Skip to main content
Advertisement
  • Loading metrics

Increase of CD4+CD25highFoxP3+ cells impairs in vitro human microbicidal activity against Mycobacterium tuberculosis during latent and acute pulmonary tuberculosis

Abstract

Background

Regulatory T cells (Tregs) play a critical role during Mycobacterium tuberculosis (Mtb) infection, modulating host responses while neutralizing excessive inflammation. However, their impact on regulating host protective immunity is not completely understood. Here, we demonstrate that Treg cells abrogate the in vitro microbicidal activity against Mtb.

Methods

We evaluated the in vitro microbicidal activity of peripheral blood mononuclear cells (PBMCs) from patients with active tuberculosis (TB), individuals with latent tuberculosis infection (LTBI, TST+/IGRA+) and healthy control (HC, TST-/IGRA-) volunteers. PBMCs, depleted or not of CD4+CD25+ T-cells, were analyzed to determine frequency and influence on microbicidal activity during in vitro Mtb infection with four clinical isolates (S1, S5, R3, and R6) and one reference strain (H37Rv).

Results

The frequency of CD4+CD25highFoxP3+ cells were significantly higher in Mtb infected whole blood cultures from both TB patients and LTBI individuals when compared to HC. Data from CD4+CD25+ T-cells depletion demonstrate that increase of CD4+CD25highFoxP3+ is associated with an impairment of Th-1 responses and a diminished in vitro microbicidal activity of LTBI and TB groups.

Conclusions

Tregs restrict host anti-mycobacterial immunity during active disease and latent infection and thereby may contribute to both disease progression and pathogen persistence.

Author summary

Our immune system has an enormous capacity of recognizing and responding to foreign antigens and, likewise, presents an extremely efficient mechanism of controlling these responses. Here, we investigated how a specific cell type with regulatory abilities can interfere in the immunological response against tuberculosis bacillus. For this, we used blood samples from individuals sensitized with the bacillus and patients with active pulmonary tuberculosis to understand how these cells act and their impact on the host/parasite relationship in the development of the disease. We could observe the negative impact that such regulatory cells cause during the immune response against Mycobacterium tuberculosis, decreasing the control/elimination of the bacillus in asymptomatic individuals and patients with tuberculosis. We also observed a recovery in the immune response when Treg cells were removed during in vitro challenge, restoring the capacity of Mtb clearance. Thus, these regulatory cells, when present, may represent a possible facilitator of the asymptomatic permanence of the bacillus, or even of the development of the disease itself. These data allowed us to see latency and tuberculosis from a new angle and thus postulate new approaches to fight tuberculosis.

Introduction

Although it has been estimated that one-fourth of the world’s population is latently infected with Mycobacterium tuberculosis (Mtb), 90% of these individuals may never develop active disease, suggesting that, in most cases, the immune response can control the infection [1,2]. However, despite its ability to control the infection, host immunity is not capable of eliminating Mtb bacilli and cumulative data suggest that T regulatory (Treg) cells in tuberculosis (TB) may play a negative role in Mtb clearance [3,4]. Human Treg cells represent 5–10% of CD4+ T cell population and can be characterized by the expression of Forkhead box P3 transcription factor (FoxP3) [5]. These cells are known to down-regulate the activity of CD4+ and CD8+ T-cell populations, avoiding collateral-tissue damage due to an excessive inflammation elicited during the response to pathogens [6,7]. Although this mechanism is generally beneficial to the host in the acute phase, it may be detrimental during chronic infection, when, despite the presence of active immune response, pathogen persistence is sustained, as reported during TB [8].

Tuberculosis patients present elevated frequencies of induced Treg cells when compared to healthy individuals [3,914]. Higher level of Tregs, as well as activated CD4+ T cells, was also determined in lymph nodes (LN) mononuclear cells of patients with LN TB when compared to PBMCs [15]. Moreover, in TB patients, Treg cells have a pivotal role in depressing anti-Mtb immunity, decreasing IFN-γ production, which is restored after Treg depletion [13,16,17]. Interestingly, the high frequency of Treg population is not restricted to active TB disease, but also reported during latent TB infection (LTBI) when compared to healthy controls (HC) both in PBMC [14,18,19] or in bronchoalveolar lavage [19].

It has been suggested that patients who had been treated and cured for TB present a higher risk of developing pulmonary TB if re-infected [20]. Besides, TB-case contacts with a prior positive TST response had an increased propensity to develop the active disease when compared to individuals with a negative TST result [21,22]. Recently our group showed that TST-positive individuals had an impaired in vitro microbicidal activity compared to TST-negative [23]. Together these findings indicate that immune-competent individuals infected with Mtb are not protected against disease during subsequent reinfection [2426].

In the present work, we investigated the ability of CD4+CD25highFoxP3+ cells to down-modulate in vitro microbicidal activity against Mtb. Data presented here demonstrate that Treg cells hinder anti-Mtb in vitro microbicidal activity, suggesting that these cells may inhibit the establishment of protective immunity against Mtb, contributing to pathogen persistence and/or disease progression in LTBI individuals and patients with active TB.

Materials and methods

Ethics statement

All study participants signed an informed consent form before their enrolment and before sample collection. The study protocol was approved by the Health Science Center Internal Review Board of the Universidade Federal do Espírito Santo and is registered under the number CEP/UFES 080/2011.

