GET THE APP

Hyaluronan-Binding T Regulatory Cells in Peripheral Blood of Brea
Journal of Clinical and Cellular Immunology

Journal of Clinical and Cellular Immunology
Open Access

ISSN: 2155-9899

+44 1223 790975

Research Article - (2015) Volume 6, Issue 1

Hyaluronan-Binding T Regulatory Cells in Peripheral Blood of Breast Cancer Patients

Yuliya Perfilyeva1, Yekaterina Ostapchuk1, Esin Aktas Cetin2, Abdullah Yilmaz2, Gunnur Deniz2, Shynar Talaeva3, Nazgul Omarbaeva3, Igor Oskolchenko1 and Nikolai Belyaev1*
1Laboratory of Molecular Immunology and Immunobiotechnology, M.A. Aitkhozhin’s Institute of Molecular Biology and Biochemistry, 050012 Almaty, Kazakhstan
2Department of Immunology, Institute of Experimental Medicine (DETAE), Istanbul University, 34393 Istanbul, Turkey
3Mammology Center, Research Institute of Radiology and Oncology, 480072 Almaty, Kazakhstan
*Corresponding Author: Nikolai Belyaev, Doctor of Biological Sciences, Professor, M.A. Aitkhozhin’s Institute of Molecular Biology and Biochemistry, Laboratory of Molecular Immunology and Immunobiotechnology, 86 Dosmukhamedov St., 050012, Almaty, Kazakhstan, Tel: +7-705-874-08-50, Fax: +7-727-293-70-92 Email:

Abstract

Regulatory T cells (Treg), both natural and induced, play an important role in maintaining immune homeostasis. Alterations in the number and functions of Tregs are involved in tumor growth. One of the possible regulatory mechanisms of Treg functional activity involves interaction with major component of extracellular matrix hyaluronan. It has been demonstrated that high molecular weight hyaluronan promotes Treg function via increased expression of FoxP3 and production of IL-10. Moreover, previous research has shown highly enhanced suppressor function of hyaluronan-binding CD4+CD25+ Tregs in mice. Breast cancer is characterized by upregulated production of tumor-associated hyaluronan, therefore we investigated hyaluronan-binding subset of Tregs obtained from peripheral blood of breast cancer patients. As a result, we showed that the majority of peripheral blood Tregs were able to adhere to immobilized hyaluronan, and these cells exerted superior suppressor activity, suggesting a key role in regulatory functions of these cells. The percentage of CD4+FoxP3+ Treg cells binding hyaluronan, as well as CD39+ hyaluronan-binding Tregs were significantly increased in breast cancer patients compared to healthy donors. Enhanced number of the activated Treg cells might play an important role in the suppression of antitumor immunity.

Keywords: Regulatory T cells; Hyaluronan; Breast cancer; CD39; CTLA-4

Introduction

Regulatory T cells can be described as a T cell population that functionally suppresses immune response by influencing the activity of a range of effector cells, and thereby contributes to the maintenance of immune homeostasis. CD4+ Tregs consist of two types, “natural” Tregs (nTregs) that constitutively express CD25 and FoxP3, and so-called adaptive or “inducible” Tregs (iTregs). iTreg cells can be induced in the periphery from a CD4+FoxP3- T cell population following T cell receptor (TCR) stimulation in the presence of immunoregulatory cytokines such as TGF-β, IL-10, and IL-4. nTregs and iTregs suppress immune responses through various cytokines and contact-dependent mechanisms [1,2].

One of the central mechanisms that mediate Treg recruitment from the blood to sites of inflammation or tumor growth is mediated through interaction between the activated form of CD44 on peripheral Tregs and its ligand hyaluronan (HA) on microvascular endothelium [3]. CD44, a type I transmembrane glycoprotein, is widely expressed on T lymphocytes but requires activation for binding HA. Transition from low- to high-affinity binding state can be activated in T cells by several stimuli, such as HA-binding itself, TCR engagement, and responses to cytokines/chemokines, and one mechanism involves the enzymatic removal of terminal sialic acid from two N-linked glycans in the HA-binding domain [4-7]. Therefore, the ability of Tregs to interact with HA is intrinsically related to their activation state.

