Skip to main content
Advertisement
Browse Subject Areas
?

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

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Immunophenotypic Lymphocyte Profiles in Human African Trypanosomiasis

  • Caroline Boda ,

    Contributed equally to this work with: Caroline Boda, Bertrand Courtioux

    Affiliations Université de Limoges, IFR 145 GEIST, Institut de Neurologie Tropicale, EA 3174 NeuroEpidémiologie Tropicale et Comparée, Faculté de Médecine, Limoges, France, Centre International de Recherches Médicales de Franceville (CIRMF), BP 769, Franceville, Gabon

  • Bertrand Courtioux ,

    Contributed equally to this work with: Caroline Boda, Bertrand Courtioux

    bertrand.courtioux@unilim.fr

    Affiliation Université de Limoges, IFR 145 GEIST, Institut de Neurologie Tropicale, EA 3174 NeuroEpidémiologie Tropicale et Comparée, Faculté de Médecine, Limoges, France

  • Pierre Roques,

    Affiliations Centre International de Recherches Médicales de Franceville (CIRMF), BP 769, Franceville, Gabon, CEA, Division of Immuno-Virology/Institute of Emerging Diseases and Innovative Therapies (iMETI), Fontenay-aux-Roses, France

  • Lynda Pervieux,

    Affiliation Université de Limoges, IFR 145 GEIST, EA 3174 NeuroEpidémiologie Tropicale et Comparée, Faculté de Médecine, Limoges, France

  • Gédéon Vatunga,

    Affiliation Instituto de Combate e Controlo das Tripanossomiases-ICCT/Minsa, Luanda, Angola

  • Théophile Josenando,

    Affiliation Instituto de Combate e Controlo das Tripanossomiases-ICCT/Minsa, Luanda, Angola

  • Constant Roger Ayenengoye,

    Affiliation Programme National de Lutte contre la Trypanosomose Humaine Africaine, PNLTHA, Libreville, Gabon

  • Bernard Bouteille,

    Affiliation Université de Limoges, IFR 145 GEIST, EA 3174 NeuroEpidémiologie Tropicale et Comparée, Faculté de Médecine, Limoges, France

  • Marie-Odile Jauberteau,

    Affiliation Université de Limoges, IFR 145 GEIST, EA 3842 Homéostasie cellulaire et pathologies, Faculté de Médecine, Limoges, France

  • Sylvie Bisser

    Affiliations Université de Limoges, IFR 145 GEIST, Institut de Neurologie Tropicale, EA 3174 NeuroEpidémiologie Tropicale et Comparée, Faculté de Médecine, Limoges, France, Centre International de Recherches Médicales de Franceville (CIRMF), BP 769, Franceville, Gabon

Abstract

Human African trypanosomiasis (HAT) is a deadly vector-born disease caused by an extracellular parasite, the trypanosome. Little is known about the cellular immune responses elicited by this parasite in humans. We used multiparameter flow cytometry to characterize leukocyte immunophenotypes in the blood and cerebrospinal fluid (CSF) of 33 HAT patients and 27 healthy controls identified during a screening campaign in Angola and Gabon. We evaluated the subsets and activation markers of B and T lymphocytes. Patients had a higher percentage of CD19+ B lymphocytes and activated B lymphocytes in the blood than did controls, but lacked activated CD4+ T lymphocytes (CD25+). Patients displayed no increase in the percentage of activated CD8+ T cells (HLA-DR+, CD69+ or CD25+), but memory CD8 T-cell levels (CD8+CD45RA) were significantly lower in patients than in controls, as were effector CD8 T-cell levels (CD8+CD45RA+CD62L). No relationship was found between these blood immunophenotypes and disease severity (stage 1 vs 2). However, CD19+ B-cell levels in the CSF increased with disease severity. The patterns of T and B cell activation in HAT patients suggest that immunomodulatory mechanisms may operate during infection. Determinations of CD19+ B-cell levels in the CSF could improve disease staging.

