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

Regulatory and Activated T Cells in Human Schistosoma haematobium Infections

  • Norman Nausch ,

    nnausch@staffmail.ed.ac.uk

    Affiliation Ashworth Laboratories, School of Biological Sciences, Institute for Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom

  • Nicholas Midzi,

    Affiliation National Institute of Health Research, Harare, Zimbabwe

  • Takafira Mduluza,

    Current address: Harvard AIDS Institute, Gaborone, Botswana

    Affiliation Department of Biochemistry, University of Zimbabwe, Harare, Zimbabwe

  • Rick M. Maizels,

    Affiliation Ashworth Laboratories, School of Biological Sciences, Institute for Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom

  • Francisca Mutapi

    Affiliation Ashworth Laboratories, School of Biological Sciences, Institute for Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom

Abstract

Acquired immunity against helminths is characterised by a complex interplay between the effector Th1 and Th2 immune responses and it slowly manifests with age as a result of cumulative exposure to parasite antigens. Data from experimental models suggest that immunity is also influenced by regulatory T cells (Treg), but as yet studies on Treg in human schistosome infections are limited.

This study investigated the relationship between schistosome infection intensity and the two cell populations regulatory T cells (Treg: CD4+(dim)CD25+(high)FOXP3+CD127low), and activated (Tact: CD4+CD25+FOXP3) T cells in Zimbabweans exposed to Schistosoma haematobium parasites. Participants were partitioned into two age groups, young children (8–13 years) in whom schistosome infection levels were rising to peak and older people (14+ years) with declining infection levels. The relationship between Tact proportions and schistosome infection intensity remained unchanged with age. However Treg proportions rose significantly with increasing infection in the younger age group. In contrast Treg were negatively correlated to infection intensity in the older age group.

The relative proportions of regulatory T cells differ significantly between young individuals in whom high infection is associated with an enhanced regulatory phenotype and older infected patients in whom the regulatory response is attenuated. This may influence or reflect different stages of the development of protective schistosome acquired immunity and immunopathogenesis.

Introduction

Schistosomiasis is a major human parasitic disease caused by different species of trematode parasites of the genus Schistosoma. Urogenital schistosomiasis caused by S. haematobium is the most prevalent form in sub-Saharan Africa where it is associated with pathology ranging from hematuria to more severe conditions such as hydronephrosis and bladder cancer [1].

The role of acquired immunity in reducing human schistosome infection intensity and pathology has been subject to intense study [2], [3], [4]. Early studies suggested that anti-schistosome immune responses fell into a clear Th1/Th2 dichotomy with resistance to infection being associated with Th2 responses [5], [6], but mounting evidence shows this dichotomy cannot adequately explain the balance between susceptibility and resistance to infection [7], [8]. For example, we have previously shown that neither Th1 nor Th2 cytokine responses in S. haematobium-infected Zimbabwean children show a clear pattern with infection intensity [9], [10] supporting similar reports on S. manosni vaccine candidates [11] and other immuno-epidemiology studies (reviewed in [12], [13]).

The discovery that a regulatory subset of CD4+ T cells (Treg) modulates the activity of effector Th1 and Th2 responses in mouse models of schistosomiasis [14] has led to suggestions that it is the balance between effector and regulatory responses including Treg, which determines the outcome of murine helminth infections [15], has also been postulated for human helminth infections [16], [17]. This concept is not unique to helminth infections; regulatory T cells have been invoked across a wide range of infectious diseases including fungal, bacterial and viral infections [18], [19], [20], [21], [22] and non infectious diseases such as allergies or cancer [23], [24], and modulation of immunopathology by regulatory T cells has been reported in human and mouse models [25], [26], [27], [28].

There have been several studies of the phenotype and role of Treg cells in mouse models of helminth infections [15], [29], [30], [31] including schistosomiasis [27], [32], [33], [34], but to date there is a paucity of studies of human Treg responses in helminth infections. Human Treg were initially identified as those expressing the highest levels of CD25 [35], and using this marker it was reported that regulatory and effector cells show an inverse relationship in schistosomiasis [36]. In addition a study in Kenyan children found differences in activated T cells and memory regulatory T cells between individuals with S. mansoni only compared to individuals concurrently infected with S. mansoni and Plasmodium falciparum [37].

