Skip to main content

The frequency of CD127low expressing CD4+CD25high T regulatory cells is inversely correlated with human T lymphotrophic virus type-1 (HTLV-1) proviral load in HTLV-1-infection and HTLV-1-associated myelopathy/tropical spastic paraparesis

Abstract

Background

CD4+CD25high regulatory T (TReg) cells modulate antigen-specific T cell responses, and can suppress anti-viral immunity. In HTLV-1 infection, a selective decrease in the function of TReg cell mediated HTLV-1-tax inhibition of FOXP3 expression has been described. The purpose of this study was to assess the frequency and phenotype of TReg cells in HTLV-1 asymptomatic carriers and in HTLV-1-associated neurological disease (HAM/TSP) patients, and to correlate with measures of T cell activation.

Results

We were able to confirm that HTLV-I drives activation, spontaneous IFNĪ³ production, and proliferation of CD4+ T cells. We also observed a significantly lower proportion of CTLA-4+ TReg cells (CD4+CD25high T cells) in subjects with HAM/TSP patients compared to healthy controls. Ki-67 expression was negatively correlated to the frequency of CTLA-4+ TReg cells in HAM/TSP only, although Ki-67 expression was inversely correlated with the percentage of CD127low TReg cells in healthy control subjects. Finally, the proportion of CD127low TReg cells correlated inversely with HTLV-1 proviral load.

Conclusion

Taken together, the results suggest that TReg cells may be subverted in HAM/TSP patients, which could explain the marked cellular activation, spontaneous cytokine production, and proliferation of CD4+ T cells, in particular those expressing the CD25highCD127low phenotype. TReg cells represent a potential target for therapeutic intervention for patients with HTLV-1-related neurological diseases.

Background

Between 10 and 20 million people are infected with HTLV-1 worldwide [1]. Although most subjects are clinically asymptomatic during their lifetime, a proportion (5 to 10%) develop adult T cell leukemia/lymphoma (ATLL) or HTLV-1 associated myelopathy/tropical spastic paraparesis (HAM/TSP) [2]. Epidemiological surveys have identified regions in the world where prevalence rates are considerably higher, including Japan, the Caribbean, South America, Africa, Melanesia and the Middle East [1, 3]. It has been estimated that the prevalence of HTLV-1 infection in South America ranges from 2 to 5% [4], with an estimated 1ā€“2 million infected people in Brazil [5]. The prevalence in blood donors ranges from 0.17 to 1.8% in different areas of the country [6, 7], with a 0.3% seroprevalence in the city of Sao Paulo blood donors [8].

HTVL-1 is a retrovirus encoding the group specific antigen (gag), protease (pro), polymerase (pol), and envelope (env) genes. Six proteins are encoded by the pX region of the genome, including the Tax protein, which is critical to viral replication and induction of cellular activation and transformation, increasing the expression and production of cytokines and receptors involved in T cell growth and transformation, such as IL-15 [9, 10] and IL-2 [11ā€“13]. Tax also has the ability of interfering in the expression of several transcription factors and proto-oncogenes, as well as in the nucleic acid repair and apoptosis [14ā€“17]. These effects combined seem to play a key role in the potential of HTLV-1 to induce cellular transformation and, consequently, trigger the development of ATLL.

It has been previously demonstrated that HTLV-1 proviral load is one of the key factors in the pathogenesis of HAM/TSP [18, 19], although host genetic factors are also independently associated with the development of the diseases, e.g. certain HLA [20, 21] and non-HLA [22, 23] genes. These invoke the hypothesis that both viral and genetic host factors are implicated in the pathogenesis of HAM/TSP.

The CD8+ T cell response to HTLV-1 can be readily detected [24ā€“31], commonly directed against the HTLV-1-tax protein. The contribution of the CD8+ T cell response might be particularly important for viral control in HTLV-1 infection, since infected lymphocytes produce virtually no cell-free infectious HTLV-1 particles. However, it is noteworthy that the magnitude of the HTLV-1-specific T cell response is associated with higher proviral loads, highlighting the fact that T cells frequencies are determined by proviral load, as well as being a determinant of proviral load. CD4+ T cells are the main target for HTLV-1 infection, which induces CD4+ T cell activation, including proliferation and IFNĪ³ production. The HTLV-1-specific CD4+ T cell response is directed mainly against Env, the HTLV-1 envelope surface [32].

TReg cells are crucial for the control of autoimmune disease and maintenance of peripheral T cell tolerance (reviewed in Sakagushi et al. [33]). In addition, they can suppress pathogen-specific T cell responses, including response to viruses [34ā€“37]. The mechanisms whereby TReg cells suppress T cell responses are not yet fully understood, but are likely to include both soluble factors, e.g. IL-10 and TGF-Ī², as well as cell-cell contact dependent mechanisms, e.g. through CTLA-4. CTLA-4 (CD152) is expressed by a large fraction of CD4+CD25+ T cells, and by a majority of CD4+CD25high T cells. CTLA-4 has also been shown to be one of mediators of TReg function [38, 39], and is considered a marker for TReg cells. In addition, it was recently demonstrated that TReg cells are characterized by low levels of the IL-7RĪ± (CD127low) [40ā€“42], which together with CD25 help to distinguish TReg cells from activated normal CD4+ T cells in healthy individuals. FOXP3 is a key regulator of TReg cell function, but is not exclusive to TReg cells; it has been identified in human nonregulatory activated CD4+FoxP3+ T cells. Humans with mutations in FOXP3 present with a syndrome characterized by severe autoimmune and inflammatory disorders often early in life, denominated IPEX [33]. Interestingly, it was recently shown that HTLV-1 tax can downregulate Foxp3 expression [43, 44].