Introduction

Human African trypanosomiasis (HAT) is caused by Trypanosoma brucei (T. b.) gambiense in West and Central Africa, and by T. b. rhodesiense in East and Southern Africa. Epidemics of the disease occur, with the Democratic Republic of Congo, Angola and Sudan the most afflicted countries in recent years [1]. Disease control is based on the implementation of appropriate public health policies, which remains difficult in countries with limited resources subject to political unrest. Humans contract the disease after being bitten by an infected tse-tse fly. The trypanosomes move out from the site of inoculation, invading all parts of the body via the bloodstream and lymphatic system (stage 1). The encephalitic stage (stage 2) begins when parasites cross the blood-brain barrier, modifying sleep-wake cycles, endocrinological functions and behavior. Over time, the entire central nervous system (CNS) may be affected, with sensory and motor dysfunctions progressively leading to irreversible demyelinating disease [2]. It has been suggested that an autoimmune response against neuronal and myelin antigens occurs in cases of CNS involvement. Indeed, autoantibodies reacting with galactocerebrosides [3], the major components of myelin, and with neurofilaments [4], the intermediate filaments of neurons, have been reported. The time-course of the disease varies widely due to the complexities of the host-parasite interaction, resulting in immune system-driven mechanisms with both beneficial and deleterious effects on the body [5][6]. The immunophenotyping of lymphocytes in patients with viral infections, immunodeficiencies and autoimmune diseases has provided clues to the pathogenesis of these diseases [7][8]. Furthermore, the patterns of lymphocyte activation observed during the course of Chagas' disease, caused by a trypanosome endemic in South America (Trypanosoma cruzi), may provide an explanation for the cardiac disease occurring during infection [9][10]. In HAT, the mechanisms of host-parasite interaction and the subsequent development of a demyelinating disease in the CNS are not well understood. Classically and alternatively activated macrophages play a key role in initiating the immune response and disease control [11][13]. Indeed, the protection of the host against lesion formation depends on the ability to downregulate alternatively activated macrophages. The subsequent stages of cellular immune profile during the course of the disease have not been extensively studied in humans and remain unclear. We therefore carried out a clinical study with the aim of describing the phenotypic changes occurring in subsets of lymphocyte during HAT.

Results

Enrollment of patients and clinical data

Survey 1 was performed in November 2002 in Angola (Viana and Bengo districts) and in Gabon (N'kembo Hospital). Blood samples from 17 patients (16 Angolans, 1 Gabonese) and 13 controls (Gabonese from N'kembo) were analyzed. Survey 2 was performed in June 2004 in Angola (Bengo district) and Gabon (Estuaire district). Blood samples from 16 Angolan patients and 14 controls (6 Gabonese, 8 Angolans) were analyzed. All subjects belonged to the Bantu ethnic group and were of similar socioeconomic status. They were matched for age and sex with controls whenever possible. The epidemiological and clinical characteristics and disease stages of the patients are summarized in Table 1. Briefly, clinical and biological data allowed us to identify 17 patients in stage 2 (S2), 11 patients in stage 1 (S1) and 9 patients in intermediate stage (S-int) as defined in material and methods below.

thumbnail
Table 1. Clinical and epidemiological features of the subjects studied.

https://doi.org/10.1371/journal.pone.0006184.t001

Blood lymphocyte subset profiles

B and T cell activation markers in blood.

The general profile of lymphocyte subsets common to both surveys (CD19, CD3, CD4, CD8) is presented in figure 1 for 33 HAT patients and 27 controls. Patients had significantly larger numbers of B (CD19+) cells (Table 2) and significantly fewer T (CD3+) cells (Figure 1) than controls (P = 0.003 and 0.04, respectively). The CD4/CD8 ratios of patients and controls were similar (Table 3). Lymphocyte subsets did not differ significantly between S-1, S-int and S-2 patients (P>0.05).

thumbnail
Figure 1. Distribution of B cells (CD19+), T cells (CD3+), and T-cell subpopulations (CD3+CD4+ or CD3+CD8+) in the blood of 33 HAT patients and 27 controls (surveys 1 and 2 combined).

For each box plot, the features limits correspond, from bottom to top, to the 90th, 75th, 50th (median), 25th and 10th percentiles.

https://doi.org/10.1371/journal.pone.0006184.g001

thumbnail
Table 2. B cells (CD19+) and B-cell activation markers (CD69+ and CD95+) in peripheral blood mononuclear cells for surveys 1 and 2.

https://doi.org/10.1371/journal.pone.0006184.t002

thumbnail
Table 3. T cells subsets (CD4+/CD8+) and T-cell activation markers (HLA-DR+, CD69+, CD25+, CD45RA+, CD45RA, CD62L) in peripheral blood mononuclear cells for surveys 1 and 2.

https://doi.org/10.1371/journal.pone.0006184.t003

The results for B cells (CD19+) in 33 patients and 27 controls (survey 1 and 2) and for B-cell activation markers (CD69+, CD95+) in 16 patients and 14 controls (survey 2) are presented in Table 2. Patients displayed significantly stronger expression of B-cell activation markers than did controls (P = 0.03 and 0.0001, respectively).