The discovery of the forkhead/winged helix transcription factor FOXP3 as a marker for these cells allows a more accurate characterization of Treg [38], [39], [40]. Some studies suggested that human FOXP3+ cells (in contrast to mouse FoxP3+ Treg) are functionally heterogeneous [41] and that transient expression of FOXP3 on activated T cells does not correlate with suppressive function [42]. However FOXP3 is still considered the most accurate marker for naturally occurring Treg [43]. Recent studies have further characterized the phenotype of Treg by the expression of low levels of CD127 [44] and slightly diminished levels of CD4 [45]. By using a combination of these markers we have investigated the relationship between Treg, effector T cells and schistosome infection intensity. The aim of the study was to determine if the percentage of activated T cells (Tact, defined as CD4+CD25+FOXP3) or regulatory T cells (Treg, defined as CD4+CD25+(high)FOXP3+CD127dim) differed with schistosome infection intensity.

Materials and Methods

Ethics Statement

Permission to conduct the study in the region was obtained from the Provincial Medical Director and institutional and ethical approval was received from the University of Zimbabwe's Institute Review Board (UZIRB) and the Medical Research Council of Zimbabwe (MRCZ) respectively. Only compliant participants were recruited into the study and they were free to drop out at any point during the study. At the beginning of the study, participants and their parents/guardians had the aims and procedures of the project explained fully in the local language, Shona, and oral consent was obtained from participants and parents/guardian before parasitology and blood samples were obtained. Oral informed consent was obtained because of the high levels of illiteracy and cultural reasons [46]. Both the UZIRB and MRZC approved the use of oral consent. Upon oral consent participants were enrolled in the study with a written record of their name, age, sex and case number, this served as both the record of oral consent and enrolment record.

Study population and parasitology

The study was conducted in the Mashonaland East Province of Zimbabwe (31o30′E; 17o45′S) where S. haematobium is endemic and where the participants have been participating in an ongoing study of the immuno-epidemiology of human schistosomiasis [47], [48]. The villages were selected because health surveys regularly conducted in the region showed little or no infection with other helminths and a low S. mansoni prevalence (<5%) [49], [50]. The two villages selected (Goromonzi and Mutoko) had not been included in the National Schistosome Control Programme and therefore participants had not received antihelminthic treatment for schistosomiasis or other helminth infections meaning that their natural immune responses could be studied in the absence of drug-altered schistosome responses [51]. The main activity in these villages is subsistence farming and human water contact is frequent with at least 4 contacts/person/week (assessed by questionnaire, adapted from [52], [53]) due to insufficient safe water and sanitation facilities. Drinking water is collected from open wells while bathing and washing is conducted in two main rivers in the villages. The schools surveyed in the two villages were all in close proximity to rivers.

Helminth infection intensity was assessed by microscopic examination of urine and stool samples. Urine specimens were processed by urine filtration following a standard method originally described by Mott et al. [54] for diagnosis of S. haematobium. Fresh stool specimens were processed by the Kato-Katz technique and subsequently analysed by microscopy for intestinal helminths including S. mansoni, hookworm, Ascaris lumbricoides and Trichuris trichiura. The formol-ether concentration method was performed on 50% of the samples to confirm results obtained by the Kato-Katz technique [55]. Stool and urine specimens were collected on three consecutive days.

In order to be included in the cohort, participants had to meet all the following criteria which are comparable to other studies performed for human Schistosomiasis [36], [56], [57]: 1) be life-long residents of the schistosome-endemic area; 2) not have received prior treatment for helminth infections; 3) have provided at least 2 urine and 2 stool samples on consecutive days; 4) be negative for intestinal helminths including S. mansoni and negative for Plasmodium parasites as determined by blood smears; 5) have given a blood sample for the collection of peripheral blood mononuclear cells (PBMC). Forty-nine participants aged 8–60 years met these criteria and formed our study population. Questionnaire studies indicated that there were no significant differences in water contact measures (frequency or duration) between the age groups. After collection of all samples, all participants were offered treatment with the recommended dose of praziquantel (40 mg/kg of body weight).

Collection of PBMC

Twenty millilitres of venous blood was collected in heparinised tubes of which 5 ml was used for serological assays as well as microscopic detection of malaria parasites. Fifteen ml were used to collect peripheral blood mononuclear cells (PBMC) through density gradient centrifugation using Lymphoprep™ (Axis-Shield, Cambridgeshire, UK). These PBMC were subsequently, enumerated, cryo-preserved and stored in liquid nitrogen in Zimbabwe prior to freighting to Edinburgh in dry shippers for assay.

