Arginase activity induction in macrophages is an escape mechanism developed by parasites to cope with the host’s immune defense and benefit from increased host-derived growth factor production. We report that arginase expression and activity were induced in macrophages during mouse infection by Trypanosoma musculi, a natural parasite of this host. This induction was reproduced in vitro by excreted/secreted factors of the parasite. A mAb directed to TbKHC1, an orphan kinesin H chain from Trypanosoma brucei, inhibited T. musculi excreted/secreted factor–mediated arginase induction. Anti-TbKHC1 Ab also inhibited T. musculi growth, both in vitro and in vivo. Induction of arginase activity and parasite growth involved C-type lectin receptors, because mannose injection decreased arginase activity induction and parasite load in vitro and in vivo. Accordingly, the parasite load was reduced in mice lacking mannose receptor C-type 1. The T. musculi KHC1 homolog showed high similarity with TbKHC1. Bioinformatics analysis revealed the presence of homologs of this gene in other trypanosomes, including pathogens for humans and animals. Host metabolism dysregulation represents an effective parasite mechanism to hamper the host immune response and modify host molecule production to favor parasite invasion and growth. Thus, this orphan kinesin plays an important role in promoting trypanosome infection, and its neutralization or the lock of its partner host molecules offers promising approaches to increasing resistance to infection and new developments in vaccination against trypanosomiasis.

Parasites have developed various strategies to escape the immune response and take advantage of host growth factor production. Therefore, parasites need to reduce the production of toxic molecules, including NO and its derivatives, that are synthesized by the immune system, in particular macrophages (13).

Macrophages are activated and programmed into different functional subtypes in response to signals delivered by the microenvironment. Distinct macrophage-activation states have been widely acknowledged, with classically activated and alternatively activated states as extremes (4). Classically activated macrophages produce effector molecules (reactive oxygen species, NO) and inflammatory molecules (TNF-α, IL-1β, IL-6). The various forms of alternatively activated macrophages generally exhibit high levels of mannose receptor (MR) and arginase 1 upregulation (4). In trypanosomiasis, macrophages are characterized by the induction of arginase (57). Arginase hydrolyzes l-arginine to l-ornithine and urea, decreasing the intracellular pool of l-arginine and, thus, also reducing trypanotoxic NO production by NO synthase 2. Moreover, l-ornithine is a precursor for the synthesis of polyamines and trypanothione, which are essential for trypanosomatid survival (8). Thus, increased arginase activity is likely to counteract the host immune response and favor parasite growth.

In human African trypanosomiasis caused by Trypanosoma gambiense, arginase serum levels increase and return to control values after treatment (9). Accordingly, in experimental murine infection by Trypanosoma brucei, host macrophage arginase activity represents a marker of susceptibility to the disease (10). In T. brucei, arginase activity is induced by excreted/secreted factors (ESFs); in particular, TbKHC1, a kinesin H chain, has been identified as the arginase-inducing factor (11). Arginase induction in macrophages was shown to promote infection by providing polyamine nutrients for trypanosomes, which cannot generate their own source of l-ornithine through the activity of a functional arginase enzyme (11, 12).

We investigated the role of arginase activity induction during a natural murine trypanosomiasis caused by Trypanosoma musculi. In this infection, blood parasitemia lasts 20–25 d, comprising a growth phase, a plateau, and an elimination phase. Although the control of the dynamics of T. musculi infections is, in part, nonimmunological, elimination of this parasite requires Abs and macrophages (1316). Yet, the immune mechanisms controlling early T. musculi proliferation are poorly characterized. Kidney forms persist during all host life. Blood parasites are not eliminated in T cell–deprived mice, blood parasite evolution is modified by nutrition and, curiously, previously infected pregnant mothers quickly became parasitemic (13, 17, 18). T. musculi modulates the host immune response in coinfection with various infectious agents (19). T. musculi can be considered an appropriate model of a host–parasite relationship. Moreover, T. musculi coexpress several receptors binding the Fc regions of IgM, IgG, and IgE, with the receptor common to IgM and IgE being of particular interest (20). The unsuspected immunomodulatory role of an orphan kinesin, TbKHC1, in mice infected with T. brucei, a bovine parasite, led us to investigate the presence and the role of a homolog of this kinesin in this natural murine trypanosome and to examine the presence of homologs in other trypanosomes.

Female Swiss mice, aged 6–8 wk, were purchased from Charles River Laboratories (Saint Germain Nuelles, France). SIGN-R1–knockout (KO) and congenic control (BALB/c background), C57BL/6 (B6), and MR C-type 1 (Mrc1)-KO B6 mice were bred in the Laboratory of Molecular Parasitology, Université Libre de Bruxelles.

Experiments, maintenance, and care of mice complied with the guidelines of the European Convention for the Protection of Vertebrate Animals used for Experimental and other Scientific Purposes (CETS no. 123). Experiments were approved by the Department for the Protection of Animals and Plants of the Préfecture de la Gironde (Identification no. A33-063-324) and the Ethical Committee for Animal Experiments at the Université Libre de Bruxelles (laboratory accreditation no. LA2500482).

d-Mannose, d-galactose, S-(2-boronoethyl)-l-cysteine (BEC), l-arginine, l-ornithine, protease inhibitor mixture, α-isonitrosopropiophenone, DEAE cellulose, Triton X-100, urea, Tris, propidium iodide, and MnCl2 were obtained from Sigma-Aldrich Chemie (Saint-Quentin Fallavier, France). TRIzol reagent was obtained from Invitrogen (Illkirch, France), FITC anti-mouse CD206 (Mrc1) was from BioLegend (San Diego, CA), allophycocyanin-rat anti-mouse CD14 was from BD Biosciences (Le Pont de Claix, France), and PE–anti-mouse CD209 (SIGN-R1) mAb and control Ab were from Miltenyi Biotec (Bergisch Gladbach, Germany). For cell culture and RNA analysis, all reagents were obtained from Life Technologies (Villebon-sur-Yvette, France).