The results for CD4+ and CD8+ T cells (CD3+) in 33 patients and 27 controls (surveys 1 and 2) and for T-cell activation markers are presented in Table 3. The results for lymphocyte markers analyzed exclusively in survey 1 (CD25, CD95) or survey 2 (CD45RA, CD69L, CD28, HLA-DR) were analyzed separately. We analyzed the results from 17 patients and 13 controls in survey 1 and 16 patients and 14 controls in survey 2 (Table 3).

NK cells were defined as CD56+ lymphocytes in survey 1 and as CD16+ lymphocytes in survey 2. The relative percentages in patients and controls were therefore compared separately in surveys 1 and 2. No differences were observed between patients and control (P>0.05, data not shown).

The percentages of CD4 and CD8 T cells were similar in patients and controls. Similar levels of T lymphocyte activation, as shown by evaluations of CD4+HLA-DR+, CD8+HLA-DR+, CD4+CD69+ and CD8+CD25+ cells, were observed in patients and controls (Table 3). However, CD4+CD25+ T-lymphocyte levels were significantly lower in patients than in controls (P = 0.02). The proportions of the naive CD4+CD45RA+ and CD8+CD45RA+ T-cell subsets were similar in patients and controls, but the proportion of naive T cells expressing CD62L was higher in patients than in controls (P = 0.03). Similarly, patients had larger numbers of memory CD4+CD45RACD62L+ cells (P = 0.01). The pre-effector T lymphocytes populations (CD8+CD45RACD62L) were of similar size in patients and controls (Table 3), but patients had fewer effector T lymphocytes (CD8+CD45RA+CD62L) (P = 0.01).

CSF lymphocyte subset profiles and correlation with disease stage.

Logistic problems with CSF cell storage before FC analysis resulted in some results being unexploitable. We therefore restricted our analysis to interpretable results. We thus present, in Table 4, only the results for B cells (CD19+) for 4 controls, 8 S-1, 8 S-int and 13 S-2 patients. An example of the FC images obtained is provided in Figure 2.

thumbnail
Figure 2. Results of CSF analysis by flow cytometry in a HAT patient with a cell count of 63 cells/µl.

https://doi.org/10.1371/journal.pone.0006184.g002

thumbnail
Table 4. B cells (CD19+) in the CSF of HAT patients and controls.

https://doi.org/10.1371/journal.pone.0006184.t004

No B cells (CD19+) were detected in controls (<0.01 cells/µl). The percentages of B cells in CSF samples from controls, S-1 and S-int patients were significantly lower than those in S-2 patients (P<0.01). In S-2 patients, B cells accounted for 50% of the cells present (243 cells/µl). The number of B cells increased between control and S-1 (factor of 10), between S-1 and S-int (factor of 5) and between S-int and stage 2 (factor of 500), although the differences between subgroups of patients were not significantly different.

Discussion

We provide here the first description of differences in lymphocyte subsets in blood and CSF between HAT patients and controls from the same ethnic group and geographic area. Despite the preliminary nature of our results, the small sample size and the logistic problems encountered, we identified several interesting features consistent with known data and experimental findings.

Patients had larger numbers of B cells (CD19+) and activated B cells (CD19+CD69+) than controls, consistent with the immune response classically reported for HAT, with the extensive polyclonal activation of cell likely to proliferate and to secrete large amounts of antibodies [14], [15]. We clearly observed an upregulation of Fas (CD95) expression on the B cells of patients, consistent with the activation of cells via CD95, which may be involved in subsequent lymphocyte apoptosis. Cell activation seems to be self-regulated by the increase in B-cell apoptosis, as described in T. cruzi infection [16], [17], suggesting poor conversion of B cells into memory B cells [18]. Radwanska et al. (2008) [19] recently demonstrated that the parasite causes a significant decrease in various B-cell populations in mice. These changes may account for the inability of the host to develop protective anti-parasite antibody responses and the lack of development of an effective B-cell memory response against parasites with variant antigens.