Phenotyping of PBMC

Thawing of cryopreserved PBMC was performed by rotating cryovials in a 37°C water bath until a small crystal was remaining in the cell suspension. Medium (RPMI 1640 supplemented with 10% Fetal bovine serum, 2 mM L-glutamine and 100 U Penicillin/Streptomycin; all Lonza, Verviers, Belgium) was slowly added to the PBMC. Cells were washed twice with the corresponding medium, counted and viability assessed using trypan blue (Sigma-Aldrich, Dorsert, UK). Afterwards cells were washed with Dulbecco's-PBS (Lonza) and surface stained with the following antibodies: Alexa488-conjugated anti-CD4 (clone OKT-4), PE-Cy7-conjugated anti-CD127 (clone eBioRDR5; all from eBiosciences, San Diego, USA), PE-conjugated anti-CD25 (clone M-A251, BD Biosciences, San Jose, USA). Intracellular staining was performed using APC-conjugated anti-FOXP3 (clone PCH101, eBioscience) and the intracellular staining set from eBioscience following the maufacturer's instructions. Stained cells were analysed on a FACSCalibur™ using CellQuestPro™ software (BD Biosciences).

Statistics

In an initial analysis was determined if the proportion of Treg and Tact relative to the total CD4+ T cells varied with host age. This analysis was conducted using a multivariate analysis of variance (MANOVA) allowing for sex (categorical; male, female), and residential village (categorical; Goromonzi, Mutoko). T cell percentage data were arcsine square root transformed to allow the use of parametric tests [58].

Subsequently the relationship between the proportions of Treg and Tact relative to total CD4+ T cells and infection intensity was determined using an analysis of variance (ANOVA). In this statistical test, the dependent variable was infection intensity (log 10(x+1) transformed), and the independent variables were Tact and Treg transformed as above. The possible confounding variables sex, village (categorised as before) and age (categorical; age group 1 (8–13 years), and age group 2 (14+ years)) were allowed for by using sequential sums of squares. Following the ANOVA the relationship between the between the two T cell subsets and the infection intensity was analysed using residuals for infection intensity (log10(x+1) transformed) after allowing for the effects of sex and village (as before) using a regression analysis.

A description of these statistical methods has been published previously by Mutapi and Roddam [59]. All statistical tests were conducted using the software package SPSS and p values were taken to be significant at p<0.05.

Results

Schistosome epidemiology of study population

The prevalence of S. haematobium in the analysed population was 78% with a mean egg count of 28 eggs/10 ml urine (SEM = 5) with a range of 0 to 126 eggs/10 ml of urine. The population shows a typical convex age-infection profile for schistosome infection intensity, with infection intensity rising in the youngest age group (8–10 years), peaking in people aged 11–13 years and subsequently declining above 14 years of age. This infection profile corresponds to our data published previously for this population [47], [48].

Based on this profile the study population was divided into two age groups. In the first age group (8–13 years, N = 30) infection intensity is rising to peak whereas in the second group (14+, N = 19) infection intensity is declining as summarized in Figure 1. Mean infection intensity was significantly higher (p = 0.01) in the 8–13 year-group (39.4 eggs/10 ml urine ±7.3) compared to the 14+ year group (12.7 eggs/10 ml urine ± 4.9) and there were both egg negative and egg positive individuals in both groups as shown in Figure 1. The infection prevalence was significantly higher (χ2 = 5.16, df = 1, p = 0.023) in the younger group compared to the older group at 90% versus 63% respectively. All participants were negative for Plasmodium parasites as determined by microscopic examination of blood smears.

thumbnail
Figure 1. Study population was divided in two age groups.

The 8–13 year age group (N = 30) infection intensity is rising and peaking, whereas in the 14+ year age group (N = 19) infection intensity is lower. Dots represent individual values of infection intensity to illustrate distribution within the age groups and gray line indicates means. Means were compared by Student's t-Test and p-value is shown.

https://doi.org/10.1371/journal.pone.0016860.g001

Discrimination of regulatory and activated T cells

CD4+ Treg express a characteristic combination of molecular markers which distinguish them from activated effector T cells (Tact), including CD25high and FOXP3 [38], [40]. In addition, Treg show diminished levels of CD4 [45] and low levels of CD127 [44].

CD4+ lymphocytes were analyzed for the expression of CD25, FOXP3 and CD127 (Figure 2A) and gated on either CD4+CD25+FOXP+ (Treg) or CD4+CD25+FOXP (Tact) subsets (Figure 2B, C). CD25+FOXP3+ Treg expressed CD25 at high levels and showed a clear CD127low, CD4dim phenotype, allowing the identification of Treg as CD4+(dim)CD25+(high)FOXP3+CD127low cells (Figure 2A–C). In contrast Tact, identified as CD4+CD25+FOXP3 cells, showed lower CD25 expression compared to the FOXP3+ cells. In addition most Tact expressed high levels of CD127 and normal levels of CD4. The expression of CD25high, CD4low, CD127dim and FOXP3 were significantly correlated to each other (data not shown), and hence this combination of markers was used to discriminate between Tact and Treg populations.

thumbnail
Figure 2. Treg and Tact are distinguished by a combination of several markers.