T. musculi (Partinico II strain) was originally obtained from The London School of Hygiene and Tropical Medicine (21). Parasites were stored in liquid nitrogen or maintained in vivo by a peritoneal injection (5 × 104 parasites per mouse) into naive Swiss mice. When required, animals were treated i.p. with 0.2 ml of physiological saline, mannose (50 mM), or galactose (50 mM) three times a day, beginning day 1 postinfection. Mice were also given anti-TbKHC1 Ab (10), an IgG2b isotype control (250 μg in 0.5 ml of saline), or physiological saline (0.5 ml). Parasitemia was monitored by tail blood puncture from days 5 to 24 of infection.

Parasites from 10-d–infected mice were purified from blood by chromatography on DEAE cellulose (22). Soluble parasite ESFs were prepared in a protein-free culture medium, according to a previously described method (23). Purified parasites (2 × 108 per milliliter) were incubated in this medium for 2 h at 37°C. Parasite viability was initially assessed by microscopic examination and then by flow cytometry after propidium iodide staining. Supernatant containing ESFs was separated from trypanosomes by centrifugation (1000 × g, 10 min, 4°C) and filtered using a 0.22-μm low-binding protein filter (Millipore, Molsheim, France). Protein concentration was estimated by the Bradford method, using a kit from Bio-Rad (Marnes-la-Coquette, France).

SDS–PAGE was carried out under nonreducing conditions in 10% polyacrylamide gels (24) and subsequently stained with silver. Protein samples were transferred to a nitrocellulose membrane (Amersham, Buckinghamshire, U.K.) and subsequently blocked, as previously described (25, 26). The membrane was incubated with mouse mAbs at a 1/100 dilution at 37°C for 2 h. After three washings, the membranes were immersed in peroxidase-conjugated anti-murine Ig (Dako, Les Ulis, France), diluted at 1:10,000, at room temperature for 2 h. After washing, membranes were exposed to Clarity ECL reagent (cat. no. 170–5061) at room temperature for 2 min and visualized using a ChemiDoc MP (cat. no. 170–8280; both from Bio-Rad). Band intensities were detected and quantified using Image Lab 5.0 software (Bio-Rad).

The T. musculi KHC sequence was amplified from gDNA with primers designed from the alignment of KHC sequences of T. brucei (Tb927.6.4390) and T. theileri (provided by Dr. Ivens and Prof. Matthews). The resulting fragment, corresponding to nt 275–2780 of T. brucei, was fully sequenced after subcloning in Zero BluntTOPO vector (Invitrogen). Primers specific for the T. musculi sequence were then designed to amplify the 5′ and 3′ extremities of the TmKHC transcript from total RNA by 5′ and 3′ RACE (First Choice RLM-RACE Kit; Ambion, Merelbeke, Belgium). The resulting PCR fragments were cloned into the Zero BluntTOPO vector and sequenced. The following primers were used: TthKHC forward, 5′-AATTTTGATGGTGCGTTTG-3′; TthKHC reverse, 5′-ATTGACATATAATCATCCATTGCAAT-3′; TmKHC 5′ reverse out, 5′-AAACGCGTGGTCAACGCACG-3′; TmKHC 5′ reverse in, 5′-GCAACACTCTCGAAGATCTGC-3′; TmKHC 3′ forward out, 5′-GTGAACTTCGGGAAATAATTGCGTCAGC-3′; and TmKHC 3′ forward in, 5′-GTCAGCGCATTTATCTGTTCCGGAATC-3′.

Peritoneal macrophages were cultured in 96-well plates (105 per well) in RPMI 1640 medium supplemented with 2% FBS, 20 mM HEPES, 2 mM sodium pyruvate, 10−4 M 2-ME, 10 μg/ml gentamicin, and 3% glucose in a humidified 5% CO2 atmosphere at 37°C, as previously described (4). Viability, as determined by trypan blue dye exclusion, was >98%, and May–Grünwald–Giemsa and nonspecific esterase staining revealed that >96% of the cells were macrophages. ESFs (10 μg/200 μl), BEC (100 μM), mannose and galactose (up to 50 mM), l-ornithine (2.5 mM), and Abs (3 μg/200 μl) were added to the culture medium when required.

The macrophage culture medium was removed 48 h later to obtain a macrophage-conditioned medium (MCM) (27). A medium, set under the same conditions but not exposed to macrophages, was used as the control medium. After collection, these media were centrifuged at 1000 × g for 5 min and stored at −80°C until use.

Macrophages from naive mice (105 per well) were cocultured with purified T. musculi (400 per well) in culture medium (28). Parasites were counted after 3 d in coculture.

Arginase activity was measured in macrophages cultured with T. musculi or ESFs for 48 h or in ex vivo macrophages from uninfected mice or infected animals at days 7, 10, 15, 20, and 24 postinfection. Arginase activity was measured by a colorimetric method, as previously described (29). Briefly, 10 mM MnCl2 and 0.5 M l-arginine were added in turn to macrophage lysates and incubated at 37°C for 1 h. The reaction was stopped by adding an acid solution, and the urea formed by arginase was revealed by adding α-isonitrosopropiophenone at 100°C for 45 min. The colored product was quantified by absorption at 540 nm.

Mouse peritoneal macrophages were plated in 60-mm dishes at a density of 2 × 106 cells. Total RNA was extracted using TRIzol reagent. Total RNA (2 μg) was retrotranscribed using the Transcriptor First Strand cDNA Synthesis Kit, according to the manufacturer’s instructions. Amplification of specific PCR products was detected using FastStart Universal SYBR Green Master (ROX) (Roche, Basel, Switzerland) and the following primers: Arg1, 5′-AAGAAAAGGCCGATTCACCT-3′ and 5′-CACCTCCTCTGCTGTCTTCC-3′, 201 bp; Arg2, 5′-ACAGGGTTGCTGTCAGCTCT-3′ and 5′-TGATCCAGACAGCCATTTCA-3′, 298 bp. PCR was performed using an ABI PRISM 7900HT (Applied Biosystems, New York, NY). Fold differences in gene expression were calculated as 2−ΔΔ cycle threshold using β-actin as the housekeeping gene (10).