We hypothesized that HTLV-1 compromises TReg cell function, resulting in higher T cell activation, which contributes to HAM/TSP development. We found a significantly higher frequency of CD4+Ki-67+ T cells and a lower proportion of CTLA-4+ TReg cells in subjects with HAM/TSP, compared to healthy controls. Moreover, we found an inverse correlation between HTLV-1 proviral load and frequency of CD127low/CTLA-4+TReg cells. Our data suggest a role for TReg cells in the pathogenesis of HAM/TSP, and reveal a potential new therapeutic target for patients with HAM/TSP.

Results

Study subjects

Blood samples were collected at the Federal University of Sao Paulo outpatient clinics, after informed consent. PBMC were isolated by Ficoll-Paque PLUS density gradient centrifugation and cryopreserved. The demographics of the study subjects are shown in Table 1, including gender, age, proviral load, CD3, CD4 and CD8 absolute T cell counts. No statistically significant differences were observed in gender and age distribution among groups.

Table 1 Characteristics of study subjects.

CD4+ T cell activation and IFNĪ³ production in healthy donors, HTLV-1 seropositive asymptomatics, and HAM/TSP patients

We initially investigated the expression of Ki-67, HLA-DR and CD38 on CD4+ T cells in PBMC from healthy donors (Control), HTLV-1 infected patients who were clinically asymptomatic (HTLV), or had associated neurological disease (HAM/TSP). HAM/TSP had significantly higher frequencies of CD4+Ki-67+ T cells compared to HTLV or Control subjects (Fig. 1A, 1C). In addition, HAM/TSP patients had an increase in the frequency of CD4+ HLA-DR+ T cells compared to Controls (Fig. 1E), whereas no statistically significant difference in frequency of CD4+CD38+ T cells was noted (data not shown). Furthermore, CD4+ T cells from both HTLV and HAM/TSP groups had an increase in the spontaneous expression of IFNĪ³ (Fig. 1B, 1D).

Figure 1
figure 1

Expression of Ki-67, HLA-DR and spontaneous production of IFNĪ³ by CD4+ T cells from Control, HTLV and HAM/TSP subjects. (A) Representative dot plots showing Ki-67 expression in CD4+ T cells from a Control, a HTLV, and a HAM/TSP subject. (B) Representative dot plots showing spontaneous IFNĪ³ production by CD4+ T cells. (C) Percent CD4+Ki-67+ T cells; and (D) percent CD4+IFNĪ³+ T cells; or (E) percent CD4+HLA-DR+ T cells from Control, HTLV, or HAM/TSP. The line shown represents the mean.

Decreased frequency of TReg cells in HAM/TSP patients

In order to assess the frequency of TReg cells in HTLV-1 infected subjects, we measured the expression of CD25, CTLA-4, CD127 and GITR on CD4+ T cells by flow cytometry. Gating strategies are shown in Fig. 2Aā€“C. The frequency of CD4+ T cells expressing CD25 was very similar between the groups (Fig. 2D). As CD25 is upregulated on activated CD4+ T cells, and thus is not a specific marker for TReg cells, we sought to determine the frequency of TReg cells using more specific phenotypes. First, we analyzed the frequency of CD4+CD25high T cells, known to be composed mainly of TReg cells [45], and no significant difference in the frequency of TReg cells could be found between controls and HTLV-1 subjects (Fig. 2E). We determined the intracellular expression of CTLA-4 in CD4+CD25+, and CD4+CD25high T cells and observed a decrease in frequency of CTLA-4+ from HAM/TSP patients (Fig. 2F and 2G). Next, we assessed the expression of CD127. HAM/TSP patients had a statistically significant increase in the frequency of CD4+CD25+CD127low T cells compared to Controls (Fig. 2H). In contrast, there was a slight decrease in frequency of CD4+CD25highCD127low T cells in HTLV-1 infected groups, although this did not reach statistical significance (Fig. 2I). There were no differences in percent expression of GITR between the groups (Fig. 2J, 2K).

Figure 2
figure 2

Immunophenotypes of CD25+ or CD25high CD4+ T cells in Control, HTLV, or HAM/TSP subjects. Dot plots depict the gating strategies for CD25+ and CD25high cells (A); CD127low (B); and CTLA-4 expression (C) are shown. The gates for CD25+ and CD4+CD25high T cells were set based on the expression level of CD25 on CD4- T cells, where the pattern of CD25 expression is more distinct. Accordingly, CD4+CD25high T cells were defined as those expressing CD25 at a level higher than the bulk CD4-CD25+ population. For D-K, individual results are represented in symbols, and the line represents the mean. (D) % CD4+CD25+T cells; (E) % CD4+CD25high T cells; (F) % CD4+CD25+CTLA-4+ T cells; (G) % of CD4+CD25highCTLA-4+ T cells; (H) % CD4+CD25+CD127low T cells; (I) % CD4+CD25highCD127low T cells; (J) % CD4+CD25highGITR+ T cells; and (K) % CD4+CD25highGITR+ T cells.