Previous studies raised the possibility that CD44 interactions with HA may be integrally related to Treg functions. In mice HA-binding CD4+CD25+ Treg cells showed highly enhanced suppressor activity in vitro [8]. Tregs from CD44-deficient mice have an impaired capacity to inhibit T cell responses. In vitro ligation of CD44 on activated Tregs promotes persistent expression of FoxP3, increased production of IL-10 and expression of membrane TGF-β, which are necessary for immunoregulatory activity. These effects on Tregs are shown to depend upon interaction with a high molecular weight form of HA [9].

The rates of HA synthesis and degradation are much higher in cancer than in healthy tissues [10,11]. It has been demonstrated that in advanced cancer, aberrant synthesis and degradation of HA by transformed cells result in the formation of an extremely unusual microenvironment characterized by the accumulation of high molecular weight HA, which may facilitate the malignant transformation and survival of tumor cells, and affect functions of immune cells [12]. Several lines of evidence indicate that malignant breast tissue contains more HA than normal breast tissue or benign lesions [13-18].

These studies with other indirect evidences lead us to formulate the hypothesis that in cancer, Tregs bind with HA whose gradient exists in peripheral blood, and migrate to the tumor, where interaction of CD44 with high molecular weight HA increases the suppressive potential of Tregs and results in the suppression of antitumor immunity. To test the hypothesis, HA-binding Treg cells from the peripheral blood of breast cancer patients were purified, and their phenotype related to specific suppressor activity was assessed.

Materials and Methods

Study Subjects

Peripheral blood samples were obtained from healthy volunteers and patients with breast cancer. Only patients without prior chemotherapy or other treatments and surgical removal of tumor were included in the study. Both the volunteers and the patients provided their informed consent for participation in the investigation. Of 16 patients, 14 carried stage II or higher disease (Table 1).

  Study subjects
Normal Breast cancer patients
Donors 20 16
Average age 38.2 ± 11.7 52.8 ± 6.6
Stage I - 2
Stage II - 11
Stage III - 3
Stage IV - 0

Table 1: Patient characteristics.

Isolation of CD4+ and Treg cells from peripheral blood

Human peripheral blood mononuclear cells (PBMCs) were obtained from heparinized blood by density gradient centrifugation over Histopaque-1077 (Sigma-Aldrich, St. Louis, MO). The CD4+ cell rich fraction was isolated by negative selection on a Miltenyi Biotec Vario Max separator using the CD4+ T Cell Isolation Kit, human (Miltenyi Biotec AG, Bergisch Gladbach, Germany) according to the manufacturer’s instructions. The purity of the resultant CD4+ cell fraction was evaluated on a BD FACS Calibur (BD Biosciences, San Jose, CA) using monoclonal antibody (mAB) anti-CD4-PerCP (Miltenyi Biotec AG, Bergisch Gladbach, Germany) and resulted in 95%. For some experiments, CD4+CD25+ Treg cells and CD4+CD25- conventional T cells were sorted on a BD FACSAria II from freshly obtained PBMC using anti-CD4-APC-Cy7 (BD Biosciences, San Jose, CA) and anti-CD25-PerCP-Cy5,5 (BD Biosciences, San Jose, CA). The purity of sorted cells was >98%.

Separation of hyaluronan-binding CD4+ cells

For 107 cells, 15µg of biotinylated hyaluronan (Sigma-Aldrich. Co., St. Louis, MO) and 50µl of anti-biotin MACSi Bead Particles (Miltenyi Biotec AG, Bergisch Gladbach, Germany) were mixed in a 50µl solution containing phosphate buffer saline (PBS) supplemented with 0.5% Bovine Serum Albumin (BSA) and 2mM EDTA, and incubated for 2 h at 4-8°C under gentle rotation. After washing by centrifugation at 300g for 15 min at 7°C the conjugate was resuspended in 100µl of the same buffer solution. Cell suspension was mixed with conjugate at a 1:1 cell-to-bead ratio and incubated for 20 min at 4-8°C. Then the cells were magnetically separated into HA-binding (HA+) and HA-nonbinding (HA-) fractions using a BD IMag Separator (BD Biosciences, San Jose, CA).