Peritoneal cells from T. musculi–infected mice were removed at 13 d postinfection. Peritoneal cells from noninfected mice were used as control. Cells (106 per milliliter) were incubated with allophycocyanin–anti-mouse CD14 and FITC–anti-mouse CD206 (MR), PE-anti-mouse CD209 (SIGN-R1), or the corresponding control Abs. After 20 min, cells were washed and resuspended in PBS containing 0.5% FCS. Cells were analyzed by flow cytometry (BD FACSCanto and FACSDiva software version 6.1.2).

The protein sequences of T. musculi, T. brucei, and Trypanosoma theileri KHC were compared with homologs in GenBank using the algorithm “protein BLAST” (Basic Local Alignment Search Tool) from the National Center for Biotechnology Information (https://blast.ncbi.nlm.nih.gov/Blast.cgi). To reconstruct a phylogenetic tree, we selected the sequences from the nonredundant protein sequences database according to three criteria: identity >50%, query cover >50%, and the sequence species were previously characterized as T. brucei, Trypanosoma vivax, Trypanosoma equiperdum, Trypanosoma congolense, Trypanosoma evansi, Trypanosoma cruzi, T. theileri, and Trypanosoma grayi and their subspecies. MEGA6 (30) was used for sequence alignment and tree reconstruction. The best amino acid substitution model was estimated by the Bayesian Information Criterion and used to build a maximum likelihood tree itself tested by the bootstrap method with 100 replications.

Quantitative data are expressed as mean ± SEM. The Student t test was used for comparison between two groups. Prism 5.0 (Graph Pad, La Jolla, CA) was used for analyses.

Arginase activity was measured in macrophages from infected mice at different days postinfection. This activity peaked between days 13 and 17 and paralleled the number of circulating parasites (Fig. 1A) (regression analysis between arginase activity and parasitemia in Supplemental Fig. 1).

FIGURE 1.

Parasitemia, arginase activity, and arginase expression in T. musculi–infected mice. (A) Parasitemia and arginase activity in macrophages at days 5, 7, 10, 13, 15, 20, and 24 postinfection. Day 0 represents arginase activity of macrophages from uninfected mice (control macrophages). Data are the mean ± SEM of eight mice. (B) mRNA expression levels of arginase 1 and arginase 2 in macrophages were assessed by real-time PCR. Data are standardized to β-actin and presented as the fold increase versus naive mice. Data are the mean ± SEM from four mice.

FIGURE 1.

Parasitemia, arginase activity, and arginase expression in T. musculi–infected mice. (A) Parasitemia and arginase activity in macrophages at days 5, 7, 10, 13, 15, 20, and 24 postinfection. Day 0 represents arginase activity of macrophages from uninfected mice (control macrophages). Data are the mean ± SEM of eight mice. (B) mRNA expression levels of arginase 1 and arginase 2 in macrophages were assessed by real-time PCR. Data are standardized to β-actin and presented as the fold increase versus naive mice. Data are the mean ± SEM from four mice.

Close modal

Levels of Arg1 and Arg2 mRNA expression in peritoneal macrophages were determined by RT-PCR. The level of Arg1 was ∼12-fold higher in macrophages from mice 13 d postinfection with T. musculi compared with macrophages from uninfected mice, whereas Arg2 expression was not induced (Fig. 1B).

In macrophages cocultured with T. musculi, arginase activity increased compared with controls (Fig. 2). Arginase activity was still induced, even after separation of parasites from macrophages by a 0.22-μm filter support. Finally, arginase activity induction was also observed in macrophages cultured with ESFs (Fig. 2).

FIGURE 2.

Arginase activity in macrophages cocultured in vitro with T. musculi. Macrophages were cocultured for 3 d with T. musculi (Tm), Tm in an insert, or ESFs or were grown in culture medium (CM) before measuring arginase activity. Data are the mean ± SEM from five separate experiments. ***p < 0.001.

FIGURE 2.

Arginase activity in macrophages cocultured in vitro with T. musculi. Macrophages were cocultured for 3 d with T. musculi (Tm), Tm in an insert, or ESFs or were grown in culture medium (CM) before measuring arginase activity. Data are the mean ± SEM from five separate experiments. ***p < 0.001.

Close modal

T. musculi was grown in the presence of macrophages or in a 2-d parasite-free MCM (27, 28). Adding BEC, an arginase inhibitor, to the parasite–macrophage culture dramatically reduced parasite growth, which was restored by adding l-ornithine (Fig. 3). MCM promoted parasite growth, whereas control medium had no effect. Addition of BEC to MCM just prior to incubation with parasites had no effect on parasite growth, indicating that BEC had no direct effect on parasites (data not shown).

FIGURE 3.

Induction of arginase activity favors in vitro parasite growth. Parasites were counted after a 3-d coculture with macrophages. Parasite growth was assessed as the percentage compared with growth in the presence of controls (cocultures of parasite and macrophages from naive mice). The effects of the arginase inhibitor BEC, with or without l-ornithine, on parasite growth were assessed. Data are the mean ± SEM from four separate experiments. ***p < 0.001.

FIGURE 3.

Induction of arginase activity favors in vitro parasite growth. Parasites were counted after a 3-d coculture with macrophages. Parasite growth was assessed as the percentage compared with growth in the presence of controls (cocultures of parasite and macrophages from naive mice). The effects of the arginase inhibitor BEC, with or without l-ornithine, on parasite growth were assessed. Data are the mean ± SEM from four separate experiments. ***p < 0.001.

Close modal

Using Western blot, a mAb directed to TbKHC1, a previously described arginase-inducing factor from T. brucei (11), recognized a band in T. musculi ESFs corresponding to the molecular mass of TbKHC1 (122 kDa) (Fig. 4).

FIGURE 4.

T. musculi express a TbKHC1 cross-reactive protein. After SDS-PAGE and blotting, T. musculi ESFs were incubated with anti-TbKHC1 (1/200) or IgG2b isotype control (1/200) Ab. Peroxidase-labeled goat anti-mouse IgG was added as secondary Ab (1/5000).

FIGURE 4.

T. musculi express a TbKHC1 cross-reactive protein. After SDS-PAGE and blotting, T. musculi ESFs were incubated with anti-TbKHC1 (1/200) or IgG2b isotype control (1/200) Ab. Peroxidase-labeled goat anti-mouse IgG was added as secondary Ab (1/5000).