This strong B-cell response during infection may affect the homeostasis of T-cell subsets. This phenomenon has been observed in HIV infections [20], [21]. HAT-infected patients have higher percentages of T cells than HIV patients, with no change in CD4/CD8 ratio and an increase in the proportion of naive T (CD62L+CD45RA+) cells [22]. Indeed, the CD4/CD8 ratio has been shown to increase in autoimmune diseases and to decrease during viral infections, but remains unaffected in HAT. Naive lymphocytes, when stimulated by an antigen, develop into memory lymphocytes, which subsequently differentiate into effector lymphocytes. This process facilitates the rapid and effective secondary response required for long-term protection of the host [23], [24]. We found that the proportion of CD8 T effector cells (CD8+CD45RA+CD62L) was lower in patients than in controls. Patients also had fewer CD4+CD25+ cells than controls, accounting for the lower level of activation/proliferation of CD4 T cells in patients. These findings suggest that there may be a T-cell response dysfunction, in patients, possibly due to continuous stimulation of the immune system by new antigenic variants from trypanosomes, resulting in a lack of memory and effector lymphocytes. Thus, the adaptive immune response seems to be impaired in HAT. T cells act as effectors either through cell-mediated cytotoxicity, following the release of dense cytoplasmic granules containing perforin and granzymes, or through the production of cytokines, including IFN-γ in particular, thereby activating other effector cells, such as antigen-presenting cells. On the other hand, some of CD25-expressing T cells belong to the Treg subset populations that represent 10% of the CD4 T cells in humans. The natural Treg cells, CD4+CD25highFoxp3+ were recently studied in experimental model of African trypanosomiasis in Trypanosoma congolense - infected mice [25]. This study demonstrated that Treg cells play an important role in trypanotolerance, through a decrease of INF-γ production and downregulation of the inflammatory response depending on IL-10 [25]. Experiment with injection of anti-CD25 on animals before challenge, resulted in the absence of development of parasitemia and elimination of all parasites [26].

Furthermore, the proportion of NK cells was not higher in patients than in controls suggesting that this cell subset of innate immune response was not modified in HAT. However, these results require confirmation with a larger cohort and specific markers.

Finally, the B- and T-cell subset profiles observed are consistent with the classical immunomodulatory mechanisms described in experimental models of HAT [27][29]. Direct (parasite-released) and indirect (host cells) mechanisms of immunomodulation are observed. The trypanosome-derived lymphocyte triggering factor (TLTF), also known as trypanin, induces the production of TGF-β mRNA in CD8 T cells and has an immunosuppressive effect [30]. In lymph node T-cell proliferation responses, the classical subset of macrophages plays a central role in inhibiting T-cell proliferation. Early-activated macrophages (by IFN-γ activation, Th1 cytokine pattern) inhibit T-cell proliferation most strongly, through several mechanisms: i) the production of prostaglandin, leading to the abolition of IL-2 production [31], ii) the induction of a prostaglandin-independent mechanism accounting for the abolition of IL-2 receptor (IL-2R/CD25) expression [32] and iii) NO synthesis induced by IFN-γ and resulting in impaired mitogen-induced T-cell proliferation in the spleen, peritoneal cavity and lymph nodes of T. brucei-infected mice [33]. Later in the course of infection, T-cell proliferation in lymph nodes may be inhibited through other mechanisms, mediated by an NO-independent pathway and depending on IFN-γ release from CD8 T cells [33], [34]. Alternatively activated macrophages have also been reported to play a role in experimental models of African trypanosomiasis [12], [33]. These cells are activated in the presence of IL-4 and contribute to the development of a Th2 cytokine pattern, which decreases inflammatory responses in the host [35]. Together, these mechanisms impair T-cell activation, resulting in a hyporesponsiveness of T cells to parasite-related and unrelated antigens [12], [33].

We report here the first analysis of the lymphocyte subsets expressed in patients with HAT. The relationship between lymphocyte phenotype and final clinical outcome remains unclear, but our results suggest that various immunosuppressive mechanisms may enable parasites to evade the immune system long enough to cross the blood-brain barrier and hide within the CNS [36]. Logistic problems prevented a more comprehensive analysis of the CSF samples, but our results for this small sample strongly suggest that the progressive increase in the proportion of activated B lymphocytes in the CSF may be an important indicator of stage 2 disease.