Surface and Intracellular Staining

The following mouse mAbs were used: anti-CD4-PerCP, anti-CD25-PE, anti-FoxP3-PE, anti-FoxP3-APC, anti-CD39-PE, anti-CD39-FITC, anti-CTLA4-APC, anti-IL-17-FITC, anti-LAP-APC, anti-GITR-APC (Miltenyi Biotec AG, Bergisch Gladbach, Germany), anti-CD25-PerCP-Cy5.5, anti-CD25-FITC (BD Biosciences, San Jose, CA), anti-TGF-β-FITC, anti-IL-10-PE, anti-IL-10-FITC, anti-IL-35-APC (R&D Systems, Inc., Minneapolis, MN) and their relevant control isotypes. Cells were incubated with mAbs specific for surface markers for 20 min at 4-8°C in the dark and then fixed in Cytofix buffer (BD Biosciences, San Jose, CA). For intracellular staining, after surface labeling cells were permeabilized with paraformaldehyde/saponin solution (Cytofix & Cytoperm kit, BD Bioscience, San Jose, CA). Permeabilized cells were stained with mAbs specific for intracellular markers for 30 min at 4°C in the dark. FoxP3 Staining Buffer Set (Miltenyi Biotec AG, Bergisch Gladbach, Germany) was used for intracellular labeling of FoxP3 according to the manufacturer’s instruction. Afterward cells were analyzed by BD FACSCalibur with CellQuest Pro software (BD Bioscience, San Jose, CA).

For detection of HA-binding Treg cells, freshly purified PBMC were stained with biotinylated hyaluronan (15µg for 107 cells), mixed well and incubated for 20 min at 4-8°C. Then cells were washed, labeled with streptavidin-PE (R&D Systems, Inc., Minneapolis, MN) and Treg-associated markers and analyzed by flow cytometry.

In vitro cell culture

All the cultures were maintained in RPMI-1640 (Sigma Chem. Co., St. Louis, MO) supplemented with 10% fetal calf serum (FCS), penicillin (100U/ml), streptomycin (100mg/ml) and L-glutamine (2mM) at 37°C, 5% CO2 and 95% humidity. Freshly isolated HA+ and HA- fractions of CD4+ cells were activated with anti-CD3/anti-CD28/anti-CD2 bound to beads (Treg Suppression Inspector assay, TSI, Miltenyi Biotec AG, Bergisch Gladbach, Germany) at a cell-to-bead ratio of 2:1 for 18 h. After washing cells were stained with fluorochrome-labeled mAb and analyzed.

Suppression of CD4+CD25- conventional T lymphocytes

The suppressor effect of HA+ and HA- Tregs was determined by the ability to inhibit proliferation of autologous CFSE-labeled CD4+CD25- conventional T lymphocytes. In brief, sorted conventional T cells were resuspended at 2–3×106/ml in PBS with 0.1% BSA and kept on ice. CFSE solution (5μM) was added at volume equivalent to cell suspension and incubated at room temperature without agitation for 10 min. The reaction was quenched as quickly as possible with ice-cold fetal bovine serum (FBS). Subsequently, cells were immediately put on ice for 2 min, washed twice and resuspended at 5×105/ml. CD4+CD25+ Treg cells were fractionated into HA+ and HA- using biotinylated HA conjugated with anti-biotin MACSi Bead Particles. The obtained fractions were cultured in 96-well plates with CFSE-labeled conventional T cells at different effector-to-target ratios (1:1, 3:1, 5:1) in the presence of 10µg/ml PHA (Sigma-Aldrich, St. Louis, MO) for 72 h at 37°C.

Statistical analysis

Data are expressed as mean ± SD or median and inter quartile range, p25-p75. The Wilcoxon signed-rank test was used to determine pairwise differences. The Mann–Whitney U test and Student t-test were used to determine differences between groups. A probability value of equal to or less than 0.05 (p ≤ 0.05) was considered statistically significant.

Results

Increased number of FoxP3+ cells in hyaluronan-binding CD4+ T cell population in breast cancer patients

Although soluble HA failed to detect HA+ Tregs in freshly obtained PBMC, HA+ and HA- fractions of CD4+ cells were separated using immobilized high molecular weight HA. In healthy individuals, the fraction of CD4+ cells able to bind HA was small (6.5 ± 2.6%), but it was significantly increased in breast cancer patients (14.8 ± 8.3%, p=0.01) (Figure 1A). In healthy donors, the prevalence of CD4+CD25+ cells was significantly higher in HA+ fraction compared to HA- fraction (median 9.9%, IQR 6.6-12.6 and median 4.6%, IQR 3.3-8.7, correspondingly, p=0.03). There was no significant difference in the percentage of CD4+CD25+ cells between HA+ and HA- fractions in breast cancer patients (data not shown).