Close modal

To identify a putative T. musculi KHC gene, we first aligned the TbKHC1 sequence with the T. theileri homolog, which is more closely related to T. musculi. Two primers were designed in conserved regions to amplify a 2505-nt fragment from T. musculi gDNA. Sequencing of this fragment showed 70 and 63% identity with TbKHC1 at the DNA and protein levels, respectively. This sequence was used to design two sets of primers for amplification of the 5′ and 3′ ends of the T. musculi gene by retrotranscription of total RNA, followed by nested PCR. Assembly of these 5′ and 3′ sequences with the first PCR product revealed that the TmKHC gene encoded a protein of 1145 aa that had 59% identity with TbKHC1 at the protein level (Fig. 5). The protein N-terminal domain contained the highly conserved kinesin motor domain with a typical ATP binding site.

FIGURE 5.

Protein sequence alignment of KHC genes in T. brucei brucei, T. musculi, and T. theileri. Kinesin motor domains with ATP binding sites (▪) and the start of the shorter cDNA recognized by anti-TbKHC1 in T. brucei brucei (▼) are indicated.

FIGURE 5.

Protein sequence alignment of KHC genes in T. brucei brucei, T. musculi, and T. theileri. Kinesin motor domains with ATP binding sites (▪) and the start of the shorter cDNA recognized by anti-TbKHC1 in T. brucei brucei (▼) are indicated.

Close modal

When anti-TbKHC1 mAb was added to T. musculi–macrophage cocultures, arginase induction and parasite growth were markedly reduced compared with the isotype-control Ab (Fig. 6A).

FIGURE 6.

In vitro and in vivo parasite growth in the presence of anti-TbKHC1. (A) Parasites were counted after a 3-d coculture with macrophages in the presence of anti-TbKHC1 or IgG2b isotype-control Ab (3 μg/200 μl). Data are the mean ± SEM from four separate experiments. (B) Mice were injected i.p. with physiological saline (500 μl), anti-TbKHC1, or IgG2b isotype-control Ab (250 μg in 500 μl saline) at day 2 post–T. musculi infection. Parasitemia at days 5, 7, 10, 13, 15, 20, and 24 postinfection is shown. Data are the mean ± SEM from five mice. **p < 0.01, ***p < 0.001.

FIGURE 6.

In vitro and in vivo parasite growth in the presence of anti-TbKHC1. (A) Parasites were counted after a 3-d coculture with macrophages in the presence of anti-TbKHC1 or IgG2b isotype-control Ab (3 μg/200 μl). Data are the mean ± SEM from four separate experiments. (B) Mice were injected i.p. with physiological saline (500 μl), anti-TbKHC1, or IgG2b isotype-control Ab (250 μg in 500 μl saline) at day 2 post–T. musculi infection. Parasitemia at days 5, 7, 10, 13, 15, 20, and 24 postinfection is shown. Data are the mean ± SEM from five mice. **p < 0.01, ***p < 0.001.

Close modal

In addition, when anti-TbKHC1 Ab was injected i.p. into mice on day 2 post–T. musculi infection, the parasite load strongly decreased, whereas isotype-control Ab treatment had no effect (Fig. 6B). Arginase activity also decreased in macrophages from 13-d infected mice in the anti-TbKHC1 Ab–treated group (38 ± 11 mU per 105 macrophages) compared with those from the isotype-control group (117 ± 20 mU per 105 macrophages) or the saline group (125 ± 18 mU per 105 macrophages).

In cells obtained from the peritoneal cavity of an unmanipulated mouse, ∼50–60% are B cells, ∼30% are macrophages, and 5–10% are T cells (31). In murine trypanosomiasis, C-type lectin receptor expression increases in macrophages (6). The number of peritoneal macrophages was increased in T. musculi–infected mice, and granulocytes did not exceed 1% (14). In the peritoneal cavity from 14-d infected mice, CD14high cells have been considered macrophages, and expression levels of CD206 (Mrc1) on these cells (Fig. 7) increased 4-fold compared with control macrophages (mean fluorescence intensity, 1987 ± 89 versus 476 ± 51), whereas CD209b (SIGN-R1) expression was unchanged (mean fluorescence intensity, 685 ± 102 versus 648 ± 91).

FIGURE 7.

MR was expressed on macrophages from T. musculi–infected mice. Peritoneal cells (106 per milliliter) from 14-d infected mice and from noninfected mice were labeled with allophycocyanin-rat anti-mouse CD14 and FITC–anti-mouse CD206 Ab. The percentage of CD14+ CD206+ cells was determined. Data are the mean ± SEM from six mice. ***p < 0.001.

FIGURE 7.

MR was expressed on macrophages from T. musculi–infected mice. Peritoneal cells (106 per milliliter) from 14-d infected mice and from noninfected mice were labeled with allophycocyanin-rat anti-mouse CD14 and FITC–anti-mouse CD206 Ab. The percentage of CD14+ CD206+ cells was determined. Data are the mean ± SEM from six mice. ***p < 0.001.

Close modal

Mannose or galactose was added to parasite–macrophage cocultures. Mannose inhibited parasite growth in a dose-dependent manner, whereas galactose had only a marginal effect (Fig. 8). Supplementing MCM with mannose or galactose just before adding it to purified parasite cultures had no effect on parasite growth, indicating the absence of a direct toxic effect of sugars on the parasites (data not shown).

FIGURE 8.

(A) Effect of sugars on in vitro T. musculi proliferation. Parasite growth was assessed, after 3 d in parasite–macrophage cocultures containing mannose or galactose, as a percentage compared with growth in the presence of controls (macrophages from naive mice without sugars). Data are the mean ± SEM from six culture wells. (B) Inhibition of parasites and ESF-induced arginase activity in vitro by sugars. Macrophages were cultured in medium containing parasites or ESFs and supplemented with mannose or galactose (10 mM). Arginase activity was assessed 72 h later. Data are the mean ± SEM from six culture wells. **p < 0.01, ***p < 0.001. ns, nonsignificant.

FIGURE 8.