Finally, our findings provide new insight into the immunopathological mechanisms underlying HAT and highlight the importance of B lymphocytes for the diagnosis of CNS invasion and disease. B lymphocytes marked with beads coupling with anti-CD19 can be visualized under the microscope for fresh lumbar puncture samples, and this detection could be used in a field-adapted diagnostic tool for determining the stage of the disease. We are currently validating these results in a larger sample.

Materials and Methods

Subjects

The study was approved by the Angolan and Gabonese ministries of health and was conducted in accordance with the Helsinki declaration. Informed, written consent was obtained from all included subjects. Subjects were selected actively, during field surveys in Angola and Gabon, and passively, at the reference hospitals at Viana (Angola) and N'kembo (Gabon). Two surveys were carried out in 2002 (survey 1) and 2004 (survey 2). Patients were selected on the basis of whole-blood screening for antibodies with the card agglutination trypanosomiasis test (CATT) [37]. Patients were defined as subjects with a positive CATT test confirmed by parasite detection. Controls were obtained in the same area of endemic disease and had a negative CATT test, with no detection of parasites in the blood after the use of concentration techniques. Subjects presenting evidence of coinfections (malaria, filariasis, HIV, syphilis) or under the age of 10 years were excluded from this study. All subjects were examined by a doctor and clinical examination findings were noted on a case report form.

Sampling

Blood was obtained during normal diagnostic procedures. We collected 10 ml of blood by venipuncture. The blood sample was then divided in two, with one tube used for subtyping by immunostaining and flow cytometry (FC) (tube containing EDTA) and the other (tube without anticoagulant) used for serological analysis. The storage of blood samples for FC analysis differed between the two surveys:

- In survey 1, staining was carried out with whole blood and monoclonal antibodies (mAbs), as described below, in the field and was then fixed in Cytocheck® (Dako France, Trappes, France) and stored at +4°C until analysis within five days.

- In survey 2, peripheral blood mononuclear cells were isolated in the field by density gradient centrifugation in Vacutainer® CPT™ cell preparation tubes containing sodium heparin (Becton Dickinson, Franklin Lakes, NJ). These cells were then frozen in the presence of fetal bovine serum-10% dimethyl sulfoxide (Sigma-Aldrich, Lyons, France), in liquid nitrogen.

CSF samples were obtained from patients during normal diagnostic procedures. Four CSF samples were obtained from patients consulting for disturbed sleep with minor neurological signs but no detection of trypanosomes in the blood or CSF and a normal CSF cell count. We collected 6 ml of CSF into a conical tube. We centrifuged 4 ml of the sample and dispensed the supernatant into aliquots, which were stored in liquid nitrogen. The remaining 2 ml of the CSF sample was used to determine disease stage in the field. Lumbar puncture samples containing blood (>500 red blood cells/µl) were discarded and the patients from whom such samples were obtained were excluded from the study. CSF samples were immediately stained for FC, fixed with 10% CellFix (Becton Dickinson) and stored at +4°C.

Parasitological and stage determination procedures

Positive CATT results were confirmed by parasite detection in blood or swollen lymph nodes. A modified miniature anion-exchange centrifugation technique was used for blood [38]. Lymph nodes were punctured and the fluid obtained was observed under the microscope.

Disease stage was diagnosed on the basis of clinical examination and lumbar puncture. CSF was examined immediately after puncture. Cells were counted on Kova slides (Hycor Biomedical Inc., Gardengrove, CA). Parasite detection was carried out by microscopic examination of 2 ml of CSF, after simple centrifugation [39].

(i) Stage 1 (S-1) was defined as the presence of fewer than 5 cells/µl in CSF, and the absence of trypanosomes on microscopic examination.

(ii) Stage 2 (S-2) was defined as the presence of more than 20 cells/µl CSF and/or the presence of trypanosomes on microscopic examination.

(iii) Patients were classified as having intermediate stage disease (S-int) if their CSF profile differed from those described above - i.e, between 5 and 20 cells/µl - or if trypanosomes were observed on microscopy but there were fewer than 5 cells/µl CSF [40].

Clinical examination was performed jointly to confirm the diagnosis. The detection of neurological signs in S-int patients was indicative to treat with second stage drugs.