clinical-cellular-immunology-cancer-patients

Figure 1: The frequency of FoxP3+ Tregs is higher in HA+ fraction compared to HA- fraction of CD4+ cells isolated from both healthy donors and breast cancer patients. CD4+ cells obtained from peripheral blood were magnetically separated into HA+ and HA- fractions. The frequency of HA+ and HA- cells in CD4+ was determined (A). The percentage of CD4+CD25+FoxP3+ (B) and CD4+FoxP3+ (C) Tregs in HA+ and HA- fractions was analyzed; paired data and medians for each group of variables are represented.

While CD25 can be expressed on activated conventional T cells ”contaminating” CD4+CD25+ Treg cell subpopulation, transcription factor FoxP3 has a central role in Treg identification. As a highly characterized marker of Tregs, FoxP3 has been shown to be essential for their suppressive activity [19,20]. With gating on CD4+CD25+, the frequency of FoxP3+ cells was higher in HA+ compared to HA- both in healthy donors and breast cancer patients (Figure 1B). The difference in the number of CD4+CD25+FOXP3+ Tregs between HA+ and HA- fractions of breast cancer patients was not associated with stage of the disease. The percentage of CD4+CD25+FOXP3+ cells in HA- fraction was significantly increased in the breast cancer group compared to the healthy donor group (Figure 1B).

Previous studies have demonstrated that FoxP3 does not always correlate with CD25 expression [21,22]. We also observed that not all FoxP3+ cells expressed CD25. FACS analysis showed that the majority of CD4+FoxP3+ cells were able to bind HA in both groups. The percentage of HA+CD4+FoxP3+ cells was significantly elevated in breast cancer patients compared to healthy subjects (Figure 1C).

Higher number of CD39+ cells in circulating hyaluronan-binding CD4+CD25+ Treg subpopulation

Recent findings reveal an important role of CD4+CD25+CD39+ Tregs in cancer pathogenesis [23-26]. Considering the importance of this cell population, frequency of CD4+CD25+CD39+ Tregs in HA+ and HA- fractions of CD4+ was evaluated. No difference in CD39 expression between HA+ and HA- fractions either in healthy donors and breast cancer patients was obtained (Figure 2).

clinical-cellular-immunology-breast-cancer

Figure 2: Analysis of CD39 expression on HA+ and HA- Tregs of healthy donors and breast cancer patients. The percentage of CD4+CD25+CD39+ cells in freshly obtained HA+ and HA- fractions of CD4+ was determined. One representative result for each group (A) as well as summarized data (B) are shown.

However, CD39 expression was significantly increased on HA+CD4+CD25+ Tregs in breast cancer patients when compared to healthy donors. Notably, there was no difference in the number of CD39+ Tregs in HA- fractions between the healthy and breast cancer groups (Figure 2).

Increased level of CTLA-4 in hyaluronan-binding CD4+FoxP3+ Tregs

It is well known that the Treg cell population bears elevated expression of suppression markers. Therefore, we investigated whether the expression of such markers is associated with the ability to bind HA. HA+ and HA- fractions of CD4+ T cells were activated with TSI and analyzed for expression of suppression markers. After overnight activation, higher prevalence of CD4+CD25+FoxP3+ and CD4+FoxP3+ Tregs in HA+ fraction compared to HA- fraction was observed both in healthy individuals and breast cancer patients (Figure 3).

clinical-cellular-immunology-overnight-activation

Figure 3: After overnight activation with TSI, frequency of CD4+CD25+FoxP3+ and CD4+FoxP3+ Tregs is upregulated in HA+ when compared to HA- fraction of CD4+ cells in healthy donors and breast cancer patients. Representative results (A) and summarized data (B) are shown.