Decreased CTLA-4+ TReg cells correlate with increased CD4+ T cell proliferation in HAM/TSP patients

Our initial analysis indicated that the frequency of CD4+CD25+CD127low T cells was higher in HAM/TSP patients. However, there were a lower percentage of CTLA-4+ TReg cells, indicating that HAM/TSP subjects might have TReg cells with a dysfunctional phenotype. Moreover, we showed that HAM/TSP patients had an increased proliferation and T cell activation, as evidenced by higher frequencies of HLA-DR, Ki-67 and INFĪ³-expressing CD4+ T cells. To test whether the higher frequency of TReg cells was associated with lower levels of activation or proliferation of CD4+ T cells, we compared the frequency of CTLA-4+, CD127low or GITR+ TReg cells with the percentage of CD4+Ki-67+ T cells. The frequency of CTLA-4+ TReg was negatively correlated with the frequency of CD4+Ki-67+ T cells in HAM/TSP patients only (Fig. 3A). In addition, there was a negative correlation between the frequency of CD127low TReg cells and the percentage of CD4+Ki-67+ T cells in controls, whereas no such correlation was found in HTLV-1 infected subjects (Fig. 3B). There was no association between GITR+ TReg cells with the percentage of CD4+Ki-67+ T cells in any group (Fig. 3C).

Figure 3
figure 3

Correlation between different immunophenotypes of T reg with expression of Ki-67. Frequency of CTLA-4+, CD127low, or GITR+ Treg were correlated with proportion of CD4+Ki67+ T cells in Control (Blue diamond), HTLV (Green triangle) and HAM/TSP (Red circle). A. There was a negative correlation between CTLA-4+ Treg with percentage of CD4+Ki67+ T cells in HAM/TSP only (r = -0.7833, p = 0.017). B. CD127low Treg were inversely correlated with proportion of CD4+Ki67+ T cells in Control group (r = -0.8108, p = 0.034). C. There was no association between GITR+ Treg with percentage of CD4+Ki67+ T cells in any group.

The frequency of CD4+CD25+CTLA-4+ and CD127low TReg cells was negatively correlated to HTLV-1 proviral load

Finally, we wanted to determine whether the frequency of TReg cells was related to HTLV-1 proviral load. High CD4+ T cells activation and elevated HTLV-1 proviral load are observed in HAM/TSP. We hypothesized that this phenomenon would be related to a lower proportion of TReg cells. We quantified HTLV-1 proviral load by real-time PCR and correlated it with the frequency of the different CD25-expressing CD4+ T cell subsets. There was no correlation between the frequency of CD4+CD25+ or CD4+CD25high T cells with proviral load (Fig. 4A and 4B). In contrast, there was a negative correlation between the frequency of CD4+CD25+CTLA-4+ T cells and proviral load in HTLV only (Fig. 4C). Moreover, there was an inverse correlation between CD127low TReg cells and HTLV-1 proviral load (Fig. 4F).

Figure 4
figure 4

Correlation between HTLV-1 proviral load of HTLV (Green triangle), and HAM/TSP (Red circle) with expression of CD127 or CTLA-4 in CD25+ or CD25high CD4+ T cells subsets. HTLV-I proviral load is expressed in copies per 1,000 cells. There was no correlation with CD4+CD25+ T cells (A). No correlation with CD4+CD25high T cells (B). Inverse correlation with the percentage of CD4+CD25+CTLA-4+ T cells in HTLV only; r = -0.7500, p = 0.0255 (C). No correlation with CD4+CD25highCTLA-4+ T cells (D). Lack of correlation with the percentage of CD4+CD25+CD127low T cells (E). (F) Negative correlation with CD4+CD25highCD127low T cells in HTLV (r = -0.7333, p = 0.03) and HAM/TSP (r = -0.8108, p = 0.034).

Discussion

Regulatory T cells are important for the maintenance of peripheral T cell tolerance to self antigens, and can also suppress T cell responses to tumors, parasites, viruses and bacteria. In this study we addressed the relationship between TReg cells, T cell activation, and HTLV-1 proviral load. Infection with HTLV-1 was associated with higher spontaneous IFNĪ³ release by CD4+ T cells, but only in HAM/TSP there was a marked increase in T cell proliferation.

The HTLV-1 derived tax protein can downregulate expression of the FOXP3, which presence is associated with TReg cell function [43, 44]. Increased expression of tax can be expected in patients with HAM/TSP, who have higher proviral loads compared to asymptomatic carries. We observed a higher proportion of CD4+ IFNĪ³+ T cells in HTLV-1 infected subjects, which could also be indicative of a decreased TReg cell fraction. Interestingly, only the HAM/TSP patients presented with a higher cell proliferation, as measured by Ki-67 staining, which correlated markedly with HTLV-1 proviral load (data not shown). These observations suggest that HTLV-1 directly affects TReg cell number, and as proviral load increases, not only is the control of IFNĪ³ lost, but controls on cell proliferation as well. Our data, together with the recent findings that HTLV-1 tax downregulates FOXP3 expression, indicate that TReg cell dysfunction can be a direct consequence of HTLV-1 infection.

In order to better understand the role of TReg cells in HTLV-I infection and disease, we used CTLA-4 and CD127 staining in CD4+CD25high cells as markers for TReg subsets. CD127 and CTLA-4 have been described as useful markers for TReg, and facilitate the identification of TReg cells, even without staining for FOXP3 [40]. In this study, we found that an increased frequency of CD127lowCD4+CD25+ TReg in controls correlated negatively with CD4+ T cell proliferation (Ki-67), indicating that these cells indeed have a regulatory immunophenotype. In contrast, increased frequency of these cells correlated with increased CD4+ T cell proliferation in HTLV-1 infected individuals, suggesting that these cells are not regulatory T cells in these individuals. In addiction, the elevated frequency of CTLA-4+ TReg cells was negatively correlated to CD4+ T cell proliferation only in HAM/TSP patients, which suggest that it is a better immunophenotype of TReg cells in HAM/TSP patients, but more studies are necessary to confirm this.