FC analysis

Samples were analyzed at the CIRMF in Gabon (2000 km from the site of enrollment and sampling in Angola), within five days of sample collection. The mAbs used were obtained from Beckman Coulter (Fullerton, CA, USA) and Becton Dickinson (BD, Franklin Lakes, NJ, USA) (see Table 5 and 6). The results of survey 1 determined the selection of mAbs for use in survey 2.

thumbnail
Table 5. Monoclonal antibodies (mAbs) used in surveys 1 and 2. Source: BC, Beckman Coulter (Fullerton, CA, USA); BD Becton Dickinson (Franklin Lakes, NJ, USA).

https://doi.org/10.1371/journal.pone.0006184.t005

thumbnail
Table 6. Cell lymphocyte populations targeted in surveys 1 and 2.

https://doi.org/10.1371/journal.pone.0006184.t006

Fluorescence intensity was determined with a three-color fluorescence-activated cell sorter machine (FACSCalibur, Becton Dickinson, Franklin Lakes, NJ, USA) in survey 1 and with a four-color machine in survey 2. Lymphocyte cells were gated on the basis of forward and side light-scattering properties and positively selected on the basis of cluster of differentiation (CD) molecule expression. Isotype-matched control mAbs were used to define background fluorescence. Before data acquisition, we checked and optimized the instrument parameters with CaliBRITE Beads (Becton Dickinson). On average, 104 events were acquired with FACSCalibur and analyzed with CELLQUEST software (Becton Dickinson).

During survey 1, we stained 50 µl of total blood from each sample (using 2 µl for each antibody) within 20 minutes. The blood sample was then lysed with a lysis buffer (8.29 g ammonium chloride, 1 g potassium bicarbonate, 0.037 g EDTA in 1l of distilled water, pH adjusted to 7.2), and washed twice with PBS to select leukocytes only. During survey 2, lymphocytes (104) were retrieved from frozen peripheral blood mononuclear cells by thawing and washed in PBS. Their viability was then checked with trypan blue and they were stained with 2 µl mAbs within 20 minutes. All samples were fixed in 4% PFA and analyzed within 5 days. For CSF, we added 2 µl of mAbs to 100 µl of CSF. We then incubated this mixture for 15 minutes at +4°C, fixed the cells and analyzed them within 5 days.

Statistical analysis

The proportions (% of total events acquired) of each lymphocyte type were calculated after acquisition on a FACSCalibur machine.

The markers used in both surveys (CD19, CD3, CD4, CD8, CD95) were analyzed for all the patients and controls tested, whereas markers used only in survey 1 (CD56, CD25, CD40) or in survey 2 (CD45RA, CD69L, CD28, CD16, HLA-DR) were analyzed separately. Due to the small sample size, the Mann-Whitney non parametric U test was used for comparisons, with P values below 0.05 considered statistically significant.

Acknowledgments

We would like to thank the staff of the Instituto de Combate e Controlo das Tripanossomiases (Angola) and Dr Carmelo Santercole for their help and assistance in the field. We would also like to thank Jean Luc Faucher and Sandrine Souquière for technical assistance for FC and Preston A. Marx for mAbs. We would also like to thank Dr Jeanne Cook-Moreau and Dr Dennis J. Grab who kindly agreed to correct this manuscript.

Author Contributions

Conceived and designed the experiments: CB BC MOJM SB. Performed the experiments: CB BC LP SB. Analyzed the data: CB BC PR MOJM SB. Contributed reagents/materials/analysis tools: CB BC GV TJ CRA BB SB. Wrote the paper: CB BC PR MOJM SB.

