Open Access

Cytokine-induced alterations of BAMBI mediate the reciprocal regulation of human Th17/Treg cells in response to cigarette smoke extract

  • Authors:
    • Hong‑Ju Liu
    • Gang Chen
    • Long Chen
    • Mei Zhou
    • Xian‑Zhi Xiong
    • Zhao‑Ji Meng
    • Sheng‑Wen Sun
    • Xiao‑Nan Tao
  • View Affiliations

  • Published online on: October 9, 2018     https://doi.org/10.3892/ijmm.2018.3919
  • Pages: 3404-3414
  • Copyright: © Liu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

In CD4+ T helper (Th) cells, transforming growth factor β (TGF‑β) is indispensable for the induction of both regulatory T (Treg) and interleukin‑17‑producing effector T helper (Th17) cells. Although BMP and activin membrane‑bound inhibitor (BAMBI) is part of a rheostat‑like mechanism for the regulation of TGF‑β signalling and autoimmune arthritis in mouse models, the underlying activity of BAMBI on the human Th17/Treg cell axis, particularly during exposure to cigarette smoke, remains to be elucidated. The present study aimed to further characterize BAMBI expression in human CD4+ cells, as well as immune imbalance during activation and cigarette smoke exposure. Results from the present study indicated that exposure to cigarette smoke extract partially suppressed Treg differentiation and promoted Th17 cell generation under stimulation by anti‑CD3/28 antibodies and TGF‑β1. Additionally, exposure to cigarette smoke induced an inhibition of phosphorylated‑Smad2/Smad3, which may have arisen from a concomitant enhancement of BAMBI expression. In conclusion, human BAMBI may function as a molecular switch to control TGF‑β signalling strength and the Th17/Treg cell balance, which may be used not only as a biomarker but also as a target of new treatment strategies for maintaining immune tolerance and for the treatment of smoking‑induced immune disorders.

Introduction

Many autoimmune diseases are caused by autoreactive T helper (Th)1 clones, and dysregulated Th1/Th2 responses have long been associated with the induction and regulation of autoimmunity (1). However, the finding that a lack of interferon γ makes otherwise experimental autoimmune encephalomyelitis mice susceptible to disease (2) raised the question of whether another T cell subset, other than Th1, may be required for the induction of autoimmune diseases. Subsequently studies identified an independent subset of interleukin (IL)‑17‑producing effector T helper (Th17) cells, which are involved in the pathogenesis of numerous experimental autoimmune diseases and human inflammatory states (3). By contrast, regulatory T cells (Tregs) are a subset of CD4+ T cells that express forkhead box P3 (FOXP3) and are crucial in controlling inflammation and maintaining self-tolerance (4).

Transforming growth factor (TGF)-β1 has been recognized as a link between Treg and Th17 cell subsets. Th17 cells are reciprocally related to Tregs given that TGF-β1 induces differentiation of naive T cells into FOXP3+ Tregs, whereas IL-6 inhibits the TGF-β1-induced expression of FOXP3 and ultimately induces the generation of Th17 cells (3). By contrast, high concentrations of TGF-β1 prevent the induction and effector functions of Th17 cells by inducing FOXP3 (3). However, numerous questions remain concerning the integration effects of TGF-β signals and inflammatory stimuli on the plasticity between Tregs and Th17 cells. Notably, BMP and activin membrane-bound inhibitor (BAMBI) was identified as a TGF‑β1 rheostat that controls Th17/Treg cell differentiation and the development of autoimmune arthritis in mice (5). BAMBI is structurally similar to TGF-β type I receptors (TGF-βRIs) but lacks an intracellular kinase domain, causing it to antagonize TGF-β family signalling (6). Although BAMBI has been shown to serve roles in human lung inflammation associated with chronic obstructive pulmonary disease (COPD) and bacterial infection (7), the precise expression and activity of BAMBI in human immune cells, especially in the Th17/Treg cell paradigm, remains to be determined.