In healthy donors, the prevalence of CTLA-4+ cells was higher in HA+CD4+FoxP3+ compared to HA-CD4+FoxP3+ cell fraction: mean ±SD percentage 21.4 ± 6.2% and 9.9 ± 6.2% correspondingly, p=0.04. In contrast, there was no difference in the expression of CTLA-4 on HA+ and HA- fractions of CD4+CD25+FoxP3+ Tregs (Figure 4A). When we analyzed the levels of TGF-β, IL-35, LAP, IL-10 and GITR, no difference was seen between HA+ and HA- fractions, either in CD4+FoxP3+ or CD4+CD25+FoxP3+ subsets in breast cancer patients and healthy donors (Figure 4B).

clinical-cellular-immunology-bead-cell

Figure 4: Characteristics of HA+ and HA- Tregs of healthy individuals and breast cancer patients. CD4+ cells were purified by paramagnetic bead cell separation from peripheral blood of 7 healthy donors and 12 breast cancer patients, fractioned into HA+ and HA- and stimulated with TSI for 18 h. CD4+FoxP3+ and CD4+CD25+FoxP3+ Tregs were evaluated for the frequency of CTLA-4+ cells (A) and TGF-β+, IL-35+, LAP+, IL-10+, GITR+ cells (B).

Hyaluronan-binding CD4+CD25+ Tregs have enhanced suppressive activity

To determine whether the ability to bind HA is correlated with distinctive functional activity, freshly sorted CD4+CD25+ Tregs from five healthy donors were fractionated into HA+ and HA- cells and co-cultured with CD4+CD25- conventional T cells in an in vitro suppressor assay. HA+Tregs suppressed conventional T cell proliferation even at 1:1 suppressor to target ratio. While HA- Tregs were suppressive at 5:1 suppressor to target ratio, they lost the suppressive capacity as the ratio decreased (3:1 and 1:1) (Figure 4). These results suggest that two subsets of CD4+CD25+ Tregs are functionally distinct; HA+ Tregs are more suppressive.

clinical-cellular-immunology-flow-cytometry

Figure 5: Suppression of autologous CD4+CD25- conventional T cell proliferation by HA+ and HA- Treg cells. CFSE-labeled CD4+CD25- T cells were co-cultured at different ratios with HA+ and HA- fractions of CD4+CD25+ Tregs isolated from 5 healthy donors. After 72 h incubation with PHA, proliferation of CFSE labeled T cells was measured by flow cytometry. Percentage of proliferating T cells is shown.

Discussion

Hyaluronic acid is a negatively charged glycosaminoglycan that is abundantly present on endothelial cells and in extracellular matrix [27]. Its production is increased at the tumor-stroma interface, including breast cancer [14,17]. It has been demonstrated that interaction of HA with CD44 which is widely regarded as the major receptor for HA, mediates entry of T cells including immunosuppressive Tregs to target sites, as well as cell motility within these tissues [4]. Tregs are well known to play a crucial role in inhibiting anticancer defenses in the tumor microenvironment, resulting in tumor progressive growth and metastatic dissemination [28]. M. Firan et al. have shown that the ability to bind HA discriminates mouse Tregs with enhanced suppressive function and state of activation [8], but if these cells are present in human peripheral blood under normal physiological conditions and if this Treg subset is distinct in cancer is unclear.

To investigate the ability of peripheral blood Tregs to bind HA we used biotinylated HA, which was subsequently identified with streptavidin-PE. Tregs did not stain with soluble HA. On the contrary, we were able to obtain Tregs that actively bound immobilized HA. For this purpose, we developed a method of cell separation using a commercially available biotin-conjugated high molecular HA in combination with paramagnetic beads coated with anti-biotin Ab, which allowed us to isolate cells that were able to adhere to HA. Apparently, the large beads with coated HA were imitating extra-cellular matrix; therefore, the ability of these cells to interact with immobilized HA was more pronounced when compared to soluble HA.

Earlier studies have demonstrated that around 1% of naïve peripheral lymph node T lymphocytes bind soluble HA without any stimulation [8]. Ariel et al. have shown that adhesion of freshly purified human T cells to immobilized HA was always between 5 and 15% [6]. Here we show that the HA+ fraction of unstimulated CD4+ T cells is enriched with naturally occurring CD4+CD25+FoxP3+ Tregs. It should be noted that evaluated characteristics did not correlate with age of the individuals involved in the study. Thus, it appears that a large proportion of naturally arising Tregs, key players of the immune regulation, is capable of HA-mediated rolling. The higher prevalence of CD4+CD25+FoxP3+ Tregs in HA+ fraction was also observed after overnight activation. Moreover, freshly separated HA+CD4+CD25+ Tregs showed upregulated suppressor activity. The reason for persistence of HA+ nTregs in the peripheral blood is unclear; we suppose that it is likely to be important for the maintenance of immune tolerance to self-antigens. The obtained data are consistent with the results received by Levine et al. They report that just as in the conventional CD4+ T cell compartment, Tregs consist of CD44loCD62Lhi ‘naive-like’ and CD44hiCD62Llo ‘effector-like’ populations [29]. It is assumed that transition to the activated state is induced in the thymus and in the periphery upon TCR activation presumably by self-peptide [30]. The question if CD44hiCD62Llo Tregs are self-reactive, remains to be elucidated.