(A) PBMC were electronically gated on lymphocyte population (left panel) followed by gate on total CD4+ cells (middle panel). CD4+ cells were analysed for the expression of CD25 and CD127 (right panel). (A–C) CD25+FOXP3+ cells were identified as Treg, whereas CD25+FOXP3 counted as Tact. A dot plot for one individual is shown for group 1 (B) and group 2 (C). Gating on Treg and Tact subsets was verified by further analysis of level of expression of CD25 (B–C, middle panel) and CD127 (B–C, right panel) expressed on Treg (red) and Tact (green). Gray line - isotype control.

https://doi.org/10.1371/journal.pone.0016860.g002

The proportion of CD4+ T cells classified as Treg in this study ranged from 1.7 to 10.4% whereas, the proportion of CD4+ T cells classified as Tact ranged from 5.3 to 44.8%. A sample from each of the two age groups is shown in Figure 2B and C. These analyses revealed that the expression levels of CD25 and CD127 are comparable on Treg in different age groups whereas minor changes in the expression of both markers on Tact were observed in the youngest age group. Specifically, younger people have a small subset of FOXP3 cells which show a CD127dim CD25high phenotype, these were not enumerated as Treg cells.

The percentage of activated but not regulatory T cells changes with host age

Since the T cell analysis focused on all T cells rather than schistosome-specific T cells, it was important to determine if the T cell populations varied with other host attributes. Sex did not affect proportions of CD4+CD25+FOXP3+ Treg and CD4+CD25+FOXP3 Tact relative to CD4+ T cells (statistics presented in Table 1). The proportion of CD4+CD25+FOXP3+ Treg as percentage of total CD4+ T cell numbers did not change significantly between age groups as shown in Figure 3A. Expression of different markers of Tregs in the different age groups was also compared by analyzing for the proportion of CD4+ T cells which were FOXP3+CD127dim cells (excluding CD25 as marker; Figure 3B) or the proportion of CD25high cells (Figure 3C). Regardless of the marker combination used to identify Treg, no differences were observed between the two age groups (Figure 3A–C). However the percentage of activated T cells relative to total CD4+ T cells increased significantly with host age (Figure 3D and Table 1). The latter result meant that in subsequent analyses the confounding effects of age had to be allowed for before testing for the effects of schistosome infection intensity on the T cell populations.

thumbnail
Figure 3. The proportion of Treg does not differ between age group regardless of markers employed.

Dot plots show the fraction of Treg identified by a combination of different markers: (A) CD25+FOXP3+ cells, (B) FOXP3+CD127dim cells and (C) CD25high cells as percentage of CD4+ T cells in the different age groups. In (D) percentages of CD25+FOXP3neg Tact are presented.

https://doi.org/10.1371/journal.pone.0016860.g003

The relationship between Treg and infection intensity varies with host age

The relationship between schistosome infection intensity and the percentage of regulatory and activated T cells to total CD4+ T cells was analyzed, after allowing for the effects of sex and host age in an analysis of variance. This analysis indicated that the relationship between Treg proportions and infection intensity varied between the two age groups (statistics presented in Table 2). In the young age group where infection was rising, the proportion of Treg of the total CD4+ T cell population increased significantly with increasing infection intensity (Figure 4A). The converse was true in the older age group where schistosome infection was declining, here the proportion of Treg declined with increasing infection intensity (Figure 4B). Grouping the participants by infection status (non-infected vs. infected as defined by the presence or absence of schistosome eggs in urine) rather than infection intensity produced similar results, so that in the younger age group infected participants had more Treg as proportion of CD4+ T cells than non-infected (p = 0.05), while in the older age group infected people had fewer Treg compared to non-infected (p = 0.02; data not shown). In contrast Tact showed no significant relationship with infection intensity (or infection status) in both age groups (Figure 4C, D).

thumbnail
Figure 4. The association of Treg and Tact proportions with infection intensity are presented partitioned by age group.