(A) Effect of sugars on in vitro T. musculi proliferation. Parasite growth was assessed, after 3 d in parasite–macrophage cocultures containing mannose or galactose, as a percentage compared with growth in the presence of controls (macrophages from naive mice without sugars). Data are the mean ± SEM from six culture wells. (B) Inhibition of parasites and ESF-induced arginase activity in vitro by sugars. Macrophages were cultured in medium containing parasites or ESFs and supplemented with mannose or galactose (10 mM). Arginase activity was assessed 72 h later. Data are the mean ± SEM from six culture wells. **p < 0.01, ***p < 0.001. ns, nonsignificant.

Close modal

In addition, mannose had a more marked inhibitory effect than galactose on parasite- and ESF-induced arginase activity (Fig. 8B).

Intraperitoneal injection of mannose from day 1 post–T. musculi infection dramatically decreased the parasite load in mice, whereas galactose had a less pronounced effect (Fig. 9). At 14 d postinfection, macrophage arginase activity had decreased in mice receiving mannose (34 ± 15 mU per 105 cells) compared with infected mice receiving saline (122 ± 17 mU per 105 cells) or galactose (88 ± 21 mU per 105 cells).

FIGURE 9.

Effect of sugars on T. musculi parasitemia. Parasitemia was assessed in mice receiving i.p. injections of physiological saline, mannose, or galactose three times a day from day 1 postinfection. Data are the mean ± SEM from six mice.

FIGURE 9.

Effect of sugars on T. musculi parasitemia. Parasitemia was assessed in mice receiving i.p. injections of physiological saline, mannose, or galactose three times a day from day 1 postinfection. Data are the mean ± SEM from six mice.

Close modal

T. musculi parasitemia was reduced 10-fold in Mrc1-KO mice compared with wild-type mice, whereas parasitemia decreased marginally in SIGN-R1 KO mice compared with controls (Fig. 10).

FIGURE 10.

MR-deficient mice exhibit improved parasite control. Parasitemia was monitored in various T. musculi–infected mice: Mrc1-KO B6 mice, wild-type (WT) B6 mice, SIGN-R1–KO BALB/c mice, and control (Ctrl SIGN-R1) mice. Data are the mean ± SEM of four individual mice in a representative experiment, repeated three times.

FIGURE 10.

MR-deficient mice exhibit improved parasite control. Parasitemia was monitored in various T. musculi–infected mice: Mrc1-KO B6 mice, wild-type (WT) B6 mice, SIGN-R1–KO BALB/c mice, and control (Ctrl SIGN-R1) mice. Data are the mean ± SEM of four individual mice in a representative experiment, repeated three times.

Close modal

Only eight GenBank sequences fulfilled the selection criteria (i.e., >50% homology and >50% cover from sequences belonging to the selected species): one T. brucei gambiense, one T. equiperdum, one T. congolense, one T. vivax, one T. grayi, two T. cruzi, and one Trypanosoma cruzi marinkellei. There was no protein sequence of KHC for T. brucei rhodesiense, T. evansi, or T. theileri. The protein sequences of T. brucei, T. musculi, and T. theileri KHCs, of 1111, 1148, and 1123 aa, respectively, were aligned with these eight GenBank sequences; after elimination of the noncommon parts at both ends, we obtained a final alignment of 1071 aa sites, including gaps. The maximum likelihood tree is shown in Fig. 11. TbKHC1 (strain TREU927 of T. brucei brucei, GenBank reference XP_845556) presented 100% identity with T. equiperdum (SCU65329), 99% identity with T. brucei gambiense (XP 011774228), and 78% identity with T. congolense (CCC91128). KHC of T. musculi presented 75–76% identity with the strains belonging to the T. cruzi clade and <66% identity for the other species, whereas KHC of T. theileri presented its maximum identity (69%) with T. grayi (XP 009307339) and <65% with the other species.

FIGURE 11.

KHC phylogenetic tree. Maximum likelihood tree based on the Le and Gascuel 2008 model. The tree with the highest log likelihood (−9652,6026) is shown. A discrete gamma distribution was used to model evolutionary rate differences among sites (five categories [+G, parameter = 0.6297]). The tree is drawn to scale, with branch lengths measured in the number of substitutions per site. The analysis involved 11 amino acid sequences. Evolutionary analyses were conducted in MEGA6. The numbers at the nodes are the bootstrap values obtained using 100 replications. The leaf texts are the National Center for Biotechnology Information reference followed to the species name and to the strain name when available.

FIGURE 11.

KHC phylogenetic tree. Maximum likelihood tree based on the Le and Gascuel 2008 model. The tree with the highest log likelihood (−9652,6026) is shown. A discrete gamma distribution was used to model evolutionary rate differences among sites (five categories [+G, parameter = 0.6297]). The tree is drawn to scale, with branch lengths measured in the number of substitutions per site. The analysis involved 11 amino acid sequences. Evolutionary analyses were conducted in MEGA6. The numbers at the nodes are the bootstrap values obtained using 100 replications. The leaf texts are the National Center for Biotechnology Information reference followed to the species name and to the strain name when available.

Close modal

This study revealed that T. musculi ESFs induced macrophage arginase 1 activity via a pathway involving mannose-binding receptor(s), promoting parasite growth. A mAb specific to the kinesin H chain TbKHC1, an orphan kinesin present in ESFs from T. brucei, inhibited arginase activity and reduced T. musculi load in vitro and in vivo, suggesting that the TbKHC1 homolog identified in T. musculi, with 59% aa identity with TbKHC1, was the active component of T. musculi ESFs. However, although TbKHC1 facilitates T. brucei parasitemia via the SIGN-R1 receptor (11), MR was apparently the main target of T. musculi ESFs. This suggested that kinesin H chain–related proteins played similar roles in promoting infection in two distant trypanosomes, via macrophage arginase induction, following distinct C-type lectin receptor targeting.