We observed that FoxP3 was expressed in a significant percentage of CD4+ T cells independently of CD25 expression. The importance of the CD4+FoxP3+ population in cancer and its negative impact on antitumor therapy has been demonstrated [31]. Analysis showed that a major portion of circulating CD4+FoxP3+ cells was able to bind HA, and their content was significantly higher in the peripheral blood of breast cancer patients. We assume that in cancer, more inducible Tregs that include a major subset of CD4+CD25-FoxP3+ cells rather than CD4+ CD25+FoxP3+ Tregs are potentially capable of HA-mediated adhesion and therefore more effective migration to tumor sites and maintenance of immune suppression [32].

In healthy donors, the ability to bind HA distinguished CD4+FoxP3+ Tregs with elevated expression of CTLA-4 after activation. The inhibitory mechanism of CTLA-4 is meditated through competition with the co-stimulatory molecule CD28, which, together with TCR, is necessary for T cell activation [33]. Massive aberrant activation and expansion of conventional T cells has been observed in transgenic mice lacking CTLA-4 expression in Tregs [34]. Apparently, under normal physiological conditions CTLA-4 on regulatory T cells dampens pathological naïve T cell activation and its elevated expression on HA+ Tregs implies a specific role of this subset in control of periphery tolerance in tissues.

Further analysis showed that CD39 is more highly expressed by HA+CD4+CD25+ Tregs in breast cancer than in healthy subjects. Catalytic inactivation and conversion of extracellular ATP by CD39 is one of the key anti-inflammatory mechanisms of Tregs [35]. CD39 drives the sequential hydrolysis of both adenosine triphosphate and adenosine diphosphate to adenosine monophosphate, while CD73 further hydrolyses it to adenosine, a nucleoside that exhibits direct immunosuppressive effects [36,37]. A significant increase of CD39 expression on Tregs in cancer patients, which is strongly associated with tumor progression, has been reported earlier [24,38]. Here, we demonstrate that particularly the frequency of CD4+CD25+CD39+ Tregs that are able to adhere to HA is increased in the periphery in breast cancer patients. It is unclear if the binding of HA promotes expression of CD39 on Tregs in breast cancer patients, but the obtained results suggest that accumulation of CD4+CD25+CD39+ Tregs in the tumor environment reported earlier may be meditated by HA [38-40].

Here we show a new approach for the isolation and investigation of Tregs, which can discriminate a new subset of Tregs potentially capable of adhesion to extracellular matrix. The approach allowed us to formulate several key notions concerning HA-binding Tregs. First, the normal immune system generates Tregs that intensively bind immobilized high molecular weight HA. The ability to bind HA discriminates Tregs with elevated suppressive activity and increased percentage of CTLA-4-expressing cells after activation in vitro. The reason for maintenance of such HA-binding Tregs under normal physiological conditions is not clear; they can represent an important subpopulation of previously activated circulating Tregs that may be involved into mechanisms of peripheral tolerance to auto-antigens. Second, the subset of HA-binding Tregs is increased in breast cancer and characterized by augmented expression of ectonucleotidase CD39. The increased proportion of HA-binding Tregs in circulation may reflect the upregulated activity of Tregs in breast cancer. Furthermore, HA-mediated rolling may be involved in infiltration of CD39+ Tregs into the tumor stroma and accumulation there. These findings, however, need to be proved in a larger cohort of patients in different phases of the disease to elucidate whether strategies targeting HA-mediated migration of Tregs may have therapeutic value.

Acknowledgements

This work was supported by the Grant #0222GF of the Ministry of Education and Science of Republic of Kazakhstan. The authors have no conflict of interest including any financial, personal or other. The authors thank Dr. G.K. Zakiryanova for contribution to the scientific research.