Human subjects

HIV-negative healthy volunteers were invited to participate in the present study at Universidade Federal do Espírito Santo (UFES), Vitória, Brazil, and divided according to their purified protein derivative (TST) and whole blood IFN-γ release assay (IGRA) status in LTBI (TST+/IGRA+) and healthy control (TST-/IGRA-) groups. Both LTBI and HC subjects underwent a comprehensive clinical evaluation before their enrollment, which included a complete hemogram, HIV test, thoracic X-ray, TST, and IGRA testing. Candidates invited to participate in the present study were excluded if positive for other chronic diseases such as HIV, intestinal helminths, hepatitis, and autoimmune diseases. HIV-negative active pulmonary TB patients were enrolled at the Tuberculosis Clinic, Hospital Universitário Cassiano Antônio de Moraes, Centro de Ciências da Saúde, Universidade Federal do Espírito Santo. TB diagnosis was based on chest X-ray, acid-fast bacilli staining of sputum smears (AFB smears), and mycobacterial culture of sputum samples from all TB patients. Blood samples from TB patients were collected before anti-TB therapy was initiated. Enrolled subjects/patients, who initiated anti-TB treatment, or the use of anti-inflammatory drugs were also excluded.

Tuberculin skin test (TST)

TST involved an intradermal injection of 0.1mL (2 tuberculin units) of the purified protein derivative RT23 (Statens Serum Institut, Copenhagen, Denmark) into the anterior surface of the forearm with a standard tuberculin syringe. Reactions were evaluated 72h after the injection by two experts and defined as positive by induration of ≥10 mm in the transverse diameter using the “Sokal ballpoint pen method” [27].

QuantiFERON TB-Gold In-Tube (QFT-IT)

Prior to the intradermal injection of PPD (tuberculin skin testing), blood samples for IGRA testing were collected by venipuncture using vials provided by the manufacturer. Blood samples were used for whole blood IFN-γ release assay (QuantiFERON TB Gold In-Tube, Qiagen, Valencia, CA, USA) according to the manufacturer’s instructions.

Mycobacterium tuberculosis reference strain and clinical isolates

The ATCC Mtb H37Rv reference strain (ATCC27294) and four clinical isolates with distinct minimal inhibitory concentration (MIC) to first-line anti-Mtb drugs and IS6110 RFLP electrophoretic band pattern [28], stored at the Núcleo de Doenças Infecciosas (NDI)/UFES repository, were used in the present study (S1 Fig and Table 1).

thumbnail
Table 1. Minimal inhibitory concentration (MIC) of Mycobacterium tuberculosis clinical isolates used in the present study.

https://doi.org/10.1371/journal.pntd.0009605.t001

Cell cultures and in vitro microbicidal activity assay (Mtb killing assay)

Peripheral blood samples were collected by venipuncture in K3-EDTA-treated tubes (BD Vacutainer Blood Collection Tube, Becton & Dickinson, USA). Peripheral blood mononuclear cells (PBMC) were isolated by density gradient separation (Ficoll-Histopaque, SIGMA-ALDRICH, Missouri, USA). The in vitro microbicidal activity assay, a modified version of an ex-vivo model previously described [29], was performed using 300 μL of heparinized peripheral whole blood or PBMC, depleted or not of Treg cells, dispensed into sterile microtubes (Sarstedt AG & Co, Nümbrecht, Germany) and incubated at 37°C, 5% CO2 in the presence of Mtb suspension (10:1 MOI) for 72h. Non-stimulated control samples received complete RPMI medium alone. After incubation, cultures were centrifuged (15000 x g for 10 min) and the supernatants were collected and stored at -80°C until further use. The cell pellet was lysed in sterile Milli-Q water, followed by serial dilution (10−1 to 10−4) in PBS 0.25% Tween 80. Aliquots were plated on Middlebrook 7H11 medium (BD Difco, Detroit, Michigan, USA) supplemented with Middlebrook OADC (BD BBL, Maryland, USA) and incubated for 14–21 days at 37°C in 5% CO2 when colony forming units (CFU) was determined. All experiments were conducted in triplicates.

Depletion of CD4+CD25+ regulatory T cells

Depletion of Treg cells was accomplished by magnetic separation using MACS human CD4 CD25 regulatory T cell isolation kit according to manufacturers instructions (Miltenyi Biotec, Germany). Briefly, CD4+ cells were first isolated by negative selection, then CD4+CD25+ cells were isolated by positive selection, resulting in 95% purity based on FoxP3 expression as reported previously [13] (S2A and S2B Fig).

Flow cytometry

Intra- and extracellular labeling were performed using monoclonal antibodies (MoAb) (BD Immunocytometry, BD Biosciences, San Jose, CA, USA; Biolegend, California, USA) conjugated with fluorescein isothiocyanate (FITC); phycoerythrin (PE), peridinin chlorophyll (PerCP), phycoerythrin-cyanine 7 (PE-Cy7). Cell surface markers were labeled using MoAb specific for CD25 (clone BC96), CD4 (SK3) markers, isotype control IgG2a (eBM2a), IgG2b (eBMG2b) and anti-IgG1 (P3). Cell surface markers were first labeled in FACS buffer (PBS 0.1% BSA 0.05% sodium azide) at 4°C for 30 min. Then, cells were washed, fixed, permeabilized using FoxP3 Staining Buffer (eBioscience, CA, USA), and incubated at 4°C for 30 min. for intracellular labeling of T-bet (a Th1 specific T-box transcription factor expressed in T cell) (clone 4B10) and FoxP3 (Forkhead box P3 transcription factor) (clone 236A/E7). Data was acquired with Attune NxT flow cytometer (Life Technologies, Carlsbad, CA, USA) and analyzed using FlowJo software (TreeStar, San Carlos, CA, USA).