Macrophages have been widely used as feeder layer cells to favor parasite cell proliferation in vitro (27, 28). Adding an arginase inhibitor reduced their parasite growth-promoting activity, which was restored by l-ornithine supplementation (Fig. 3), although synergy with the various components of FCS cannot be ruled out. The essential requirement for l-ornithine is linked to the absence of the functional enzyme arginase in trypanosomes (12), resulting in their dependence on host arginase activity for polyamine and trypanothione synthesis, which are essential for parasite survival, growth, and differentiation (7, 32). Difluoromethylornithine, a structural analog of l-ornithine, is used alone or, more recently, in combination with nifurtimox as an effective drug in human African trypanosomiasis (33). However, its administration is difficult and requires large quantities of i.v. injected fluids, which limits its use in remote areas. Therefore, it would be useful to find easier ways to target polyamine synthesis, including delivery of cytotoxic polyamine analogs, to improve resistance to trypanosome infection. Alternatively, inhibitors of the pathway triggering arginase activity are expected to reduce parasite load in infected animals, suggesting that strategies targeting trypanosome ESFs would be useful for increasing resistance to trypanosomiasis.

Parasites have to adapt and develop in successive hosts by modulating various host environments and defense mechanisms. Communication between host cells and parasites may take the form of cell–cell contact, soluble factors, and/or extracellular vesicles (exosomes or ectosomes) from parasites, involving membrane receptors of host cells. C-type lectin receptors, expressed in large quantities by dendritic cells and macrophages, play important roles in numerous basic phenomena, as well as various aspects of the immune response. These receptors induce highly diverse immune responses to pathogens (34). They are involved in protection against various fungi, such as Cryptococcus neoformans (35). Several pathogens exploit these receptors to suppress an efficient immune response (36). Distinct C-type lectin receptors can contribute positively and negatively to the macrophage response to Leishmania infantum, a parasite related to trypanosomes (37). MR binds to many microorganisms, primarily intracellular, that are coated with carbohydrates, including viruses, bacteria, fungi, and parasites (38). We showed in this article that T. musculi KHC1, a secreted kinesin, induced arginase via Mrc1, which is upregulated in macrophages from T. musculi–infected mice. This arginase induction was inhibited by a mAb directed to TbKHC1 (Fig. 6), as well as by mannose, an MR inhibitor (Fig. 8) (11, 39). In mice infected with T. cruzi, a parasite related to T. musculi, MR expression was upregulated in macrophages. The T. cruzi Ag cruzipain enhanced MR recycling, which favored arginase induction and T. cruzi survival in macrophages, whereas receptor blockade decreased arginase activity and parasite growth (40). All of these data highlighted the importance of arginase induction for extra- and intracellular trypanosomes and identified the parasite molecules and host receptors involved.

ESFs from different parasite strains induce macrophage arginase as a function of strain virulence (23); however, their contents are poorly known. A mAb directed to TbKHC1 reduced parasite load in T. musculi–infected mice (Fig. 6). This indicated that a homolog to TbKHC1 was present in T. musculi and possessed similar properties, such as macrophage arginase induction (Figs. 4 and 5). Moreover, sequence analysis revealed the presence of homologs of this gene in other trypanosomes (Fig. 11), including pathogens affecting humans (causing African trypanosomiasis [sleeping sickness] or Chagas disease) and animals (reducing production and increasing morbidity and mortality in livestock). It is worth noting that the hierarchical structure of the KHC phylogenetic tree was consistent with previous reports on other markers: monophyly of T. cruzi and T. brucei/T. equiperdum and intermediary positions for the others species making KHC as a potential phylogenetic marker for trypanosomes (41, 42).

The dysregulation of host l-arginine–inducible metabolism by ESFs represents an effective parasite mechanism to hamper host immune response and modify host molecule production to favor parasite invasion and growth. Preventing this host metabolism dysregulation by immunizing against active components in ESFs or locking partner host molecules is a promising approach. A selective targeting of TbKHC1 kinesin H chain homologs in trypanosomiasis deserves further investigation. Antigenic variations in surface glycoproteins hamper vaccination against African trypanosomiasis. Conserved epitopes in nonvariant Ags may be selected and combined and then associated with an adjuvant in an appropriate vaccine formulation to induce adequate immune responses (43, 44).

We thank Patricia Nabos for animal care, Vincent Pitard and Santiago Gonzalez (cytometry platform, University of Bordeaux), and Dr. Alasdair Ivens and Prof. Keith Matthews (University of Edinburgh) for providing T. theileri genome sequence information.

This study was supported by funding from Pôle d’Attraction Interuniversitaire P7/41 and European Research Council Grant 669007-APOLs to E.P. A.B. is funded by Pôle d’Attraction Interuniversitaire P7/41. R.N.-B. and S.S. thank le Service de Coopération et d’Action Culturelle de l’Ambassade de France à Bangui and l’Association pour le Développement de la Recherche en Parasitologie et Santé Tropicale for continuous support.

The online version of this article contains supplemental material.

Abbreviations used in this article:

B6

C57BL/6

BEC

S-(2-boronoethyl)-l-cysteine

ESF

excreted/secreted factor

KO

knockout

MCM

macrophage-conditioned medium

MR

mannose receptor

Mrc1

MR C-type 1.