References

  1. Curotto de Lafaille M, Lafaille JJ (2009) Natural and Adaptive FoxP3+ Regulatory T Cells: More of the Same or a Division of Labor? J Immun 30: 626-635.
  2. Chatenoud L, Bach JF (2006) Adaptive human regulatory T cells: myth or reality? J Clin Invest 116: 2325-2327.
  3. DeGrendele HC, Estess P, Picker LJ, Siegelman MH (1996) CD44 and its ligand hyaluronate mediate rolling under physiologic flow: a novel lymphocyte/endothelial cell primary adhesion pathway. J Exp Med 183: 1119-1130.
  4. Baaten JG, Li C-R, Bradley LM (2010) Multifaceted regulation of T cells by CD44. Commun Integr Biol 3: 508-512.
  5. Liu D, Liu T, Li R, Sy MS (1998) Mechanisms regulating the binding activity of CD44 to hyaluronic acid. Front Biosci 1: 631-636.
  6. Ariel A, Lider O, Brill A, Cahalon L, Savion N, et al. (2000) Induction of interactions between CD44 and hyaluronic acid by a short exposure of human T cells to diverse pro-inflammatory mediators. Immunol 100: 345–351.
  7. Petrey A, Motte C (2014) Hyaluronan, a crucial regulator of inflammation. Front in Immunol 5: 1-13.
  8. Firan M, Dhillon S, Estess P, Siegelman MH (2006) Suppressor activity and potency among regulatory T cells is discriminated by functionally active CD44. Blood 107: 619-627.
  9. Bollyky PL, Falk BA, Long SA, Preisinger A, Braun KR, et al. (2009) CD44 costimulation promotes FoxP3+ regulatory T cell persistence and function via production of IL-2, IL-10, and TGF-β. J Immunol 183:2232-2241.
  10. Toole BP (2004) Hyaluronan: From extracellular glue to pericellular cue. Nat Rev Cancer 4: 528-539.
  11. Hopwood JJ, Dorfman A (1977) Glycosaminoglycan synthesis by cultured human skin fibroblasts after transformation with simian virus 40. J BiolChem 252: 4777-4785.
  12. Iijima J, Konno K, Itano N (2011) Inflammatory Alterations of the Extracellular Matrix in the Tumor Microenvironment. Cancers 3: 3189-3205.
  13. Takeuchi J, Sobue M, Sato E, Shamoto M, Miura K, et al. (1976) Variation in glycosaminoglycan components of breast tumors. Cancer Res 36: 2133-2139.
  14. Bertrand P, Girard N, Delpech B, Duval C, d‘Anjou J, et al. (1992) Hyaluronan (hyaluronic acid) and hyaluronectin in the extracellular matrix of human breast carcinomas: comparison between invasive and non-invasive areas. Int J Cancer 52: 1–6.
  15. Ponting J, Kumar S, Pye D (1993) Co-localisation of hyaluronan and hyaluronectin in normal and neoplastic breast tissues. Int J Oncol 2: 889-893.
  16. Tammia RH, Kulttia A, Kosmab V-M, Pirinenc R, Auvinenc P, (2008) Hyaluronan in human tumors: Pathobiological and prognostic messages from cell-associated and stromal hyaluronan. Sem in Cancer Biol 18:288-295.
  17. de la Torre M, Wells AF, Bergh J, Lindgren A (1993) Localization of hyaluronan in normal breast tissue, radial scar, and tubular breast carcinoma. Hum Pathol 24: 1294-1297.
  18. Auvinen P, Parkkinen J, Johansson R, Ågren U, Tammi R, et al (1997) Expression of hyaluronan in benign and malignant breast lesions. Int J Cancer 74: 477-481.
  19. Hori S, Nomura T, Sakaguchi S (2003) Control of regulatory T cell development by the transcription factor FoxP3. Science 299: 1057–1061.
  20. Fontenot JD, Gavin MA, Rudensky AY (2003) FoxP3 programs the development and function of CD4+CD25+ regulatory T cells. Nature Immunol 4: 330–336.
  21. Fontenot JD, Rasmussen JP, Williams LM, Dooley JL, Farr AG (2005) Regulatory T cell lineage specification by the forkhead transcription factor FoxP3. Immunity 22: 329–341.
  22. Liu W, Putnam AL, Xu-yu Z, Szot GL, Lee MR, et al. (2006) CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4+ T reg cells. J Exp Med 203: 1701–1711.