We could detect a negative association between the frequency of CTLA-4+ or CD127low TReg cells and proviral load, extending recent findings of an association between FOXP3 expression and HTLV-1 infection [44]. We speculate that therapeutic manipulation of regulatory T cells could positively impact disease pathogenesis. Two mechanisms might be involved, the first by suppressing the exuberant anti-HTLV-1 CD8+ T cell mediated immune response, and the second by suppression of CD4+ T cell proliferation, which can result in lower proviral load. However, stimulating an expansion of TReg cells could also provide additional targets for HTLV-1 replication, so such studies should proceed with great caution.

In this study, there are some limitations. The study was cross sectional, and with a limited number of patients in each group. We hope that future longitudinal studies can assess changes in TReg cells over time in HTLV-1 infected patients. We, and others, working in the regulatory T cells field, are limited by the lack of definitive phenotypic markers of TReg, and CD4+CD25+/high remains the standard identifiers. In this study we have added other markers, but at the time the study was conducted, the FOXP3 antibody, commonly used to detect a TReg cell population was not commercially available. However, this may have been a fortuitous event, as recent reports suggest that FOXP3 is also expressed on non regulatory T cells in humans [46, 47]. As we did not have access to tissue samples from these subjects, we cannot exclude redistribution of cells out of the peripheral blood into tissues, and the study of regulatory T cells at secondary lymphoid sites and within CSF will be of interest for a future study of HTLV-1 associated disease.

In conclusion, our data suggest a role of TReg cells in the pathogenesis of HAM/TSP. Further studies should help delineate the ability of expanded TReg cells to affect T cell proliferation in HTLV-1 patients and the potential development of therapeutic modulation of regulatory T cells in HTLV-1 patients.

Conclusion

In this study, we showed that HTLV-I drives activation, spontaneous IFNĪ³ production, and proliferation of CD4+ T cells. HAM/TSP patients have a decreased frequency of TReg cells in peripheral blood, compared to healthy subjects, markedly in the CD4+CD25highCTLA+ phenotype. The proportion of CD127low TReg cells correlated inversely with HTLV-1 proviral load. These results suggest that TReg cells may be subverted in HAM/TSP patients, and contributes to the identification of novel therapeutic targets for patients with HTLV-1-related disease.

Methods

Study subjects

Three groups of volunteers were enrolled. The first consisted of seven HTLV-1-negative control volunteers; the second consisted of ten HTLV-1 seropositive volunteers without clinical and laboratory evidence of HTLV-1-associated disease, and the last group was composed of nine patients with the diagnosis of HTLV-1 associated myelopathy/tropical spastic paraparesis (HAM/TSP). After approval by the Institutional Review Board, written informed consent was obtained from all the participants according to the guidelines of Brazilian Ministry of Health. Samples were collected in EDTA-treated vacuum tubes, and PBMC were frozen into liquid nitrogen after separation using a ficoll gradient.

DNA extraction and determination of HTLV-1 proviral load

HTLV-1 proviral DNA was extracted from PBMCs using a commercial kit (Qiagen GmbH, Hilden Germany) following the manufacturer's instructions. The extracted DNA was used as a template to amplify a fragment of 158 bp from the viral tax region using previously published primers [48]. The SYBR green real-time PCR assay was carried out in 25 Ī¼l PCR mixture containing 10Ɨ Tris (pH 8.3; Invitrogen, Brazil), 1.5 mM MgCl2, 0.2 Ī¼M of each primer, 0.2 mM of each dNTPs, SYBR Green (18.75 Units/r Ɨ n; Cambrex Bio Science, Rockland, ME) and 1 unit of platinum Taq polymerase (Invitrogen, Brazil). The amplification was performed in the Bio-Rad iCycler iQ system using an initial denaturation step at 95Ā°C for 2 minutes, followed by 50 cycles of 95Ā°C for 30 seconds, 57Ā°C for 30 seconds and 72Ā°C for 30 seconds. The human housekeeping Ī² globin gene primers GH20 and PC04 [49] were used as an internal control calibrator. For each run, standard curves for the value of HTLV-1 tax were generated from MT-2 cells of log10 dilutions (from 105 to 100 copy). The threshold cycle for each clinical sample was calculated by defining the point at which the fluorescence exceeded a threshold limit. Each sample was assayed in duplicate and the mean of the two values was considered as the copy number of the sample. The amount of HTLV-1 proviral load was calculated as follows: copy number of HTLV-1 (tax) per 1,000 cells = (copy number of HTLV-1 tax)/(copy number of Ī² globin/2) Ɨ 1000 cells. The method could detect 1 copy per 103 PBMCs cells.