1
Green
,
S. J.
,
M. S.
Meltzer
,
J. B.
Hibbs
Jr.
,
C. A.
Nacy
.
1990
.
Activated macrophages destroy intracellular Leishmania major amastigotes by an L-arginine-dependent killing mechanism.
J. Immunol.
144
:
278
283
.
2
Vincendeau
,
P.
,
S.
Daulouède
.
1991
.
Macrophage cytostatic effect on Trypanosoma musculi involves an L-arginine-dependent mechanism.
J. Immunol.
146
:
4338
4343
.
3
Bogdan
,
C.
2015
.
Nitric oxide synthase in innate and adaptive immunity: an update.
Trends Immunol.
36
:
161
178
.
4
Mantovani
,
A.
,
S. K.
Biswas
,
M. R.
Galdiero
,
A.
Sica
,
M.
Locati
.
2013
.
Macrophage plasticity and polarization in tissue repair and remodelling.
J. Pathol.
229
:
176
185
.
5
Gobert
,
A. P.
,
S.
Daulouede
,
M.
Lepoivre
,
J. L.
Boucher
,
B.
Bouteille
,
A.
Buguet
,
R.
Cespuglio
,
B.
Veyret
,
P.
Vincendeau
.
2000
.
L-Arginine availability modulates local nitric oxide production and parasite killing in experimental trypanosomiasis.
Infect. Immun.
68
:
4653
4657
.
6
Namangala
,
B.
,
P.
De Baetselier
,
W.
Noël
,
L.
Brys
,
A.
Beschin
.
2001
.
Alternative versus classical macrophage activation during experimental African trypanosomosis.
J. Leukoc. Biol.
69
:
387
396
.
7
Raes
,
G.
,
L.
Brys
,
B. K.
Dahal
,
J.
Brandt
,
J.
Grooten
,
F.
Brombacher
,
G.
Vanham
,
W.
Noël
,
P.
Bogaert
,
T.
Boonefaes
, et al
.
2005
.
Macrophage galactose-type C-type lectins as novel markers for alternatively activated macrophages elicited by parasitic infections and allergic airway inflammation.
J. Leukoc. Biol.
77
:
321
327
.
8
Flohé
,
L.
2012
.
The trypanothione system and its implications in the therapy of trypanosomatid diseases.
Int. J. Med. Microbiol.
302
:
216
220
.
9
Nzoumbou-Boko
,
R.
,
M.
Dethoua
,
F.
Gabriel
,
A.
Buguet
,
R.
Cespuglio
,
P.
Courtois
,
S.
Daulouède
,
B.
Bouteille
,
S.
Ngampo
,
G.
Mpandzou
, et al
.
2013
.
Serum arginase, a biomarker of treatment efficacy in human African trypanosomiasis.
J. Clin. Microbiol.
51
:
2379
2381
.
10
Duleu
,
S.
,
P.
Vincendeau
,
P.
Courtois
,
S.
Semballa
,
I.
Lagroye
,
S.
Daulouède
,
J. L.
Boucher
,
K. T.
Wilson
,
B.
Veyret
,
A. P.
Gobert
.
2004
.
Mouse strain susceptibility to trypanosome infection: an arginase-dependent effect.
J. Immunol.
172
:
6298
6303
.
11
De Muylder
,
G.
,
S.
Daulouède
,
L.
Lecordier
,
P.
Uzureau
,
Y.
Morias
,
J.
Van Den Abbeele
,
G.
Caljon
,
M.
Hérin
,
P.
Holzmuller
,
S.
Semballa
, et al
.
2013
.
A Trypanosoma brucei kinesin heavy chain promotes parasite growth by triggering host arginase activity.
PLoS Pathog.
9
:
e1003731
.
12
Hai
,
Y.
,
E. J.
Kerkhoven
,
M. P.
Barrett
,
D. W.
Christianson
.
2015
.
Crystal structure of an arginase-like protein from Trypanosoma brucei that evolved without a binuclear manganese cluster.
Biochemistry
54
:
458
471
.
13
Viens
,
P.
,
G. A.
Targett
,
E.
Leuchars
,
A. J.
Davies
.
1974
.
The immunological response of CBA mice to Trypanosoma musculi. I. Initial control of the infection and the effect of T-cell deprivation.
Clin. Exp. Immunol.
16
:
279
294
.
14
Vincendeau
,
P.
,
A.
Caristan
,
R.
Pautrizel
.
1981
.
Macrophage function during Trypanosoma musculi infection in mice.
Infect. Immun.
34
:
378
381
.
15
Vincendeau
,
P.
,
M.
Daëron
,
S.
Daulouède
.
1986
.
Identification of antibody classes and Fc receptors responsible for phagocytosis of Trypanosoma musculi by mouse macrophages.
Infect. Immun.
53
:
600
605
.
16
Albright
,
J. W.
,
M.
Pierantoni
,
J. F.
Albright
.
1990
.
Immune and nonimmune regulation of the population of Trypanosoma musculi in infected host mice.
Infect. Immun.
58
:
1757
1762
.
17
Davies
,
A. J.
2012
.
Immigration control in the vertebrate body with special reference to chimerism.
Chimerism
3
:
1
8
.
18
Ongele
,
E. A.
,
M.
Ashraf
,
R. A.
Nesbitt
,
P. A.
Humphrey
,
C. M.
Lee
.
2002
.
Effects of selenium deficiency in the development of trypanosomes and humoral immune responses in mice infected with Trypanosoma musculi.
Parasitol. Res.
88
:
540
545
.
19
Lowry
,
J. E.
,
J. A.
Leonhardt
,
C.
Yao
,
E. L.
Belden
,
G. P.
Andrews
.
2014
.
Infection of C57BL/6 mice by Trypanosoma musculi modulates host immune responses during Brucella abortus cocolonization.
J. Wildl. Dis.
50
:
11
20
.
20
Vincendeau
,
P.
,
M.
Daëron
.
1989
.
Trypanosoma musculi co-express several receptors binding rodent IgM, IgE, and IgG subclasses.
J. Immunol.
142
:
1702
1709
.
21
Krampitz
,
H. E.
1969
.
Verbreitung, Wirt-Parasit-Beziehungen und Vermehrung sizilianischer Stämme von Trypanosoma (Herpetosoma) duttoni Thiroux, 1905 (Protozoa, Trypanosomatidae)
.
Z. Parasitenkd.
32
:
297
315
.
22
Lanham
,
S. M.
1968
.
Separation of trypanosomes from the blood of infected rats and mice by anion-exchangers.
Nature
218
:
1273
1274
.
23
Holzmuller
,
P.
,
D. G.
Biron
,
P.
Courtois
,
M.
Koffi
,
R.
Bras-Gonçalves
,
S.
Daulouède
,
P.
Solano
,
G.
Cuny
,
P.
Vincendeau
,
V.
Jamonneau
.
2008
.