Although smoking has been well established as a central factor in many pathological conditions, such as neoplasms and lung and cardiovascular diseases, little is known about the processes and factors involved in changing immune arms in response to cigarette smoke (8). Although tobacco smoke was demonstrated to stimulate the production of pro‑inflammatory cytokines (such as tumour necrosis factor α, IL-1 and IL-6) and to reduce anti‑inflammatory cytokine levels (such as IL‑10), local homeostasis and cytokine networks, as well as genetic predisposition, should be taken into account when analysing the influence of cigarette smoke (8). Our previous studies have demonstrated that cigarette smoking disrupts the survival of CD4+CD8+ Tregs in COPD, partially through muscarinic receptor-dependent mechanisms (9,10). However, the pathological processes and pathways underlying the Th17/Treg cell imbalance are still unknown.

Our previous study also demonstrated that an impaired TGF-β/BAMBI pathway promotes inflammation leading to Th17/Treg imbalance, which is a new mechanism in smokers who develop COPD (11). The present study aimed to further characterize BAMBI expression in human CD4+ cells and immune imbalance during activation and cigarette smoke exposure. These data suggested that cigarette smoke may disturb TGF-β-dependent BAMBI expression in human TCR-stimulated CD4+ T cells, which, in turn, may cause the dysfunction of Smad2/Smad3 phosphorylation following cigarette smoke exposure. The results indicated that BAMBI was functionally expressed in activated CD4+ T cells and may act as a molecular switch to control TGF-β signalling and the Th17/Treg cell balance. The TGF-β1/BAMBI pathway in T cells may serve a role in immune dysfunction associated with cigarette smoke.

Materials and methods

Cell isolation

This study was conducted between January 2016 and May 2017. Blood samples (20–30 ml) were obtained from 25 healthy adults (age, 25–55 years; 15 male, 10 female) and immediately placed on ice. Peripheral blood mononuclear cells (PBMCs) were isolated from buffy coats of blood samples. All experiments were performed in accordance with relevant manufacturer’s protocols. Naive CD4+ T cells were isolated from PBMCs by MACS, based on negative selection using the naive CD4+ T cell isolation kit II (Miltenyi Biotec GmbH, Bergisch Gladbach, Germany), according to the manufacturer’s instructions. The purity of CD4+CD45RA+ T cells was >95%, as measured by flow cytometry. This study was approved by the Ethics Committee of Union Hospital, Tongji Medical School, Huazhong University of Science and Technology (Wuhan, China); written informed consent was received from each donor.

Cigarette smoke extract (CSE) preparation

Aqueous CSE was prepared according to a modification of our previously published method (10). Briefly, the smoke from one cigarette (Huanghelou, Wuhan, China) was bubbled slowly into a tube containing 2.5 ml of sterile RPMI-1640 medium (Beijing Suolaibao Biotechnology Co., Ltd., Beijing, China). The resulting solution was further filtered through 0.22 μm filters and was defined as 100% CSE. The CSE was prepared fresh, within 30 min prior to each experiment.

Cell viability assay

Purified naive CD4+ T cells (1×105 cells/well) were cultured and incubated at 37°C in 200 μl of complete medium containing 10% heat‑inactivated (56°C for 30 min) foetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) and 50 U/ml IL-2 (PeproTech, Inc., Rocky Hill, NJ, USA) in 96-well plates and stimulated with plate-bound anti-CD3 (monoclonal, OKT3; 5 μg/ml) and anti‑CD28 (5 μg/ml) antibodies (both purchased from eBioscience; Thermo Fisher Scientific, Inc.). Cells were stimulated for 5 days in the presence of serial dilutions of CSE at 0, 0.002, 0.02 and 0.2%. Cell viability was tested by Cell Counting Kit-8 (Dojindo Molecular Technologies, Inc., Kumamoto, Japan), according to the manufacturer’s protocol. The safe concentrations of CSE at 0.002 and 0.02% used in subsequent experiments were chosen following a cytotoxicity assay (Fig. 1).