The variation in infection intensity ((log10 (egg count+1) transformed) was analysed for the potential confounding effects sex and village using ANOVA and calculated residuals are plotted against the proportion of Treg (A, B) or Tact (C, D; arcsin√ transformed) after dividing into two age groups as indicated. Obtained residuals were used for further used in a regression analysis whose p- and β- values are given.

https://doi.org/10.1371/journal.pone.0016860.g004

thumbnail
Table 2. Relationship between infection intensity and Treg varies between age groups.

https://doi.org/10.1371/journal.pone.0016860.t002

Discussion

An early paradigm of anti-helminth immune responses was based upon the Th1/Th2 dichotomy, with resistance to schistosome infection being associated with Th2 responses [5], [6]. Because it is now evident that the Th1/Th2 dichotomy does not sufficiently explain the balance between susceptibility and resistance to helminth infections (reviewed in [16]), there is increasing interest in the ability of regulatory T cells to modulate effector T cell responses. In mouse models, Treg play important roles in minimising pathology during schistosome infections [27], [34], and Treg depletion can restore immunity to filarial worm infections [15]. In human schistosomiasis, initial evidence for the association of regulatory T cells and infection was obtained by analysis of CD25high-expressing cell subsets [36], [60], showing a significant reduction in frequency of these cells following chemotherapeutic clearance of S. mansoni infection [36]. We conducted our studies on total populations of Treg and Tact cells rather than parasite-specific T cells to determine if an association with schistosome infection could be determined at the T cell population level. This is in keeping with the paradigm of helminth infections stimulating regulatory responses which affect responses to unrelated antigens by affecting effector T cell responses [61]. Therefore our objective was to determine the association of schistosome infection and these two T cell populations.

In this study we were able to characterise naturally occurring regulatory T cells in a human helminth infection with a more comprehensive marker set, namely CD4+(dim)CD25+(high)FOXP3+CD127low cells and analysed the relationship to activated T cells designated as CD4+CD25+FOXP3. First we observed that the proportions of Treg relative to CD4+ T cells in this population are slightly higher than those reported in populations not exposed to schistosomiasis as reported elsewhere [35], [45], but comparable to those reported for Kenyans exposed to S. mansoni [36] suggesting a relationship between exposure to the helminth infection and the development of Treg cells. Percentages of Tact were significantly higher in the older age group compared to the younger individuals. This is in accordance with data showing that CD4+ T cells with effector/memory, which encompass activated T cells, increase with age, a change which can be already observed below the age of 20 [62]. In contrast proportions of Tregs relative to CD4+ T cells did not differ significantly between the two age groups.

The most striking finding of this study was that the correlation between Treg proportion and infection intensity of S. haematobium differed significantly between the two age groups. After controlling for the confounding factors of age (and thus age related biological processes) we demonstrated that Treg as percentages of total CD4+ T cells were correlated with infection intensity, being significantly positive in the younger age group where infection was rising and peaking. In contrast the converse was true in the older age group where higher infection intensity was associated with lower Treg numbers. However there was no significant association between the infection intensity and Tact. The consequence is an increase of the ratio of regulatory to activated T cells with infection intensity in younger people (dictated by Treg) which may suggest a down-regulation of effector responses, but also may protect against immunopathology [27], [33], [63]. In contrast the correlation between proportions of regulatory T cells and level of infection turned negative in the older age group in whom infection levels were declining, suggesting that older individual carrying infection might be more prone to developing the characteristic immunopathology of chronic schistosomiasis.

However the relationship between different levels of Treg relative to infection intensity and the development immunopathology remains to be established. Furthermore detailed studies of parasite-specific T cell and functional characterization will clarify the nature and development of responses associated with resistance to human schistosome infections.

Acknowledgments

We are grateful for the co-operation of the Ministry of Health and Child Welfare in Zimbabwe, the Provincial Medical Director of Mashonaland East, the Environmental Health Workers, residents, teachers and school children in Mutoko and Rusike. We also thank Frances Jones and David Dunne from the University of Cambridge, UK for advice on field and laboratory protocols. Munyaradzi Mapingure and members of the National Institutes of Health in Zimbabwe are also acknowledged for technical support. We acknowledge Margo Chase-Topping for helpful discussion on the statistical analyses.

Author Contributions

Conceived and designed the experiments: NN TM FM. Performed the experiments: NN. Analyzed the data: NN FM. Contributed reagents/materials/analysis tools: NM TM. Wrote the paper: NN RMM FM.