  23. Mandapathil M, Hilldorfer B, Szczepanski MJ, Czystowska M, Szajnik M, et al. (2010) Generation and accumulation of immunosuppressive adenosine by human CD4+CD25highFOXP3+ regulatory T cells (Treg). J BiolChemistr 285: 7176-7186.
  24. Schuler PJ, Schilling B, Harasymczuk M, Hoffmann TK, Johnson J, et al. (2012) Phenotypic and functional characteristics of CD4+CD39+ FOXP3+ and CD4+CD39+FOXP3neg T-cell subsets in cancer patients. Eur J of Immun 42: 1876-1885.
  25. Deagilo S, Dwyer KM, Gao W, Friedman D, Usheva A, et al (2007) Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression. J Exp Med 204: 1257-1265.
  26. Ye ZJ, Zhou Q, Zhang JC, Li X, Wu C, et al. (2011) CD39+ regulatory T cells suppress generation and differentiation of Th17 cells in human malignant pleural effusion via a LAP-dependent mechanism. Respir Res 12: 2-10.
  27. Entwistle J, Hall CL, Turley EA. (1996) HA receptors: regulators of signaling to the cytoskeleton. J Cell Biochem 61: 569–577.
  28. Nishikawa H, Sakaguchi S (2010) Regulatory T cells in tumor immunity. Int J Cancer 127: 759-767.
  29. Levine AG, Arvey A, Jin W, Rudensky AY (2014) Continuous requirement for the TCR in regulatory T cell function. Nat Immunol 15: 1070–1078.
  30. Zhu J, Shevach EM (2014) TCR signaling fuels Treg cell suppressor function. Nat Immunol 11: 1002-1003.
  31. Yao X, Ahmadzadeh M, Lu YC, Liewehr DJ, Dudley ME, et al.(2012) Levels of peripheral CD4(+)FoxP3(+) regulatory T cells are negatively associated with clinical response to adoptive immunotherapy of human cancer. Blood 119: 5688-5696.
  32. Wan YY, Flavell RA (2005) Identifying FoxP3-expressing suppressor T cells with a bicistronic reporter. ProcNatlAcadSci 102: 5126–5131.
  33. McCoy KD, Le Gros G (1999) The role of CTLA-4 in the regulation of T cell immune response. Immun and Cell Biol 77: 1–10.
  34. Jain N, Nguyen H, Chambers C, Kang J (2010) Dual function of CTLA-4 in regulatory T cells and conventional T cells to prevent multiorgan autoimmunity. ProcNatlAcadSci U S A 107: 1524-1528.
  35. Borsellino G, Kleinewietfeld M, Di Mitri D, Sternjak A, Diamantini A, et al. (2007) Expression of ectonucleotidase CD39 by FoxP3+ Treg cells: hydrolysis of extracellular ATP and immune suppression. Blood 110: 1225-1232.
  36. Deaglio S, Dwyer KM, Gao W, Friedman D, Usheva A, et al. (2007) Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression. J Exp Med 204: 1257-1265.
  37. Kobie JJ, Shah PR., Yang L, Rebhahn JA, Fowell DJ (2006) T regulatory and primed uncommitted CD4 T cells express CD73, which suppresses effector CD4 T cells by converting 5′-adenosine monophosphate to adenosine. J Immunol 177: 6780–6786.
  38. Mandapathil M, Szczepanski MJ, Szajnik M, Ren J, Lenzner DE (2009) Increased ectonucleotidase expression and activity in regulatory T cells of patients with head and neck cancer. Clin Cancer Res15: 6348-6357.
  39. Hilchey SP, Kobie JJ, Cochran MR, Secor-Socha S, Wang JC, et al (2009) Human follicular lymphoma CD39+-infiltrating T cells contribute to adenosine-mediated T cell hyporesponsiveness. J Immunol 183: 6157-6166.
  40. Jie H-B, Gildener-Leapman N, Li J, Srivastava RM, Gibson SP, et al. (2013) Intratumoral regulatory T cells upregulate immunosuppressive molecules in head and neck cancer patients. Brit J of Cancer 109: 2629-2635.
Citation: Perfilyeva Y, Ostapchuk Y, Cetin EA, Yilmaz A, Deniz G, et al. (2015) Hyaluronan-Binding T Regulatory Cells in Peripheral Blood of Breast Cancer Patients. J Clin Cell Immunol 6:286.

Copyright: © 2015 Perfilyeva Y, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top