Detection of Soluble IFN-γ

Soluble IFN-γ concentration in culture supernatants was measured using a commercial kit according to the manufacturer’s instructions (Human ELISA Ready-Set-Go kit; eBioscience Inc. California, San Diego, USA). The absorbance was recorded at 570 nm and 450 nm on SpectraMax M3 (Molecular Devices, Sunnyvale, CA, USA), and the detection limit was 4pg to IFN-γ.

In vitro evaluation of phagocytic activity in differentiated human macrophages

PBMCs (1×106 cells/mL) were incubated at 37°C/5%CO2 in 16-well tissue culture chamber slides (Nalge Nunc International). Four hours after incubation, monolayers were washed three times with RPMI 1640 medium supplemented with 10% inactivated fetal bovine serum (FBS) to remove non-adherent cells. Adherent cells were cultivated with rGM-CSF (10μg/mL stock R&D) for 5 days, followed by infection with either H37Rv strain or R6 Mtb isolate (MOI of 10:1) for 4h, then washed three times with 10% FBS RPMI 1640. Cells were divided in two samples: one readily fixed and stained using the Kinyoun method as described previously [30], and another group incubated at 37°C for 24h, followed by staining. Slides were examined using an optical microscope (Zeiss Axiostar) and intracellular bacteria load counted (number of cells containing intracellular bacilli x 200 fields).

Statistical analyses

Normally distributed variables were compared using ANOVA analysis simple factorial test and by one-way ANOVA-Tukey’s honestly significant difference (Tukey’s HSD) post-hoc method. A 5% significance level (p<0.05) was used to determine statistical significance. Statistical analysis was performed using GraphPad Prism software 7.0 (GraphPad Software, San Diego, CA, EUA).

Results

Characteristics of the studied population

Thirty-seven HIV-negative subjects were enrolled and divided in three groups: a) Healthy Controls (n = 13), b) LTBI (n = 13), and c) TB group (n = 11). Healthy control group comprised 3 males (median age ± SD = 31±10.6 years) and 10 females (26.5±5.1 years), the LTBI group 4 males (29±6.2 years) and 9 females (30±9.8 years) and the TB group 6 males (39±8.6 years) and 5 females (28±6.6 years), as described in Table 2. BCG vaccination after birth is mandatory in Brazil since 1960; therefore, all participants were vaccinated before enrollment. Age and gender distribution did not differ significantly among the studied groups, patients with helminthiasis were excluded. A few individuals with a discordant result between TST and IGRA (TST-/IGRA+ or TST+/IGRA-) were excluded to prevent bias during further analyzes.

Ex-vivo frequency of CD4+CD25highFoxP3+ cells are high in TB patients and LTBI individuals when compared to healthy controls

After venipuncture, whole blood samples were immediately stained for CD4+CD25highFoxP3+ and analyzed. A high frequency of CD4+CD25highFoxP3+ cells was observed in patients with active pulmonary tuberculosis when compared to healthy control and LTBI subjects, and when LTBI patients were compared to healthy controls (p<0.05) (Fig 1).

thumbnail
Fig 1. Ex-vivo frequency of CD4+CD25highFoxP3+ in TB patients are higher when compared to HC and LTBI subjects.

Peripheral blood from healthy control subjects (HC, n = 10), latent infected individuals (LTBI, n = 10), and active pulmonary TB patients (TB, n = 6). Ex vivo samples were collected and immunophenotyped for the presence of CD4+CD25highFoxP3+ cells (** = p<0.01).

https://doi.org/10.1371/journal.pntd.0009605.g001

Frequency of CD4+CD25highFoxP3+ cells in LTBI individuals and TB patients is significantly augmented in whole blood cultures after Mtb infection

To investigate if CD4+CD25highFoxP3+ cell frequency differed among the studied groups, whole peripheral blood samples from TB patients, healthy control and LTBI subjects were cultured for 72h in the presence or absence of live Mtb H37Rv strain. After, in vitro stimulation, CD4+CD25highFoxP3+ cell frequency was further increased, in both LTBI and TB groups, upon exposure to live Mtb strain (H37Rv), or upon exposure to five different clinical isolates. On the other hand, no significant change was observed in the healthy control group regardless of the stimuli used (Fig 2).

thumbnail
Fig 2. Frequency of CD4+CD25highFoxP3+ cells is increased in peripheral blood of LTBI and active TB patients.