References

  1. 1. W.H.O. (2002) Prevention and control of schistosomiasis and soil-transmisted helminthiasis. Geneva: World Health Organisation.
  2. 2. King CH, el Ibiary S, el Nawawi M, Sawyer J, Griffin A, et al. (1989) Intensity of Schistosoma mansoni infection in a human population is inversely correlated with antibody response to SmW68, a protective parasite antigen. J Infect Dis 160: 686–691.
  3. 3. Woolhouse MEJ, Taylor P, Matanhire D, Chandiwana SK (1991) Acquired immunity and epidemiology of Schistosoma haematobium. Nature 351: 757–759.
  4. 4. Correa-Oliveira R, Malaquias LC, Falcao PL, Viana IR, Bahia-Oliveira LM, et al. (1998) Cytokines as determinants of resistance and pathology in human Schistosoma mansoni infection. Braz J Med Biol Res 31: 171–177.
  5. 5. Medhat A, Shehata M, Bucci K, Mohamed S, Dief AD, et al. (1998) Increased interleukin-4 and interleukin-5 production in response to Schistosoma haematobium adult worm antigens correlates with lack of reinfection after treatment. J Infect Dis 178: 512–519.
  6. 6. Capron A, Dombrowicz D, Capron M (1999) Regulation of the immune response in experimental and human schistosomiasis: the limits of an attractive paradigm. Microbes Infect 1: 485–490.
  7. 7. Cheever AW, Hoffmann KF, Wynn TA (2000) Immunopathology of schistosomiasis mansoni in mice and men. Immunol Today 21: 465–466.
  8. 8. Hoffmann KF, Wynn TA, Dunne DW (2002) Cytokine-mediated host responses during schistosome infections; walking the fine line between immunological control and immunopathology. Adv Parasitol 52: 265–307.
  9. 9. Scott JT, Turner C, Mutapi F, Woolhouse ME, Chandiwana SK, et al. (2000) Dissociation of interleukin-4 and interleukin-5 production following treatment for Schistosoma haematobium infection in humans. Parasite Immunol 22: 341–348.
  10. 10. Mduluza T, Ndhlovu PD, Midzi N, Scott JT, Mutapi F, et al. (2003) Contrasting cellular responses in Schistosoma haematobium infected and exposed individuals from areas of high and low transmission in Zimbabwe. Immunol Lett 88: 249–256.
  11. 11. Al-Sherbiny M, Osman A, Barakat R, El Morshedy H, Bergquist R, et al. (2003) In vitro cellular and humoral responses to Schistosoma mansoni vaccine candidate antigens. Acta Trop 88: 117–130.
  12. 12. Wynn TA, Hoffmann KF (2000) Defining a schistosomiasis vaccination strategy - is it really Th1 versus Th2? Parasitol Today 16: 497–501.
  13. 13. Capron A, Dombrowicz D, Capron M (2004) Helminth infections and allergic diseases: from the Th2 paradigm to regulatory networks. Clin Rev Allergy Immunol 26: 25–34.
  14. 14. McKee AS, Pearce EJ (2004) CD25+CD4+ cells contribute to Th2 polarization during helminth infection by suppressing Th1 response development. J Immunol 173: 1224–1231.
  15. 15. Taylor MD, LeGoff L, Harris A, Malone E, Allen JE, et al. (2005) Removal of regulatory T cell activity reverses hyporesponsiveness and leads to filarial parasite clearance in vivo. J Immunol 174: 4924–4933.
  16. 16. Maizels RM, Yazdanbakhsh M (2003) Immune regulation by helminth parasites: cellular and molecular mechanisms. Nat Rev Immunol 3: 733–744.
  17. 17. Babu S, Blauvelt CP, Kumaraswami V, Nutman TB (2006) Regulatory networks induced by live parasites impair both Th1 and Th2 pathways in patent lymphatic filariasis: Implications for parasite persistence. J Immunol 176: 3248–3256.
  18. 18. Cavassani KA, Campanelli AP, Moreira AP, Vancim JO, Vitali LH, et al. (2006) Systemic and local characterization of regulatory T cells in a chronic fungal infection in humans. J Immunol 177: 5811–5818.
  19. 19. Faal N, Bailey RL, Jeffries D, Joof H, Sarr I, et al. (2006) Conjunctival FOXP3 expression in trachoma: do regulatory T cells have a role in human ocular Chlamydia trachomatis infection? PLoS Med 3: e266.
  20. 20. 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.
  21. 21. Guyot-Revol V, Innes JA, Hackforth S, Hinks T, Lalvani A (2006) Regulatory T cells are expanded in blood and disease sites in patients with tuberculosis. Am J Respir Crit Care Med 173: 803–810.
  22. 22. Belkaid Y, Tarbell K (2009) Regulatory T Cells in the Control of Host-Microorganism Interactions. Annu Rev Immunol 27: 551–589.