Development of naive and differentiating T cells in CSE‑conditioned medium

To investigate the effects of cigarette smoke on the development of naive CD4+ T cells, purified cells (5×105 cells/well) were incubated at 37°C in 1 ml complete medium containing FBS and IL-2, as aforementioned, in 48-well plates with plate-bound anti-CD3/CD28, in the absence or presence of CSE (0.002 and 0.02%). In parallel experiments, naive T cells were cultured in vitro under Treg-polarizing conditions (2 ng/ml TGF-β1) or Th17 cell-polarizing conditions (2 ng/ml TGF-β1 and 30 ng/ml IL-6; or 10 ng/ml IL-1β, 30 ng/ml IL-6 and 50 ng/ml IL-23), combined with or without CSE at the initiation of culture. Recombinant human TGF-β1, IL-6, IL-1β and IL‑23 were purchased from PeproTech, Inc. To confirm that the TGF-β1 produced and activated in T cell receptor (TCR)-stimulated cells was indeed responsible for BAMBI expression, a purified anti‑TGF‑β antibody (500 ng/ml; clone 19D8; BioLegend, Inc., San Diego, CA, USA) that is able to block human TGF-β1 activity was included in the culture. The involvement of Smad3 was determined by treating cells with 1 μM of the Smad3‑specific inhibitor SIS3 (Sigma‑Aldrich; Merck KGaA, Darmstadt, Germany), which caused potent and selective inhibition of Smad3 function (12). After 5 days incubation, the expression of T cell subsets and related molecules was determined by flow cytometry.

Flow cytometry

The expression of T cells markers were analysed by flow cytometry as described previously (10); surface or intracellular proteins were stained with fluorescein isothiocy-anate-conjugated anti-CD4 (Clone RPA-T4; BD Biosciences, San Jose, CA, USA), phycoerythrin (PE)-cyanine 7-conjugated anti-CD25 (Clone M-A251; BD Biosciences), PE-conjugated anti-FOXP3 (Clone 236A/E7; eBioscience; Thermo Fisher Scientific, Inc.), eFluor660-conjugated anti-IL-17A (Clone eBio64DEC17; eBioscience; Thermo Fisher Scientific, Inc.), Alexa Fluor (AF)-647-conjugated anti-BAMBI (Polyclonal; BIOSS, Beijing, China) and AF647-conjugated anti-Smad2 (pS465/pS467)/Smad3 (pS423/pS425) (Clone O72-670; BD Biosciences) antibodies. Intracellular staining for FOXP3, IL-17 and Smad2/Smad3 was performed following fixation and permeabilization using the FOXP3 Staining Buffer Set (eBioscience; Thermo Fisher Scientific, Inc.), as described previously (10). Isotype controls were used to exclude non‑specific staining and to identify the gates for negative/positive population selection. To identify Th17 cells, 50 ng/ml phorbol 12-myristate 13-acetate (Sigma-Aldrich; Merck KGaA), 1 μg/ml ionomycin (Sigma-Aldrich; Merck KGaA) and 1 μl/ml GolgiStop (BD Biosciences) were added to the culture medium on day 5 for 5 h. To measure intracellular levels of phosphorylated (p)-Smad2/Smad3, cells were treated with 2 ng/ml TGF-β1 for 30 min at 37°C on day 5 prior to harvesting. Flow cytometry was performed using the BD LSRFortessa and analysed with FACSDiva Software v8 (BD Biosciences) and FlowJo v10 software (Tree Star, Inc., Ashland, OR, USA).

Statistics

Data are expressed as the mean ± standard error of the mean. Statistical analysis was performed by one-way ANOVA followed by Dunnett’s multiple comparisons tests. Data analysis was conducted with GraphPad Prism v7.0 (GraphPad Software, Inc., La Jolla, CA, USA). P<0.05 was considered to indicate a statistically significant difference. No statistical method was used to predetermine sample size.

Results

Treg differentiation upon CSE exposure

To better understand the induction of Tregs, the expression levels of CD25 and FOXP3 were determined in naive CD4+ T cells stimulated with anti-CD3/28 antibodies and TGF-β1, in combination with or without the Smad3‑specific inhibitor SIS3. According to classical in vitro Treg-polarizing conditions (with anti-CD3/28 antibodies in the presence of TGF-β1) (13,14), high levels of CD25+FOXP3+ Tregs were induced successfully during differentiation; whereas this induction was blocked by SIS3 treatment (Fig. 2).

To determine whether the stimulation of cigarette smoke was associated with a change in Treg induction, CSE was added to CD4+ T cell cultures at different non-cytotoxic concentrations (0.002 and 0.02%; Fig. 1). Exposure to CSE alone did not induce naive CD4+ T cells to become CD25+FOXP3+ suppressor cells (15). Under classical Treg-polarizing conditions, however, CSE treatment notably reduced the differentiation rate of Tregs (Fig. 2).