Flow cytometry

PBMCs were thawed and stained with directly conjugated antibodies. Three different panels of antibodies were used to evaluate the expression of proteins associated with TReg cells and T cell activation. All antibodies were from BD Biosciences, unless otherwise noted. All panels contained PerCP-conjugated anti-CD4 and allophycocyanin-conjugated anti-CD25, and in addition contained (1) FITC-conjugated anti-GITR and PE-conjugated anti-CD127 (Beckman Coulter, Miami, FL), (2) FITC-conjugated anti-CD45RA and PE-conjugated anti-HLA-DR and (3) FITC-conjugated anti-Ki-67 and PE-conjugated anti-CD152 (CTLA-4). Cells stained with PerCP-conjugated anti-CD4 alone and allophycocyanin-conjugated CD25 alone were used to establish positive gates for FITC- and PE-conjugated antibodies. For panel 1 and 2, cells were stained with all antibodies in PBS supplemented with 0.5% bovine serum albumin (BSA) and 2 mM EDTA (FACS buffer), followed by two washes in FACS buffer and fixation in 1% paraformaldehyde (PFA). For panel 3, cells were first stained with PerCP-conjugated anti-CD4 and allophycocyanin-conjugated anti-CD25, followed by two washes in FACS buffer and fixation in 1% PFA. The cells were subsequently washed twice with PBS containing 0.1% saponin (perm buffer), prior to staining with PE-conjugated anti-CD152 and FITC-conjugated anti-Ki-67 diluted in perm buffer. All samples were analyzed on a FACSCalibur flow cytometer (Becton Dickinson) equipped with a 488 nm argon and a 633 nm red-diode lasers for four color detection. Acquisition and analyses were performed using CellQuest software (Becton Dickinson). Fluorescence voltages and compensation values were determined using unstained cells and cells single-stained with each of the fluorochrome-conjugated antibodies, respectively. The gating strategy used was to gate on lymphocytes using a forward scatter versus side scatter gate, followed by gating on CD4+ cells. The gate for CD4+CD25+ cells was set using cells cells stained with the PerCP-conjugated anti-CD4 antibody alone. Positive gates for the FITC- and PE-conjugated antibodies were set using cells stained with only PerCP-conjugated anti-CD4 and APC-conjugated anti-CD25 antibodies.

Cytokine flow cytometry

PBMCs were thawed and cultured for 24 hours in 96-well U-bottom plates at a concentration of 4 Ɨ 105 cells/well. Brefeldin A (BFA) was added at a concentration of 5 Ī¼g/ml for the last 5 hours of the culture. After culture, cells were harvested, stained with PE-conjugated anti-CD4, fixed in 4% PFA for 20 min, prior to being washed twice with perm buffer. The cells were subsequently stained with PerCP-conjugated anti-CD3 and allophycocyanin-conjugated anti-IFNĪ³, washed twice in perm buffer and resuspended in FACS buffer, prior to being analyzed on a FACSCalibur. All antibodies were from BD Biosciences.

Statistical analyses

Data sets were compiled and analyzed in Statistica, release 6.0 (Statsoft, Tulsa, OK) and Prism, release 4.0 (GraphPad Software, San Diego, CA). Groups comparisons were performed using non-parametric Kruskal Wallis ANOVA by ranks test; associations between variables were evaluated by Spearman rank order correlation's test. Critical p values were considered statistically significant if below 0.05.

References

  1. Edlich RF, Hill LG, Williams FM: Global epidemic of human T-cell lymphotrophic virus type-I (HTLV-I): an update. J Long Term Eff Med Implants. 2003, 13 (2): 127-140. 10.1615/JLongTermEffMedImplants.v13.i2.70.

    ArticleĀ  PubMedĀ  Google ScholarĀ 

  2. Tajima K: The 4th nation-wide study of adult T-cell leukemia/lymphoma (ATL) in Japan: estimates of risk of ATL and its geographical and clinical features. The T- and B-cell Malignancy Study Group. Int J Cancer. 1990, 45 (2): 237-243. 10.1002/ijc.2910450206.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  3. Edlich RF, Arnette JA, Williams FM: Global epidemic of human T-cell lymphotropic virus type-I (HTLV-I). J Emerg Med. 2000, 18 (1): 109-119. 10.1016/S0736-4679(99)00173-0.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  4. Gotuzzo E, Arango C, de Queiroz-Campos A, Isturiz RE: Human T-cell lymphotropic virus-I in Latin America. Infect Dis Clin North Am. 2000, 14 (1): 211-39, x-xi. 10.1016/S0891-5520(05)70225-7.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  5. Carneiro-Proietti AB, Ribas JG, Catalan-Soares BC, Martins ML, Brito-Melo GE, Martins-Filho OA, Pinheiro SR, Araujo Ade Q, Galvao-Castro B, de Oliveira MS, Guedes AC, Proietti FA: [Infection and disease caused by the human T cell lymphotropic viruses type I and II in Brazil]. Rev Soc Bras Med Trop. 2002, 35 (5): 499-508. 10.1590/S0037-86822002000500013.

    ArticleĀ  PubMedĀ  Google ScholarĀ 

  6. Farias de Carvalho SM, Pombo de Oliveira MS, Thuler LC, Rios M, Coelho RC, Rubim LC, Silva EM, Reis AM, Catovsky D: HTLV-I and HTLV-II infections in hematologic disorder patients, cancer patients, and healthy individuals from Rio de Janeiro, Brazil. J Acquir Immune Defic Syndr Hum Retrovirol. 1997, 15 (3): 238-242.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  7. Galvao-Castro B, Loures L, Rodriques LG, Sereno A, Ferreira Junior OC, Franco LG, Muller M, Sampaio DA, Santana A, Passos LM, Proietti F: Distribution of human T-lymphotropic virus type I among blood donors: a nationwide Brazilian study. Transfusion. 1997, 37 (2): 242-243. 10.1046/j.1537-2995.1997.37297203532.x.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  8. Segurado AA, Malaque CM, Sumita LM, Pannuti CS, Lal RB: Laboratory characterization of human T cell lymphotropic virus types 1 (HTLV-1) and 2 (HTLV-2) infections in blood donors from Sao Paulo, Brazil. Am J Trop Med Hyg. 1997, 57 (2): 142-148.

    CASĀ  PubMedĀ  Google ScholarĀ 

  9. Mariner JM, Lantz V, Waldmann TA, Azimi N: Human T cell lymphotropic virus type I Tax activates IL-15R alpha gene expression through an NF-kappa B site. J Immunol. 2001, 166 (4): 2602-2609.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  10. Azimi N, Brown K, Bamford RN, Tagaya Y, Siebenlist U, Waldmann TA: Human T cell lymphotropic virus type I Tax protein trans-activates interleukin 15 gene transcription through an NF-kappaB site. Proc Natl Acad Sci U S A. 1998, 95 (5): 2452-2457. 10.1073/pnas.95.5.2452.