  23. 23. Nishikawa H, Sakaguchi S (2010) Regulatory T cells in tumor immunity. Int J Cancer 127: 759–767.
  24. 24. Ozdemir C, Akdis M, Akdis CA (2009) T regulatory cells and their counterparts: masters of immune regulation. Clin Exp Allergy 39: 626–639.
  25. 25. Suvas S, Azkur AK, Kim BS, Kumaraguru U, Rouse BT (2004) CD4+CD25+ regulatory T cells control the severity of viral immunoinflammatory lesions. J Immunol 172: 4123–4132.
  26. 26. McKinley L, Logar AJ, McAllister F, Zheng M, Steele C, et al. (2006) Regulatory T cells dampen pulmonary inflammation and lung injury in an animal model of pneumocystis pneumonia. J Immunol 177: 6215–6226.
  27. 27. Taylor JJ, Mohrs M, Pearce EJ (2006) Regulatory T cell responses develop in parallel to Th responses and control the magnitude and phenotype of the Th effector population. J Immunol 176: 5839–5847.
  28. 28. Fulton RB, Meyerholz DK, Varga SM (2010) Foxp3+ CD4 regulatory T cells limit pulmonary immunopathology by modulating the CD8 T cell response during respiratory syncytial virus infection. J Immunol 185: 2382–2392.
  29. 29. McSorley HJ, Harcus YM, Murray J, Taylor MD, Maizels RM (2008) Expansion of Foxp3(+) regulatory T cells in mice infected with the filarial parasite Brugia malayi. J Immunol 181: 6456–6466.
  30. 30. Rausch S, Huehn J, Mrchhoff D, Rzepecka J, Schnoeller C, et al. (2008) Functional analysis of effector and regulatory T cells in a parasitic nematode infection. Infect Immun 76: 1908–1919.
  31. 31. Layland LE, Mages J, Loddenkemper C, Hoerauf A, Wagner H, et al. (2010) Pronounced phenotype in activated regulatory T cells during a chronic helminth infection. J Immunol 184: 713–724.
  32. 32. Watanabe K, Carter JM, Neely-Burnam M, Colley DG (2009) Relative imbalance between T regulatory cells and activated T cells in mice with differential morbidity in chronic Schistosoma mansoni infections. Parasite Immunol 31: 440–446.
  33. 33. Baumgart M, Tompkins F, Leng J, Hesse M (2006) Naturally occurring CD4+Foxp3+ regulatory T cells are an essential, IL-10-independent part of the immunoregulatory network in Schistosoma mansoni egg-induced inflammation. J Immunol 176: 5374–5387.
  34. 34. Layland LE, Rad R, Wagner H, Da Costa CUP (2007) Immunopathology in schistosomiasis is controlled by antigen-specific regulatory T cells primed in the presence of TLR2. Eur J Immunol 37: 2174–2184.
  35. 35. Baecher-Allan C, Brown JA, Freeman GJ, Hafler DA (2001) CD4+CD25high regulatory cells in human peripheral blood. J Immunol 167: 1245–1253.
  36. 36. Watanabe K, Mwinzi PN, Black CL, Muok EM, Karanja DM, et al. (2007) T regulatory cell levels decrease in people infected with Schistosoma mansoni on effective treatment. Am J Trop Med Hyg 77: 676–682.
  37. 37. Muok EM, Mwinzi PN, Black CL, Carter JM, Ng'ang'a ZW, et al. (2009) Short report: Childhood coinfections with Plasmodium falciparum and Schistosoma mansoni result in lower percentages of activated T cells and T regulatory memory cells than schistosomiasis only. Am J Trop Med Hyg 80: 475–478.
  38. 38. Fontenot JD, Gavin MA, Rudensky AY (2003) Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol 4: 330–336.
  39. 39. Hori S, Nomura T, Sakaguchi S (2003) Control of regulatory T cell development by the transcription factor Foxp3. Science 299: 1057–1061.
  40. 40. Yagi H, Nomura T, Nakamura K, Yamazaki S, Kitawaki T, et al. (2004) Crucial role of FOXP3 in the development and function of human CD25+CD4+ regulatory T cells. Int Immunol 16: 1643–1656.
  41. 41. Miyara M, Yoshioka Y, Kitoh A, Shima T, Wing K, et al. (2009) Functional Delineation and Differentiation Dynamics of Human CD4(+) T Cells Expressing the FoxP3 Transcription Factor. Immunity 30: 899–911.
  42. 42. Wang J, Ioan-Facsinay A, van der Voort Ellen IH, Huizinga Tom WJ, Toes René EM (2007) Transient expression of FOXP3 in human activated nonregulatory CD4+ T cells. Eur J Immunol 37: 129–138.
  43. 43. Kryczek I, Liu R, Wang G, Wu K, Shu X, et al. (2009) FOXP3 Defines Regulatory T Cells in Human Tumor and Autoimmune Disease. Cancer Res 69: 3995–4000.