Peripheral blood from healthy control subjects (HC, n = 10), latent infected individuals (LTBI, n = 10), and active pulmonary TB patients (TB, n = 6) were cultivated in the presence or absence of live Mtb by 72 hours, followed by staining with fluorochrome-conjugated antibodies to CD4, CD25 and FoxP3 and flow cytometry analysis. Frequency of CD4+CD25highFoxP3+ cells was determined after in vitro infection with multidrug sensitive (S1 and S5) clinical isolates, multidrug-resistant (R3 and R6) and reference strain H37RV. Statistical relevance was assessed using the ANOVA test. Bars indicating statistical significance between groups are shown (* = p<0.05); Ctrl = Control.

https://doi.org/10.1371/journal.pntd.0009605.g002

To determine if the observed increase on CD4+CD25highFoxP3+ cell frequency was a strain-dependent phenomenon, the experiment was also conducted with both multidrug-resistant (R3 and R6) and drug-susceptible (S1 and S5) clinical isolates. The same profile observed for H37Rv was also reported for all the isolates used, regardless of its drug susceptibility status (Fig 2), suggesting that the expansion on the Treg population was neither strain/isolate dependent. These results concur with the hypothesis that there is an increase in antigen-specific Treg cell frequency in latent and active TB disease.

Increase in CD4+CD25highFoxP3+ population is accompanied by a decline in microbicidal activity in whole blood cultures from LTBI and TB donors

Considering that CD4+CD25highFoxP3+ cell frequency was elevated on both LTBI and TB groups (Fig 2), we analyzed if it could hinder Th1-dependent immune responses and negatively impact Mtb clearance. Data presented here show that the increase in CD4+CD25highFoxP3+ cell frequency (Fig 2) was accompanied by a significant expansion of CFU/mL numbers, in both LTBI and TB groups, for all the strains/isolates examined (Fig 3).

thumbnail
Fig 3. Whole blood microbicidal activity is significantly reduced on both LTBI individuals and active TB patients.

Data represent Colony Forming Units (CFU/mL) counts for all the strain and clinical isolates (H37Rv, S1, S5, R3 and R6) in healthy control (n = 10), LTBI (n = 10), and TB (n = 6) groups. Bar indicates the median of CFU/mL counts. Statistical relevance (*** = p< 0.0001) was assessed using ANOVA test. CFU = Colony Forming Unit.

https://doi.org/10.1371/journal.pntd.0009605.g003

To rule out the possibility that the observed lower CFU/mL counts in the control group was due to impairment of phagocytosis, an in-vitro assay to evaluate the phagocytic activity was performed using in vitro differentiated macrophages derived from PBMCs of LTBI and healthy controls. As depicted in S3 Fig, a similar phagocytic activity was reported in both groups, regardless of incubation time (4 vs 24hs), TST status, or strain/clinical isolate used. These data suggest that microbicidal activity is hindered in LTBI and TB groups and might be associated with the observed Treg cell upregulation.

Depletion of Treg-like cells restores in vitro microbicidal capacity

To assess whether the reduced microbicidal activity reported for LTBI and TB groups was indeed associated with an increase in CD4+CD25highFoxP3+ cell frequency, in vitro microbicidal activity assays using either whole PBMC or Treg-depleted PBMC samples were performed. Before carrying out the depletion experiments, the microbicidal assay using PBMC instead of whole blood samples was validated. PBMC and whole blood samples were obtained from the same donors and processed simultaneously. As demonstrated in S4 Fig, results from microbicidal assays using either PBMC or whole blood samples are comparable.

As hypothesized, depletion of CD4+CD25+ cells restored the in vitro microbicidal activity on PBMC samples from LTBI and TB groups to levels comparable to those observed in healthy control subjects (Fig 4). In vitro microbicidal activity on both LTBI and TB groups were significantly enhanced after CD4+CD25+ cell depletion (p<0.05), as demonstrated by a strong decline in CFU/mL counts (Fig 4). As observed previously, results were similar regardless of the isolate/strain used (Fig 4), confirming that the depletion of these cells boosted the microbicidal activity in PBMC samples from both LTBI and TB groups. Together, these results support our hypothesis that the expansion of Treg cells observed during LTBI, and active TB disease is associated with an impairment of host’s microbicidal activity.

thumbnail
Fig 4. Depletion of CD4+CD25+ T cells restores in-vitro microbicidal activity in PBMC samples from LTBI individuals and TB patients.

PBMC samples from healthy control subjects (HC, n = 10), latent infected individuals (LTBI, n = 10) and active TB patients (TB, n = 8) were depleted of CD4+CD25+ cells. Whole PBMC and Treg-depleted PBMC samples were infected with Mtb strains for 72 hours and then processed for quantification of Colony Forming Units (CFU). Individual data from CFU/mL counts on whole PBMC and CD4+CD25+ cells depleted PBMC cultures infected with the reference strain H37RV, drug-susceptible (S1 and S5) or multidrug-resistant (R3 and R6) isolates. Bar indicates the median value of CFU/mL counts. Statistical relevance (* = p<0.05) was assessed using the ANOVA test. CFU = Colony Forming Unit.

https://doi.org/10.1371/journal.pntd.0009605.g004

Depletion of CD4+CD25+FoxP3+ cells restored Th1 profile in LTBI subjects

Treg cell expansion has been associated with hindered Th1-dependent immune responses [13]. Therefore, we investigated if intracellular expression of T-bet and secretion of soluble IFN-γ in H37RV infected PBMC cultures were altered in presence or absence of Treg cells. In the presence of Tregs, T-bet expression in healthy controls was close to eight-fold greater than in LTBI subjects. As expected, following Treg depletion, intracellular expression of T-bet on CD4+ cells was significantly increased in both groups (Fig 5A), with a remarkable intensification in the LTBI group. After Treg depletion, the expression of intracellular T-bet was similar in both control and LTBI groups (Fig 5A).

thumbnail
Fig 5. Depletion of CD4+CD25+FoxP3+ T cells restored the TH1 profile in LTBI individuals.