    ArticleĀ  PubMed CentralĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  11. Ballard DW, Bohnlein E, Lowenthal JW, Wano Y, Franza BR, Greene WC: HTLV-I tax induces cellular proteins that activate the kappa B element in the IL-2 receptor alpha gene. Science. 1988, 241 (4873): 1652-1655. 10.1126/science.2843985.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  12. Crenon I, Beraud C, Simard P, Montagne J, Veschambre P, Jalinot P: The transcriptionally active factors mediating the effect of the HTLV-I Tax transactivator on the IL-2R alpha kappa B enhancer include the product of the c-rel proto-oncogene. Oncogene. 1993, 8 (4): 867-875.

    CASĀ  PubMedĀ  Google ScholarĀ 

  13. Good L, Maggirwar SB, Sun SC: Activation of the IL-2 gene promoter by HTLV-I tax involves induction of NF-AT complexes bound to the CD28-responsive element. Embo J. 1996, 15 (14): 3744-3750.

    PubMed CentralĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  14. Kao SY, Marriott SJ: Disruption of nucleotide excision repair by the human T-cell leukemia virus type 1 Tax protein. J Virol. 1999, 73 (5): 4299-4304.

    PubMed CentralĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  15. Laherty CD, Perkins ND, Dixit VM: Human T cell leukemia virus type I Tax and phorbol 12-myristate 13-acetate induce expression of the A20 zinc finger protein by distinct mechanisms involving nuclear factor kappa B. J Biol Chem. 1993, 268 (7): 5032-5039.

    CASĀ  PubMedĀ  Google ScholarĀ 

  16. Brauweiler A, Garrus JE, Reed JC, Nyborg JK: Repression of bax gene expression by the HTLV-1 Tax protein: implications for suppression of apoptosis in virally infected cells. Virology. 1997, 231 (1): 135-140. 10.1006/viro.1997.8509.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  17. Uittenbogaard MN, Giebler HA, Reisman D, Nyborg JK: Transcriptional repression of p53 by human T-cell leukemia virus type I Tax protein. J Biol Chem. 1995, 270 (48): 28503-28506. 10.1074/jbc.270.48.28503.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  18. Nagai M, Usuku K, Matsumoto W, Kodama D, Takenouchi N, Moritoyo T, Hashiguchi S, Ichinose M, Bangham CR, Izumo S, Osame M: Analysis of HTLV-I proviral load in 202 HAM/TSP patients and 243 asymptomatic HTLV-I carriers: high proviral load strongly predisposes to HAM/TSP. J Neurovirol. 1998, 4 (6): 586-593. 10.3109/13550289809114225.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  19. Yoshida M, Miyoshi I, Hinuma Y: Isolation and characterization of retrovirus from cell lines of human adult T-cell leukemia and its implication in the disease. Proc Natl Acad Sci U S A. 1982, 79 (6): 2031-2035. 10.1073/pnas.79.6.2031.

    ArticleĀ  PubMed CentralĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  20. Jeffery KJ, Siddiqui AA, Bunce M, Lloyd AL, Vine AM, Witkover AD, Izumo S, Usuku K, Welsh KI, Osame M, Bangham CR: The influence of HLA class I alleles and heterozygosity on the outcome of human T cell lymphotropic virus type I infection. J Immunol. 2000, 165 (12): 7278-7284.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  21. Jeffery KJ, Usuku K, Hall SE, Matsumoto W, Taylor GP, Procter J, Bunce M, Ogg GS, Welsh KI, Weber JN, Lloyd AL, Nowak MA, Nagai M, Kodama D, Izumo S, Osame M, Bangham CR: HLA alleles determine human T-lymphotropic virus-I (HTLV-I) proviral load and the risk of HTLV-I-associated myelopathy. Proc Natl Acad Sci U S A. 1999, 96 (7): 3848-3853. 10.1073/pnas.96.7.3848.

    ArticleĀ  PubMed CentralĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  22. Sabouri AH, Saito M, Lloyd AL, Vine AM, Witkover AW, Furukawa Y, Izumo S, Arimura K, Marshall SE, Usuku K, Bangham CR, Osame M: Polymorphism in the interleukin-10 promoter affects both provirus load and the risk of human T lymphotropic virus type I-associated myelopathy/tropical spastic paraparesis. J Infect Dis. 2004, 190 (7): 1279-1285. 10.1086/423942.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  23. Vine AM, Witkover AD, Lloyd AL, Jeffery KJ, Siddiqui A, Marshall SE, Bunce M, Eiraku N, Izumo S, Usuku K, Osame M, Bangham CR: Polygenic control of human T lymphotropic virus type I (HTLV-I) provirus load and the risk of HTLV-I-associated myelopathy/tropical spastic paraparesis. J Infect Dis. 2002, 186 (7): 932-939. 10.1086/342953.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  24. Lim DG, Bieganowska Bourcier K, Freeman GJ, Hafler DA: Examination of CD8+ T cell function in humans using MHC class I tetramers: similar cytotoxicity but variable proliferation and cytokine production among different clonal CD8+ T cells specific to a single viral epitope. J Immunol. 2000, 165 (11): 6214-6220.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  25. Tanaka Y, Tozawa H, Koyanagi Y, Shida H: Recognition of human T cell leukemia virus type I (HTLV-I) gag and pX gene products by MHC-restricted cytotoxic T lymphocytes induced in rats against syngeneic HTLV-I-infected cells. J Immunol. 1990, 144 (11): 4202-4211.