  44. 44. Hartigan-O'Connor DJ, Poon C, Sinclair E, McCune JM (2007) Human CD4+regulatory T cells express lower levels of the IL-7 receptor alpha chain (CD127), allowing consistent identification and sorting of live cells. J Immunol Methods 319: 41–52.
  45. 45. Luhn K, Simmons CP, Moran E, Dung NT, Chau TN, et al. (2007) Increased frequencies of CD4+ CD25(high) regulatory T cells in acute dengue infection. J Exp Med 204: 979–985.
  46. 46. Mduluza M (2007) A Gateway to Biomedical Research in Africa. New York: Nova Science Publishers Inc.
  47. 47. Mutapi F, Mduluza T, Gomez-Escobar N, Gregory WF, Fernandez C, et al. (2006) Immuno-epidemiology of human Schistosoma haematobium infection: preferential IgG3 antibody responsiveness to a recombinant antigen dependent on age and parasite burden. BMC Infect Dis 6: 96.
  48. 48. Reilly LJ, Magkrioti C, Cavanagh DR, Mduluza T, Mutapi F (2008) Effect of treating Schistosoma haematobium infection on Plasmodium falciparum-specific antibody responses. BMC Infect Dis 8: 158.
  49. 49. Ndhlovu P, Chimbari M, Ndamba J, Chandiwana SK (1992) 1992 National Schistosomiasis Survey. Harare, Zimbabwe: Blair Research Laboratory.
  50. 50. Midzi N, Sangweme D, Zinyowera S, Mapingure MP, Brouwer KC, et al. (2008) The burden of polyparasitism among primary schoolchildren in rural and farming areas in Zimbabwe. Trans R Soc Trop Med Hyg 102: 1039–1045.
  51. 51. Mutapi F, Ndhlovu PD, Hagan P, Spicer JT, Mduluza T, et al. (1998) Chemotherapy accelerates the development of acquired immune responses to Schistosoma haematobium infection. J Infect Dis 178: 289–293.
  52. 52. Stothard JR, Mgeni AF, Khamis S, Seto E, Ramsan M, et al. (2002) Urinary schistosomiasis in schoolchildren on Zanzibar Island (Unguja), Tanzania: a parasitological survey supplemented with questionnaires. Trans R Soc Trop Med Hyg 96: 507–514.
  53. 53. Utzinger J, Booth M, N'Goran EK, Muller I, Tanner M, et al. (2001) Relative contribution of day-to-day and intra-specimen variation in faecal egg counts of Schistosoma mansoni before and after treatment with praziquantel. Parasitology 122: 537–544.
  54. 54. Mott KE (1983) A reusable polyamide filter for diagnosis of S. haematobium infection by urine filtration. Bull Soc Pathol Exot 76: 101–104.
  55. 55. Mevin MD, Brooke MM, editors. (1982) Laboratory procedures for the diagnosing of intestinal parasites. 3rd ed. Atlanata (GA): Centers for Disease Control and Prevention.
  56. 56. Black CL, Muok EM, Mwinzi PN, Carter JM, Karanja DM, et al. (2010) Increases in levels of schistosome-specific immunoglobulin E and CD23(+) B cells in a cohort of Kenyan children undergoing repeated treatment and reinfection with Schistosoma mansoni. J Infect Dis 202: 399–405.
  57. 57. Everts B, Adegnika AA, Kruize YC, Smits HH, Kremsner PG, et al. (2010) Functional impairment of human myeloid dendritic cells during Schistosoma haematobium infection. PLoS Negl Trop Dis 4: e667.
  58. 58. Sokal RR, Rohlf J (1995) Biometry: the principles and practice of statistics in biological research: Freeman and Company.
  59. 59. Mutapi F, Roddam A (2002) p-values for pathogens: statistical inference from infectious-disease data. Lancet Infectious Disease 2: 219–230.
  60. 60. Teixeira-Carvalho A, Martins-Filho OA, Peruhype-Magalhaes V, Silveira-Lemos D, Malaquias LCC, et al. (2008) Cytokines, chemokine receptors, CD4(+)CD25(HIGH+) T-cells and clinical forms of human schistosomiasis. Acta Tropica 108: 139–149.
  61. 61. Wammes LJ, Hamid F, Wiria AE, Gier Bd, Sartono E, et al. (2010) Regulatory T cells in human geohelminth infection suppress immune responses to BCG and Plasmodium falciparum. Eur J Immunol 40: 437–442.
  62. 62. Saule P, Trauet J, Dutriez V, Lekeux V, Dessaint JP, et al. (2006) Accumulation of memory T cells from childhood to old age: central and effector memory cells in CD4(+) versus effector memory and terminally differentiated memory cells in CD8(+) compartment. Mech Ageing Dev 127: 274–281.
  63. 63. Wilson MS, Mentink-Kane MM, Pesce JT, Ramalingam TR, Thompson R, et al. (2006) Immunopathology of schistosomiasis. Immunol Cell Biol 85: 148–154.