PBMC samples from healthy control subjects (HC, n = 5) and latent infected individuals (LTBI, n = 5) were depleted of CD4+CD25+ cells. Whole PBMC and Treg-depleted PBMC samples were infected with Mtb H37Rv strain for 72 hours in vitro. (A) Intracellular expression of T-bet analyzed on CD4-gated population by flow cytometry; (B) Soluble levels of IFN-y in PBMC culture supernatants determined by ELISA. Statistical relevance (* = p<0.05; *** = p<0,0001) was assessed using ANOVA test.

https://doi.org/10.1371/journal.pntd.0009605.g005

As depicted in Fig 5B, the concentration of soluble IFN-γ in PBMC culture supernatants in the LTBI group was boosted significantly after Treg depletion, which corroborates with the T-bet data and the hypothesis that the increase of CD4+CD25highFoxP3+ cells hinders Th1 response.

Discussion

Progress to TB disease following Mtb infection depends on the outcome of a subtle balance between genetic factors involved in susceptibility and resistance [31] and the ability of the host to mount an effective immune response [3234]. Immune regulatory mechanisms, such as Treg cells, are known to limit inflammatory immune responses [8,35,36] and have been associated with thwarted immune responses against Mtb which if overrepresented, may contribute to the persistence and/or establishment of chronic infections [1,4]. Therefore, it is plausible to expect that upregulation in the Treg cell population may lead to disease reactivation [37] or re-infection [13,18,35,38,39]. Data presented here demonstrate that ex-vivo (Fig 1) and in vitro (Fig 2) Treg population is significantly augmented in LTBI subjects and patients with active TB when compared to healthy controls. These findings are supported by previous studies showing a higher frequency of CD4+CD25highFoxP3+ cells in both pulmonary TB patients and LTBI subjects, when compared to other groups, suggesting a potential role of these cells in disease progression [3,10,11,13,14,1618,35,40]. Although it is still not clear whether increased in vitro Treg cell frequency correlates with progression of TB disease in humans, data exhibiting a significant in vitro expansion of these cells on samples from TB patients after stimulation [11,13,36,37,4143] support this hypothesis. A possible explanation for the observed significant increase in Treg cell frequency in LTBI individuals comes from data showing that CD4+ T cells expansion during exposure to Mtb could induce the upregulation of Treg cells specific to Mtb antigens [13,44].

Other studies suggest that different Mtb strains induced Foxp3+ cells expansion distinctively [45,46]. Data presented here demonstrated that the observed Treg frequencies associated with drug-susceptible (DS strains, H37Rv, S1 and S5) and drug-resistant (DR strains, R3 and R6) Mtb strains in whole blood cultures did not differ (Fig 2), concurring with Lim et al. [47]. These authors showed that frequencies of Treg cells in TB patients infected with either drug-susceptible Mtb or multidrug-resistant Mtb were similar but significantly higher when compared to healthy controls [47], corroborating with our findings.

A modified ex vivo whole blood bactericidal assay [29] allowed us to confirm that the upregulation of CD4+CD25highFoxP3+ cells was associated with impaired microbicidal activity in latent and active TB groups when compared to healthy control subjects, regardless of the strain used. This difference was not due to alterations in bacterial phagocytosis assay (S3 Fig) but the depletion of CD4+CD25highFoxP3+ cells in the culture restored the microbicidal activity in LTBI and TB group regardless of Mtb strain/isolates used. This result corroborates previous findings showing that peripheral blood Tregs from patients with pulmonary TB hindered the ability of alveolar and monocyte-derived macrophages to restrict Mtb growth [48]. Data presented here also confirm previous results showing that CD4+CD25highFoxP3+ cells depletion in PBMC cultures from LTBI subjects restores IFN-γ production and expression of T-bet, enhancing the Th1-immune response (Fig 5A and 5B) [12,13].

Our work has limitations. First, since a large proportion of patients with pulmonary tuberculosis were under the use of anti-inflammatory drugs, which could hinder the assessment of cytokines, cell phenotype, and the “in vitro” killing assay; enrolling TB patients who were not using such medications to participate in the present study was a challenge, which limited the number of enrolled TB patients. Second, frequently the final volume of collected blood samples was smaller than required, impacting directly on the number of experiments that could be performed per subject. Third, the great majority of enrolled TB patients refused to provide stool samples for intestinal worms testing; therefore, the impact of the presence of intestinal worms could not be assessed. Fourth, all enrolled patients with pulmonary tuberculosis presented a 3+ or higher grade bacilloscopy result, which hindered the correlation of bacillary load and the “in vitro” killing assay results. And finally, we were unable to enroll discordant cases separately, which due to the necessary sampling size would require mass testing for both IGRA and TST; therefore, analyzing discordant patients as a separate group was not possible. Limitations, notwithstanding, in this study, we demonstrate that an increase in CD4+CD25highFoxP3+ population thwarts the host´s ability to restrict Mtb growth. Therefore, it may facilitate both mycobacterial survival and intracellular persistence in vivo. Taken together, the results presented here may contribute to further our knowledge about the reason why the age-adjusted incidence rate of TB reinfection after treatment was four times greater than new TB cases [20]. Additionally, it also may help to elucidate why case contacts with prior positive TST and people who had TB previously present a significantly higher risk of developing active TB disease if reinfected, when compared to healthy subjects [21,22]. Our findings support the hypothesis that both latent and active Mtb infection led to an upregulation in Treg cell frequency, which in turn may down-regulate the ability of the host to kill intracellular mycobacteria, significantly increasing the risk of TB disease progression/reactivation in LTBI individuals or of reinfection of those treated and cured after a re-exposure to Mtb.