    CASĀ  PubMedĀ  Google ScholarĀ 

  26. Jacobson S, Reuben JS, Streilein RD, Palker TJ: Induction of CD4+, human T lymphotropic virus type-1-specific cytotoxic T lymphocytes from patients with HAM/TSP. Recognition of an immunogenic region of the gp46 envelope glycoprotein of human T lymphotropic virus type-1. J Immunol. 1991, 146 (4): 1155-1162.

    CASĀ  PubMedĀ  Google ScholarĀ 

  27. Kannagi M, Shida H, Igarashi H, Kuruma K, Murai H, Aono Y, Maruyama I, Osame M, Hattori T, Inoko H, et al.: Target epitope in the Tax protein of human T-cell leukemia virus type I recognized by class I major histocompatibility complex-restricted cytotoxic T cells. J Virol. 1992, 66 (5): 2928-2933.

    PubMed CentralĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  28. Daenke S, Kermode AG, Hall SE, Taylor G, Weber J, Nightingale S, Bangham CR: High activated and memory cytotoxic T-cell responses to HTLV-1 in healthy carriers and patients with tropical spastic paraparesis. Virology. 1996, 217 (1): 139-146. 10.1006/viro.1996.0101.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  29. Kozako T, Arima N, Toji S, Masamoto I, Akimoto M, Hamada H, Che XF, Fujiwara H, Matsushita K, Tokunaga M, Haraguchi K, Uozumi K, Suzuki S, Takezaki T, Sonoda S: Reduced frequency, diversity, and function of human T cell leukemia virus type 1-specific CD8+ T cell in adult T cell leukemia patients. J Immunol. 2006, 177 (8): 5718-5726.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  30. Furukawa K, Mori M, Ohta N, Ikeda H, Shida H, Furukawa K, Shiku H: Clonal expansion of CD8+ cytotoxic T lymphocytes against human T cell lymphotropic virus type I (HTLV-I) genome products in HTLV-I-associated myelopathy/tropical spastic paraparesis patients. J Clin Invest. 1994, 94 (5): 1830-1839. 10.1172/JCI117532.

    ArticleĀ  PubMed CentralĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  31. Pique C, Connan F, Levilain JP, Choppin J, Dokhelar MC: Among all human T-cell leukemia virus type 1 proteins, tax, polymerase, and envelope proteins are predicted as preferential targets for the HLA-A2-restricted cytotoxic T-cell response. J Virol. 1996, 70 (8): 4919-4926.

    PubMed CentralĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  32. Goon PK, Hanon E, Igakura T, Tanaka Y, Weber JN, Taylor GP, Bangham CR: High frequencies of Th1-type CD4(+) T cells specific to HTLV-1 Env and Tax proteins in patients with HTLV-1-associated myelopathy/tropical spastic paraparesis. Blood. 2002, 99 (9): 3335-3341. 10.1182/blood.V99.9.3335.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  33. Sakaguchi S, Ono M, Setoguchi R, Yagi H, Hori S, Fehervari Z, Shimizu J, Takahashi T, Nomura T: Foxp3+ CD25+ CD4+ natural regulatory T cells in dominant self-tolerance and autoimmune disease. Immunol Rev. 2006, 212: 8-27. 10.1111/j.0105-2896.2006.00427.x.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  34. Nixon DF, Aandahl EM, Michaelsson J: CD4+CD25+ regulatory T cells in HIV infection. Microbes Infect. 2005, 7 (7-8): 1063-1065. 10.1016/j.micinf.2005.03.026.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  35. Dittmer U, He H, Messer RJ, Schimmer S, Olbrich AR, Ohlen C, Greenberg PD, Stromnes IM, Iwashiro M, Sakaguchi S, Evans LH, Peterson KE, Yang G, Hasenkrug KJ: Functional impairment of CD8(+) T cells by regulatory T cells during persistent retroviral infection. Immunity. 2004, 20 (3): 293-303. 10.1016/S1074-7613(04)00054-8.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  36. Suvas S, Kumaraguru U, Pack CD, Lee S, Rouse BT: CD4+CD25+ T cells regulate virus-specific primary and memory CD8+ T cell responses. J Exp Med. 2003, 198 (6): 889-901. 10.1084/jem.20030171.

    ArticleĀ  PubMed CentralĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  37. Ward SM, Fox BC, Brown PJ, Worthington J, Fox SB, Chapman RW, Fleming KA, Banham AH, Klenerman P: Quantification and localisation of FOXP3+ T lymphocytes and relation to hepatic inflammation during chronic HCV infection. J Hepatol. 2007

    Google ScholarĀ 

  38. Read S, Malmstrom V, Powrie F: Cytotoxic T lymphocyte-associated antigen 4 plays an essential role in the function of CD25(+)CD4(+) regulatory cells that control intestinal inflammation. J Exp Med. 2000, 192 (2): 295-302. 10.1084/jem.192.2.295.

    ArticleĀ  PubMed CentralĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  39. Takahashi T, Tagami T, Yamazaki S, Uede T, Shimizu J, Sakaguchi N, Mak TW, Sakaguchi S: Immunologic self-tolerance maintained by CD25(+)CD4(+) regulatory T cells constitutively expressing cytotoxic T lymphocyte-associated antigen 4. J Exp Med. 2000, 192 (2): 303-310. 10.1084/jem.192.2.303.