Virulence and pathogenicity patterns of Trypanosoma brucei gambiense field isolates in experimentally infected mouse: differences in host immune response modulation by secretome and proteomics.
Microbes Infect.
10
:
79
86
.
24
Laemmli
,
U. K.
1970
.
Cleavage of structural proteins during the assembly of the head of bacteriophage T4.
Nature
227
:
680
685
.
25
Semballa
,
S.
,
M. C.
Okomo-Assoumou
,
P.
Holzmuller
,
P.
Büscher
,
S.
Magez
,
J. L.
Lemesre
,
S.
Daulouede
,
P.
Courtois
,
M.
Geffard
,
P.
Vincendeau
.
2007
.
Identification of a tryptophan-like epitope borne by the variable surface glycoprotein (VSG) of African trypanosomes.
Exp. Parasitol.
115
:
173
180
.
26
Bras-Gonçalves
,
R.
,
E.
Petitdidier
,
J.
Pagniez
,
R.
Veyrier
,
P.
Cibrelus
,
M.
Cavaleyra
,
S.
Maquaire
,
J.
Moreaux
,
J. L.
Lemesre
.
2014
.
Identification and characterization of new Leishmania promastigote surface antigens, LaPSA-38S and LiPSA-50S, as major immunodominant excreted/secreted components of L. amazonensis and L. infantum.
Infect. Genet. Evol.
24
:
1
14
.
27
Albright
,
J. W.
,
J. F.
Albright
.
1980
.
In vitro growth of Trypanosoma musculi in cell-free medium conditioned by rodent macrophages and mercaptoethanol.
Int. J. Parasitol.
10
:
137
142
.
28
Vincendeau
,
P.
,
B.
Guillemain
,
S.
Daulouède
,
C.
Ripert
.
1986
.
In vitro growth of Trypanosoma musculi: requirements of cells and serum free culture medium.
Int. J. Parasitol.
16
:
387
390
.
29
Corraliza
,
I. M.
,
M. L.
Campo
,
G.
Soler
,
M.
Modolell
.
1994
.
Determination of arginase activity in macrophages: a micromethod.
J. Immunol. Methods
174
:
231
235
.
30
Tamura
,
K.
,
G.
Stecher
,
D.
Peterson
,
A.
Filipski
,
S.
Kumar
.
2013
.
MEGA6: molecular evolutionary genetics analysis version 6.0.
Mol. Biol. Evol.
30
:
2725
2729
.
31
Ray
,
A
,
B. N.
Dittel
.
2010
.
Isolation of mouse peritoneal cavity cells.
J. Vis. Exp.
35
:
e1488
.
32
Fairlamb
,
A. H.
,
A.
Cerami
.
1992
.
Metabolism and functions of trypanothione in the Kinetoplastida.
Annu. Rev. Microbiol.
46
:
695
729
.
33
Priotto
,
G.
,
S.
Kasparian
,
W.
Mutombo
,
D.
Ngouama
,
S.
Ghorashian
,
U.
Arnold
,
S.
Ghabri
,
E.
Baudin
,
V.
Buard
,
S.
Kazadi-Kyanza
, et al
.
2009
.
Nifurtimox-eflornithine combination therapy for second-stage African Trypanosoma brucei gambiense trypanosomiasis: a multicentre, randomised, phase III, non-inferiority trial.
Lancet
374
:
56
64
.
34
van den Berg
,
L. M.
,
S. I.
Gringhuis
,
T. B.
Geijtenbeek
.
2012
.
An evolutionary perspective on C-type lectins in infection and immunity.
Ann. N. Y. Acad. Sci.
1253
:
149
158
.
35
Wüthrich
,
M.
,
G. S.
Deepe
Jr.
,
B.
Klein
.
2012
.
Adaptive immunity to fungi.
Annu. Rev. Immunol.
30
:
115
148
.
36
Engering
,
A.
,
T. B.
Geijtenbeek
,
Y.
van Kooyk
.
2002
.
Immune escape through C-type lectins on dendritic cells.
Trends Immunol.
23
:
480
485
.
37
Lefèvre
,
L.
,
G.
Lugo-Villarino
,
E.
Meunier
,
A.
Valentin
,
D.
Olagnier
,
H.
Authier
,
C.
Duval
,
C.
Dardenne
,
J.
Bernad
,
J. L.
Lemesre
, et al
.
2013
.
The C-type lectin receptors dectin-1, MR, and SIGNR3 contribute both positively and negatively to the macrophage response to Leishmania infantum.
Immunity
38
:
1038
1049
.
38
Martinez-Pomares
,
L.
2012
.
The mannose receptor.
J. Leukoc. Biol.
92
:
1177
1186
.
39
deSchoolmeester
,
M. L.
,
L.
Martinez-Pomares
,
S.
Gordon
,
K. J.
Else
.
2009
.
The mannose receptor binds Trichuris muris excretory/secretory proteins but is not essential for protective immunity.
Immunology
126
:
246
255
.
40
Garrido
,
V. V.
,
L. R.
Dulgerian
,
C. C.
Stempin
,
F. M.
Cerbán
.
2011
.
The increase in mannose receptor recycling favors arginase induction and Trypanosoma cruzi survival in macrophages.
Int. J. Biol. Sci.
7
:
1257
1272
.
41
Stevens
,
J. R.
,
W.
Gibson
.
1999
.
The molecular evolution of trypanosomes.
Parasitol. Today
15
:
432
437
.
42
Da Silva
,
F. M.
,
H.
Noyes
,
M.
Campaner
,
A. C.
Junqueira
,
J. R.
Coura
,
N.
Añez
,
J. J.
Shaw
,
J. R.
Stevens
,
M. M.
Teixeira
.
2004
.
Phylogeny, taxonomy and grouping of Trypanosoma rangeli isolates from man, triatomines and sylvatic mammals from widespread geographical origin based on SSU and ITS ribosomal sequences.
Parasitology
129
:
549
561
.
43
La Greca
,
F.
,
S.
Magez
.
2011
.
Vaccination against trypanosomiasis: can it be done or is the trypanosome truly the ultimate immune destroyer and escape artist?
Hum. Vaccin.
7
:
1225
1233
.
44
Gimenez
,
A. M.
,
K. S.
Françoso
,
J.
Ersching
,
M. Y.
Icimoto
,
V.
Oliveira
,
A. E.
Rodriguez
,
L.
Schnittger
,
M.
Florin-Christensen
,
M. M.
Rodrigues
,
I. S.
Soares
.
2016
.
A recombinant multi-antigen vaccine formulation containing Babesia bovis merozoite surface antigens MSA-2a1, MSA-2b and MSA-2c elicits invasion-inhibitory antibodies and IFN-γ producing cells.
Parasit. Vectors
9
:
577
.

The authors have no financial conflicts of interest.

Supplementary data