References

  1. 1. Barrett MP (2006) The rise and fall of sleeping sickness. Lancet 29: 1377–1378.
  2. 2. Bouteille B, Dumas M (2003) Human African trypanosomiasis. In: Aminoff M, Daroff R, editors. Encyclopedia of the Neurological Sciences. San Diego (CA, USA): Academic Press. vol. 2.
  3. 3. Amevigbe M, Jauberteau-Marchan MO, Bouteille B, Doua F, Breton JC, et al. (1992) Human African trypanosomiasis: presence of antibodies to galactocerebrosides. Am J Trop Med Hyg 47: 652–662.
  4. 4. Ayed Z, Brindel I, Bouteille B, Van Meirvenne N, Doua F, et al. (1997) Detection and characterization of autoantibodies directed against neurofilament proteins in human African trypanosomiasis. Am J Trop Med Hyg 57: 1–6.
  5. 5. Bisser S, Ouwe-Missi-Oukem-Boyer ON, Toure FS, Taoufiq Z, Bouteille B, et al. (2006) Harbouring in the brain: A focus on immune evasion mechanisms and their deleterious effects in malaria and human African trypanosomiasis. Int J Parasitol 36: 529–540.
  6. 6. Kennedy PG (2006) Diagnostic and neuropathogenesis issues in human African trypanosomiasis. Int J Parasitol 36: 505–512.
  7. 7. Heinrich A, Ahrens N, Schmidt S, Khaw AV (2006) Immunophenotypic patterns of T-cell activation in neuroinflammatory diseases. Acta Neurol Scand 113: 248–255.
  8. 8. Xu D, Fu J, Jin L, Zhang H, Zhou C, et al. (2006) Circulating and liver resident CD4+CD25+ regulatory T cells actively influence the antiviral immune response and disease progression in patients with hepatitis. B J Immunol 177: 739–747.
  9. 9. Mirkin GA, Celentano AM, Malchiodi EL, Jones M, Gonzalez Cappa SM (1997) Different Trypanosoma cruzi strains promote neuromyopathic damage mediated by distinct T lymphocyte subsets. Clin Exp Immunol 107: 328–334.
  10. 10. Piedras J, Gutierrez S, Reyes-Lopez PA, Reyes K, Lopez-Karpovitch X, et al. (1997) Circulating lymphocyte subpopulations and activated T and B cells in patients with chagasic and non-chagasic myocardiopathy. Cytometry 30: 28–32.
  11. 11. Boda C, Courtioux B, Jauberteau-Marchan MO, Bisser S (2005) Immunity to African trypanosomiasis. Garraud O, editor. Trivandrum: Research Signpost. pp. 255–277. Update in Tropical Immunology.
  12. 12. Namangala B, Brys L, Magez S, De Baetselier P, Beschin A (2000) Trypanosoma brucei brucei infection impairs MHC class II antigen presentation capacity of macrophages. Parasite Immunol 22: 361–370.
  13. 13. Vincendeau P, Bouteille B (2006) Immunology and immunopathology of African trypanosomiasis. An Acad Bras Cienc 78: 645–665.
  14. 14. Anthoons JA, Van Marck EA, Gigase PL (1986) Autoantibodies to intermediate filaments in experimental infections with Trypanosoma brucei gambiense. Z Parasitenkd 72: 443–452.
  15. 15. Kazyumba G, Berney M, Brighouse G, Cruchaud A, Lambert PH (1986) Expression of the B cell repertoire and autoantibodies in human African trypanosomiasis. Clin Exp Immunol 65: 10–18.
  16. 16. Minoprio P, Itohara S, Heusser C, Tonegawa S, Coutinho A (1989) Immunobiology of murine T. cruzi infection: the predominance of parasite-nonspecific responses and the activation of TCRI T cells. Immunol Rev 112: 183–207.
  17. 17. Zuniga E, Motran C, Montes CL, Diaz FL, Bocco JL, et al. (2000) Trypanosoma cruzi-induced immunosuppression: B cells undergo spontaneous apoptosis and lipopolysaccharide (LPS) arrests their proliferation during acute infection. Clin Exp Immunol 119: 507–515.
  18. 18. Acosta-Rodriguez EV, Montes CL, Motran CC, Zuniga EI, Liu FT, et al. (2004) Galectin-3 mediates IL-4-induced survival and differentiation of B cells: functional cross-talk and implications during Trypanosoma cruzi infection. J Immunol 172: 493–502.
  19. 19. Radwanska M, Guirnalda P, De Trez C, Ryffel B, Black S, et al. (2008) Trypanosomiasis-induced B cell apoptosis results in loss of protective anti-parasite antibody responses and abolishment of vaccine-induced memory responses. PLoS Pathog 4: e1000078.
  20. 20. Ho DD, Neumann AU, Perelson AS, Chen W, Leonard JM, et al. (1995) Rapid turnover of plasma virions and CD4 lymphocytes in HIV-1 infection. Nature 373: 123–126.
