-related protein(high), Foxp3-expressing CD25+ and CD25- regulatory T cells. J Immunol. 2006;176(8):4748–56.

    Article  CAS  PubMed  Google Scholar 

  47. Beiting DP, Gagliardo LF, Hesse M, Bliss SK, Meskill D, Appleton JA. Coordinated control of immunity to muscle stage Trichinella spiralis by IL-10, regulatory T cells, and TGF-beta. J Immunol. 2007;178(2):1039–47.

    Article  CAS  PubMed  Google Scholar 

  48. Wen X, He L, Chi Y, Zhou S, Hoellwarth J, Zhang C, et al. Dynamics of Th17 cells and their role in Schistosoma japonicum infection in C57BL/6 mice. PLoS Negl Trop Dis. 2011;5(11):e1399.

  49. Osorio F, LeibundGut-Landmann S, Lochner M, Lahl K, Sparwasser T, Eberl G, et al. DC activated via dectin-1 convert Treg into IL-17 producers. Eur J Immunol. 2008;38(12):3274–81.

  50. Peck A, Mellins ED. Precarious balance: Th17 cells in host defense. Infect Immun. 2010;78(1):32–8.

  51. Zhou L, Chong MM, Littman DR. Plasticity of CD4+ T cell lineage differentiation. Immunity. 2009;30(5):646–55.

Download references

Acknowledgments

We thank Bin Zhan and Yuli Cheng, Jing Yang, Xiaohuan Wang, Ran Sun, Limei Zhao and Rui Zhang for their technical assistance and helpful suggestions.

Funding

This study was supported by grants from the National Natural Science Foundation of China (81371837, 81401681), Natural Science Foundation of Beijing (7144192). Grant No.81371837 was used to support experiment reagents and publication. Grants No. 81401681 and No. 7144192 were used to purchase experiment animals, reagents and other relevant materials.

Availability of data and materials

The data supporting the conclusions of this article are included within the article.

Authors’ contributions

KG and XMS performed the experiments. YG, ZXW and JJH performed some of the experiments. The manuscript was written by KG and XMS. XPZ revised the manuscript. XMS and XPZ designed the study. All authors read and approved the final manuscript.

Competing interests

The authors declare that they have no competing interests.

Consent for publication

Not applicable.

Ethics approval and consent to participate

All animal experimentation was carried out in compliance with Capital Medical University IACUC. Capital Medical University ethics committee approved the study and the committee’s reference numbers were AEEI-2015-183 and AEEI-2015-184.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Xinping Zhu.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Guo, K., Sun, X., Gu, Y. et al. Trichinella spiralis paramyosin activates mouse bone marrow-derived dendritic cells and induces regulatory T cells. Parasites Vectors 9, 569 (2016). https://doi.org/10.1186/s13071-016-1857-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13071-016-1857-y

Keywords