Supporting information

S1 Fig. IS6110 RFLP pattern for Mtb clinical isolates used in the study.

RFLP patterns demonstrate that selected clinical isolates are distinct from each other. Legend: S1 and S5 –drug-susceptible isolates, R3 and R6—drug-resistant isolates, and CR–MT 14323 reference strain used for IS6110 RFLP typing.

https://doi.org/10.1371/journal.pntd.0009605.s001

(TIF)

S2 Fig. Gating strategy.

(A) Specific gating strategy was performed using CD4+, CD25high+ and FoxP3+ expression before and after depletion of CD4+CD25+ cells. (B) FoxP3+ expression assessed by flow cytometry analysis of intracellular FOXP3+ on CD4+ cells before and after CD4+CD25+ depletion assay (*** = p<0.0001).

https://doi.org/10.1371/journal.pntd.0009605.s002

(TIF)

S3 Fig. Evaluation of in vitro phagocytic activity in human differentiated macrophages upon infection with live M. tuberculosis.

Phagocytic activity was evaluated in vitro in PBMC samples from healthy control subjects (HC) or latently infected individuals (LTBI) by quantification of intracellular mycobacteria after 4h and 24h of exposure to live Mtb H37Rv strain and R6 isolate, via Kinyoun acid-fast staining. Bars indicating statistical significance between groups are shown (* = p<0.05).

https://doi.org/10.1371/journal.pntd.0009605.s003

(TIF)

S4 Fig. Validation of the in-vitro microbicidal activity assay using PBMC samples.

Paired whole blood (WB) and PBMC samples from healthy control subjects (n = 10), LTBI individuals (n = 10) and TB patients (n = 7) were infected with the reference strain H37RV or drug-susceptible (S1 and S5) or multidrug-resistant (R3 and R6) clinical isolates.

https://doi.org/10.1371/journal.pntd.0009605.s004

(TIF)

Acknowledgments

We thank physicians and clinical staff at UFES TB outpatient clinic who have kindly assisted us in collecting blood samples from the study participants.