  21. 21. Mohri H, Bonhoeffer S, Monard S, Perelson AS, Ho DD (1998) Rapid turnover of T lymphocytes in SIV-infected rhesus macaques. Science 279: 1223–1227.
  22. 22. Roederer M, Dubs JG, Anderson MT, Raju PA, Herzenberg LA (1995) CD8 naive T cell counts decrease progressively in HIV-infected adults. J Clin Invest 95: 2061–2066.
  23. 23. Ahmed R, Gray D (1996) Immunological memory and protective immunity: understanding their relation. Science 272: 54–60.
  24. 24. Fearon DT, Manders P, Wagner SD (2001) Arrested differentiation, the self-renewing memory lymphocyte, and vaccination. Science 293: 248–250.
  25. 25. Guilliams M, Oldenhove G, Noel W, Hérin M, Brys L, et al. (2007) African trypanosomiasis: naturally occurring regulatory T cells favor trypanotolerance by limiting pathology associated with sustained type 1 inflammation. J Immunol 179: 2748–2757.
  26. 26. Tabel H, Wei G, Shi M (2008) T cells and immunopathogenesis of experimental African trypanosomiasis. Immunol Rev 225: 128–139.
  27. 27. Millar AE, Sternberg J, McSharry C, Wei XQ, Liew FY, et al. (1999) T-cell responses during Trypanosoma brucei infections in mice deficient in inducible nitric oxide synthase. Infect Immun 67: 3334–33348.
  28. 28. Reinitz DM, Mansfield JM (1990) T-cell-independent and T-cell-dependent B-cell responses to exposed variant surface glycoprotein epitopes in trypanosome-infected mice. Infect Immun 58: 2337–2342.
  29. 29. Sendashonga CN, Black SJ (1986) Analysis of B cell and T cell proliferative responses induced by monomorphic and pleomorphic Trypanosoma brucei parasites in mice. Parasite Immunol 8: 443–453.
  30. 30. Bakhiet M, Büscher P, Harris RA, Kristensson K, Wigzell H, et al. (1996) Induction of interferon-gamma, transforming growth factor-beta, and interleukin-4 in mouse strains with different susceptibilities to Trypanosoma brucei brucei. J Interferon Cytokine Res 16: 427–433.
  31. 31. Sileghem M, Darji A, Hamers R, Van de Winkel M, De Baetselier P (1989a) Dual role of macrophages in the suppression of interleukin 2 production and interleukin 2 receptor expression in trypanosome-infected mice. Eur J Immunol 19: 829–835.
  32. 32. Sileghem M, Darji A, Remels L, Hamers R, De Baetselier P (1989b) Different mechanisms account for the suppression of interleukin 2 production and the suppression of interleukin 2 receptor expression in Trypanosoma brucei-infected mice. Eur J Immunol 19: 119–124.
  33. 33. Namangala B, De Baetselier P, Noel W, Brys L, Beschin A (2001) Alternative versus classical macrophage activation during experimental African trypanosomosis. J Leukoc Biol 69: 387–396.
  34. 34. De Baetselier PD, Namangala B, Noël W, Brys L, Pays E, et al. (2001) Alternative versus classical macrophage activation during experimental African trypanosomosis. Int J Parasitol 31: 575–587.
  35. 35. Gobert AP, Daulouede S, Lepoivre M, Boucher JL, Bouteille B, et al. (2000) L-Arginine availability modulates local nitric oxide production and parasite killing in experimental trypanosomiasis. Infect Immun 68: 4653–4657.
  36. 36. Grab DJ, Kennedy PG (2008) Traversal of human and animal trypanosomes across the blood-brain barrier. J Neurovirol 14: 344–351.
  37. 37. Magnus E, Vervoort T, Van Meirvenne N (1978) A CARD-agglutination test with stained trypanosomes (C.A.T.T.) for the serological diagnosis of T. b. gambiense trypanosomiasis. Ann Soc Belg Med Trop 58: 169–176.
  38. 38. Lumsden WH, Kimber CD, Strange M (1977) Trypanosoma brucei: detection of low parasitaemias in mice by a miniature anion-exchanger/centrifugation technique. Trans R Soc Trop Med Hyg 71: 421–424.
  39. 39. Miezan TW, Meda HA, Doua F, Dje NN, Lejon V, et al. (2000) Single centrifugation of cerebrospinal fluid in a sealed Pasteur pipette for simple, rapid and sensitive detection of trypanosomes. Trans R Soc Trop Med Hyg 94: 293.
  40. 40. Bisser S, Lejon V, Preux PM, Bouteille B, Stanghellini A, et al. (2002) Blood-cerebrospinal fluid barrier and intrathecal immunoglobulins compared to field diagnosis of central nervous system involvement in sleeping sickness. J Neurol Sci 193: 127–135.