    ArticleĀ  PubMed CentralĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  40. Liu W, Putnam AL, Xu-Yu Z, Szot GL, Lee MR, Zhu S, Gottlieb PA, Kapranov P, Gingeras TR, Fazekas de St Groth B, Clayberger C, Soper DM, Ziegler SF, Bluestone JA: CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4+ T reg cells. J Exp Med. 2006, 203 (7): 1701-1711. 10.1084/jem.20060772.

    ArticleĀ  PubMed CentralĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  41. Seddiki N, Santner-Nanan B, Martinson J, Zaunders J, Sasson S, Landay A, Solomon M, Selby W, Alexander SI, Nanan R, Kelleher A, Fazekas de St Groth B: Expression of interleukin (IL)-2 and IL-7 receptors discriminates between human regulatory and activated T cells. J Exp Med. 2006, 203 (7): 1693-1700. 10.1084/jem.20060468.

    ArticleĀ  PubMed CentralĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  42. Michaelsson J, Mold JE, McCune JM, Nixon DF: Regulation of T cell responses in the developing human fetus. J Immunol. 2006, 176 (10): 5741-5748.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  43. Yamano Y, Takenouchi N, Li HC, Tomaru U, Yao K, Grant CW, Maric DA, Jacobson S: Virus-induced dysfunction of CD4+CD25+ T cells in patients with HTLV-I-associated neuroimmunological disease. J Clin Invest. 2005, 115 (5): 1361-1368.

    ArticleĀ  PubMed CentralĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  44. Oh U, Grant C, Griffith C, Fugo K, Takenouchi N, Jacobson S: Reduced Foxp3 protein expression is associated with inflammatory disease during human t lymphotropic virus type 1 Infection. J Infect Dis. 2006, 193 (11): 1557-1566. 10.1086/503874.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  45. Baecher-Allan C, Brown JA, Freeman GJ, Hafler DA: CD4+CD25high regulatory cells in human peripheral blood. J Immunol. 2001, 167 (3): 1245-1253.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  46. Allan SE, Crome SQ, Crellin NK, Passerini L, Steiner TS, Bacchetta R, Roncarolo MG, Levings MK: Activation-induced FOXP3 in human T effector cells does not suppress proliferation or cytokine production. Int Immunol. 2007, 19 (4): 345-354. 10.1093/intimm/dxm014.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  47. Wang J, Ioan-Facsinay A, van der Voort EI, Huizinga TW, Toes RE: Transient expression of FOXP3 in human activated nonregulatory CD4+ T cells. Eur J Immunol. 2007, 37 (1): 129-138. 10.1002/eji.200636435.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  48. Innis MA: PCR protocols : a guide to methods and applications. 1990, San Diego , Academic Press, xviii, 482 p.-

    Google ScholarĀ 

  49. Iannone R, Sherman MP, Rodgers-Johnson PE, Beilke MA, Mora CA, Amin RM, Tinsley SR, Papsidero LD, Poiesz BJ, Gibbs CJ: HTLV-I DNA sequences in CNS tissue of a patient with tropical spastic paraparesis and HTLV-I-associated myelopathy. J Acquir Immune Defic Syndr. 1992, 5 (8): 810-816.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

Download references

Acknowledgements

We would like to thank Marli Campos, Rosemeire Gabriel, and PerpĆ©tua for the assistance in blood collection, and Dr. Lishomwa Ndhlovu for helpful scientific discussions. We are also grateful to Karina I. Carvalho, Helena Tomiyama, and Raphaella Goulart for the continuous laboratory support. This work was supported by the FundaĆ§Ć£o de Amparo a Pesquisa do Estado de SĆ£o Paulo (FAPESP), grant #04/10918-6, NIH grants (AI41531, AI52731, AI060379, AI064520), and The Fogarty International Center, grant #D43 TW00003. Dr. Barbosa and Ms. Brunialti were supported by the CoordenaĆ§Ć£o de AperfeiƧoamento de Pessoal de NĆ­vel Superior (CAPES), Brazilian Ministry of Education.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Esper G Kallas.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

JM planed and conducted the experiments, performed the statistical analyses, and wrote the manuscript; HMRB wrote the original protocol, collected the samples, prepared the database, helped in the experiments, performed statistical analyses, and wrote the manuscript; KAJ conducted the experiments and helped in the flow cytometry analyses; JMC conducted the experiments and helped in the flow cytometry analyses; MKCB conducted the experiments and helped in the flow cytometry analyses; WKN performed the HTLV-I viral load; YN supported the protocol design and the conduction of HTLV-I viral load; ECS performed the HTLV-I serology, helped in the project design, the molecular biology experiments, and writing the manuscript; MAC selected subjects for the cohort and supervised the blood collection of samples from HAM/TSP patients; ASBO selected subjects for the cohort and supervised the blood collection of samples from HAM/TSP patients; DFN participated in the experimental design, obtained funds for the project, discussed the results, and wrote the manuscript; EGK wrote the original protocol, participated in the experimental design, obtained funds for the project, discussed the results, and wrote the manuscript.

Jakob Michaƫlsson, Hugo Marcelo R Barbosa contributed equally to this work.

Authorsā€™ original submitted files for images

Rights and permissions

Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://creativecommons.org/licenses/by/2.0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Michaƫlsson, J., Barbosa, H.M.R., Jordan, K.A. et al. The frequency of CD127low expressing CD4+CD25high T regulatory cells is inversely correlated with human T lymphotrophic virus type-1 (HTLV-1) proviral load in HTLV-1-infection and HTLV-1-associated myelopathy/tropical spastic paraparesis. BMC Immunol 9, 41 (2008). https://0-doi-org.brum.beds.ac.uk/10.1186/1471-2172-9-41

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/1471-2172-9-41

Keywords