References

  1. 1. Kursar M, Koch M, Mittrucker HW, Nouailles G, Bonhagen K, Kamradt T, et al. Cutting Edge: Regulatory T cells prevent efficient clearance of Mycobacterium tuberculosis. J Immunol. 2007;178(5):2661–5. pmid:17312107
  2. 2. World_Health_Organization. Latent tuberculosis infection: updated and consolidated guidelines for programmatic management. Geneva: World Health Organization;. WHO; 2018.
  3. 3. Pandey P, Bhatnagar AK, Mohan A, Sachdeva KS, Vajpayee M, Das BK, et al. Insights in tuberculosis immunology: Role of NKT and T regulatory cells. Int J Mycobacteriol. 2019;8(4):333–40. pmid:31793502
  4. 4. Parkash O, Agrawal S, Madhan Kumar M. T regulatory cells: Achilles’ heel of Mycobacterium tuberculosis infection? Immunol Res. 2015;62(3):386–98. pmid:25948475
  5. 5. Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science. 2003;299(5609):1057–61. pmid:12522256
  6. 6. Chatenoud L. Natural and induced T CD4+CD25+FOXP3+ regulatory T cells. Methods Mol Biol. 2011;677:3–13. pmid:20941599
  7. 7. Curotto de Lafaille MA, Lafaille JJ. Natural and adaptive foxp3+ regulatory T cells: more of the same or a division of labor? Immunity. 2009;30(5):626–35. pmid:19464985
  8. 8. Belkaid Y, Tarbell KV. Arming Treg cells at the inflammatory site. Immunity. 2009;30(3):322–3. pmid:19303386
  9. 9. de Almeida AS, Fiske CT, Sterling TR, Kalams SA. Increased frequency of regulatory T cells and T lymphocyte activation in persons with previously treated extrapulmonary tuberculosis. Clin Vaccine Immunol. 2012;19(1):45–52. pmid:22038848
  10. 10. Feruglio SL, Tonby K, Kvale D, Dyrhol-Riise AM. Early dynamics of T helper cell cytokines and T regulatory cells in response to treatment of active Mycobacterium tuberculosis infection. Clin Exp Immunol. 2015;179(3):454–65. pmid:25313008
  11. 11. Guyot-Revol V, Innes JA, Hackforth S, Hinks T, Lalvani A. Regulatory T cells are expanded in blood and disease sites in patients with tuberculosis. Am J Respir Crit Care Med. 2006;173(7):803–10. pmid:16339919
  12. 12. Marin ND, Paris SC, Velez VM, Rojas CA, Rojas M, Garcia LF. Regulatory T cell frequency and modulation of IFN-gamma and IL-17 in active and latent tuberculosis. Tuberculosis (Edinb). 2010;90(4):252–61. pmid:20594914
  13. 13. Ribeiro-Rodrigues R, Resende Co T, Rojas R, Toossi Z, Dietze R, Boom WH, et al. A role for CD4+CD25+ T cells in regulation of the immune response during human tuberculosis. Clin Exp Immunol. 2006;144(1):25–34. pmid:16542361
  14. 14. Wergeland I, Assmus J, Dyrhol-Riise AM. T regulatory cells and immune activation in Mycobacterium tuberculosis infection and the effect of preventive therapy. Scand J Immunol. 2011;73(3):234–42. pmid:21204895
  15. 15. Sahmoudi K, Abbassi H, Bouklata N, El Alami MN, Sadak A, Burant C, et al. Immune activation and regulatory T cells in Mycobacterium tuberculosis infected lymph nodes. BMC Immunol. 2018;19(1):33. pmid:30409122
  16. 16. Whittaker E, Nicol M, Zar HJ, Kampmann B. Regulatory T Cells and Pro-inflammatory Responses Predominate in Children with Tuberculosis. Front Immunol. 2017;8:448. pmid:28487695
  17. 17. Hougardy JM, Place S, Hildebrand M, Drowart A, Debrie AS, Locht C, et al. Regulatory T cells depress immune responses to protective antigens in active tuberculosis. Am J Respir Crit Care Med. 2007;176(4):409–16. pmid:17541018
  18. 18. Serrano CJ, Castaneda-Delgado JE, Trujillo-Ochoa JL, Gonzalez-Amaro R, Garcia-Hernandez MH, Enciso-Moreno JA. Regulatory T-cell subsets in response to specific Mycobacterium tuberculosis antigens in vitro distinguish among individuals with different QTF and TST reactivity. Clin Immunol. 2015;157(2):145–55. pmid:25728490
  19. 19. Herzmann C, Ernst M, Ehlers S, Stenger S, Maertzdorf J, Sotgiu G, et al. Increased frequencies of pulmonary regulatory T-cells in latent Mycobacterium tuberculosis infection. Eur Respir J. 2012;40(6):1450–7. pmid:22441737
  20. 20. Verver S, Warren RM, Beyers N, Richardson M, van der Spuy GD, Borgdorff MW, et al. Rate of reinfection tuberculosis after successful treatment is higher than rate of new tuberculosis. Am J Respir Crit Care Med. 2005;171(12):1430–5. pmid:15831840
  21. 21. Andrews JR, Noubary F, Walensky RP, Cerda R, Losina E, Horsburgh CR. Risk of progression to active tuberculosis following reinfection with Mycobacterium tuberculosis. Clin Infect Dis. 2012;54(6):784–91. pmid:22267721
  22. 22. Gounder PP, Harris TG, Anger H, Trieu L, Meissner JS, Cadwell BL, et al. Risk for Tuberculosis Disease Among Contacts with Prior Positive Tuberculin Skin Test: A retrospective Cohort Study, New York City. J Gen Intern Med. 2015;30(6):742–8. pmid:25605533
  23. 23. Coelho da Silva FD, Covre LP, Stringari LL, Palaci M, Dietze R, Gomes DCO, et al. Toll-like receptors blocking restores in vitro microbicidal activity in latent tuberculosis-infected subjects. Int J Tuberc Lung Dis. 2019;23(2):212–8. pmid:30808454
  24. 24. Barnes PF, Cave MD. Molecular epidemiology of tuberculosis. N Engl J Med. 2003;349(12):1149–56. pmid:13679530
  25. 25. Vynnycky E, Fine PE. The natural history of tuberculosis: the implications of age-dependent risks of disease and the role of reinfection. Epidemiol Infect. 1997;119(2):183–201. pmid:9363017
  26. 26. Bjartveit K. Olaf Scheel and Johannes Heimbeck: their contribution to understanding the pathogenesis and prevention of tuberculosis. Int J Tuberc Lung Dis. 2003;7(4):306–11. pmid:12729334
  27. 27. Sokal JE. Editorial: Measurement of delayed skin-test responses. N Engl J Med. 1975;293(10):501–2. pmid:1152865
  28. 28. Ribeiro FK, Pan W, Bertolde A, Vinhas SA, Peres RL, Riley L, et al. Genotypic and Spatial Analysis of Mycobacterium tuberculosis Transmission in a High-Incidence Urban Setting. Clin Infect Dis. 2015;61(5):758–66. pmid:25948063
  29. 29. Wallis RS, Palaci M, Vinhas S, Hise AG, Ribeiro FC, Landen K, et al. A whole blood bactericidal assay for tuberculosis. J Infect Dis. 2001;183(8):1300–3. pmid:11262217
  30. 30. Selvakumar N, Gomathi M, Rehman F, Narayanan PR. Evaluation of a two-reagent cold staining method for detection of acid-fast bacilli. Int J Tuberc Lung Dis. 2002;6(8):728–31. pmid:12150486
  31. 31. Davies PD, Grange JM. Factors affecting susceptibility and resistance to tuberculosis. Thorax. 2001;56 Suppl 2:ii23–9. pmid:11514703
  32. 32. Geffner L, Yokobori N, Basile J, Schierloh P, Balboa L, Romero MM, et al. Patients with multidrug-resistant tuberculosis display impaired Th1 responses and enhanced regulatory T-cell levels in response to an outbreak of multidrug-resistant Mycobacterium tuberculosis M and Ra strains. Infect Immun. 2009;77(11):5025–34. pmid:19720756
  33. 33. Manca C, Reed MB, Freeman S, Mathema B, Kreiswirth B, Barry CE 3rd, et al. Differential monocyte activation underlies strain-specific Mycobacterium tuberculosis pathogenesis. Infect Immun. 2004;72(9):5511–4. pmid:15322056
  34. 34. Rajavelu P, Das SD. Th2-type immune response observed in healthy individuals to sonicate antigen prepared from the most prevalent Mycobacterium tuberculosis strain with single copy of IS6110. FEMS Immunol Med Microbiol. 2005;45(1):95–102. pmid:15985228
  35. 35. Chen X, Zhou B, Li M, Deng Q, Wu X, Le X, et al. CD4(+)CD25(+)FoxP3(+) regulatory T cells suppress Mycobacterium tuberculosis immunity in patients with active disease. Clin Immunol. 2007;123(1):50–9. pmid:17234458
  36. 36. Chiacchio T, Casetti R, Butera O, Vanini V, Carrara S, Girardi E, et al. Characterization of regulatory T cells identified as CD4(+)CD25(high)CD39(+) in patients with active tuberculosis. Clin Exp Immunol. 2009;156(3):463–70. pmid:19438599
  37. 37. Hougardy JM, Verscheure V, Locht C, Mascart F. In vitro expansion of CD4+CD25highFOXP3+CD127low/- regulatory T cells from peripheral blood lymphocytes of healthy Mycobacterium tuberculosis-infected humans. Microbes Infect. 2007;9(11):1325–32. pmid:17890131
  38. 38. Luo J, Zhang M, Yan B, Zhang K, Chen M, Deng S. Imbalance of Th17 and Treg in peripheral blood mononuclear cells of active tuberculosis patients. Braz J Infect Dis. 2017;21(2):155–61. pmid:27932286
  39. 39. Sterling TR, Dorman SE, Chaisson RE, Ding L, Hackman J, Moore K, et al. Human immunodeficiency virus-seronegative adults with extrapulmonary tuberculosis have abnormal innate immune responses. Clin Infect Dis. 2001;33(7):976–82. pmid:11528568
  40. 40. Singh A, Dey AB, Mohan A, Sharma PK, Mitra DK. Foxp3+ regulatory T cells among tuberculosis patients: impact on prognosis and restoration of antigen specific IFN-γ producing T cells. PLoS One. 2012;7(9):e44728. pmid:23028594
  41. 41. Henao-Tamayo MI, Obregon-Henao A, Arnett K, Shanley CA, Podell B, Orme IM, et al. Effect of bacillus Calmette-Guerin vaccination on CD4+Foxp3+ T cells during acquired immune response to Mycobacterium tuberculosis infection. J Leukoc Biol. 2016;99(4):605–17. pmid:26590147
  42. 42. Li L, Lao SH, Wu CY. Increased frequency of CD4(+)CD25(high) Treg cells inhibit BCG-specific induction of IFN-gamma by CD4(+) T cells from TB patients. Tuberculosis (Edinb). 2007;87(6):526–34. pmid:17851131
  43. 43. Orme IM, Robinson RT, Cooper AM. The balance between protective and pathogenic immune responses in the TB-infected lung. Nat Immunol. 2015;16(1):57–63. pmid:25521685
  44. 44. Shafiani S, Tucker-Heard G, Kariyone A, Takatsu K, Urdahl KB. Pathogen-specific regulatory T cells delay the arrival of effector T cells in the lung during early tuberculosis. J Exp Med. 2010;207(7):1409–20. pmid:20547826
  45. 45. Li N, Xie WP, Kong H, Min R, Hu CM, Zhou XB, et al. Enrichment of regulatory T-cells in blood of patients with multidrug-resistant tuberculosis. Int J Tuberc Lung Dis. 2015;19(10):1230–8. pmid:26459539
  46. 46. Ordway D, Henao-Tamayo M, Harton M, Palanisamy G, Troudt J, Shanley C, et al. The hypervirulent Mycobacterium tuberculosis strain HN878 induces a potent TH1 response followed by rapid down-regulation. J Immunol. 2007;179(1):522–31. pmid:17579073
  47. 47. Lim HJ, Park JS, Cho YJ, Yoon HI, Park KU, Lee CT, et al. CD4(+)FoxP3(+) T regulatory cells in drug-susceptible and multidrug-resistant tuberculosis. Tuberculosis (Edinb). 2013;93(5):523–8.
  48. 48. Semple PL, Binder AB, Davids M, Maredza A, van Zyl-Smit RN, Dheda K. Regulatory T cells attenuate mycobacterial stasis in alveolar and blood-derived macrophages from patients with tuberculosis. Am J Respir Crit Care Med. 2013;187(11):1249–58. pmid:23590266