CD25 expression is one of the activation markers of T cells. During Treg cell differentiation, a high induction of CD25 was also observed in CD4+ T cells following activation with anti-CD3/28 antibodies in the presence of TGF-β1 (Fig. 2). Similar to the observed trend in Treg generation, CD25 induction was inhibited by SIS3 and 0.02% CSE treatment (Fig. 2).

CSE exposure in Th17 cell differentiation

Classical differentiation of pro-inflammatory Th17 cells was also examined. In naive CD4+ T cells incubated in the presence of TGF-β1 + IL-6 (the first protocol), Th17 cells were successfully detected (Fig. 3). Notably, this induction was further enhanced in the presence of SIS3, which indicated that weakened Smad3 signalling may act as a regulator of Th17 cell skewing and Treg suppression. Subsequently, the underlying effects of cigarette smoking on Th17 cell induction were further examined. A previous study reported that the addition of CSE alone was unable to induce IL-17 expression in naive CD4+ T cells (15). Noatbly, under Th17 cell-polarizing conditions (the first protocol), CSE induced the differentiation of Th17 cells (Fig. 3).

In addition, the combination of IL-1β, IL-6 and IL-23 stimulation (the second protocol) was also able to initiate in vitro Th17 cell differentiation (Fig. 4). The combination of IL-6, IL-1β and IL-23 stimulation induced similar production levels of IL-17 between Th17 cells co-treated with or without SIS3 (Fig. 4). Notably, by the second protocol, CSE induced statistically stronger expression of Th17 cells compared with SIS3 treatment (Fig. 4).

TGF‑β/Smad3 signalling is attenuated by CSE exposure

As crucial downstream mediators in the TGF-β signalling pathway, the potential role of Smad2/Smad3 phosphorylation was further evaluated in TCR-stimulated naive cells. Results from flow cytometric analysis revealed a significant increase in Smad2/Smad3 phosphorylation TGF-β1-treated cells compared with expression in cells cultured in complete medium without stimulation (Fig. 5). Smad2/Smad3 phosphorylation was significantly inhibited in TCR-stimulated cells co‑treated with SIS3, which corroborated the specificity of the inhibitor.

To identify the potential for TGF-β/Smad signalling as a possible mechanism for the differentiation of naive T cells by CSE, the role of CSE in Smad2/Smad3 phosphorylation was evaluated. Cells co-treated with 0.02% CSE exhibited a significant reduction of Smad2/Smad3 phosphorylation compared with TGF-β-treated cells (Fig. 5), but to a lesser extent compared with SIS3 treatment. It is noteworthy that CSE treatment did not completely inhibit p-Smad2/Smad3 expression to that level of untreated cells.

BAMBI expression is increased during activation and CSE exposure

BAMBI acts as a TGF-β1 rheostat that regulates Th17/Treg cell differentiation and the development of COPD and autoimmune arthritis (5,9). Therefore, the potential expression characteristics of BAMBI under the various activation levels and the cytokine milieu conditions were investigated. Naive CD4+ T cells from peripheral blood express low or undetectable levels of BAMBI protein (5). Although treatment with TGF-β1 alone was able to induce mild BAMBI expression compared with untreated cells, anti-CD3/CD28 stimulation significantly increased BAMBI expression compared with untreated and TGF-β-treated cells (Fig. 6). In addition, the endogenous active TGF-β produced by the TCR-activated cells was necessary for sustained expression of BAMBI, since neutralization of TGF-β1 with anti-TGF-β antibody significantly reduced BAMBI induction in TCR‑stimulated cells (Fig. 6). Furthermore, it was observed that the addition of exogenous TGF-β1 further enhanced TCR-induced BAMBI expression compared with TCR-induction alone.

The potential effects of CSE on BAMBI expression were also investigated. Under stimulation by anti-CD3/28 antibodies in the presence of TGF-β1, CSE treatment induced a significant increase of BAMBI expression at 0.02% concentration (Fig. 6).

Discussion

Although previous studies have demonstrated key roles for tobacco smoke in immunity and inflammation, the cytokine milieu as well as genetic susceptibility should also be taken into account during analyses of effects of cigarette smoke (8). A previous study indicated that CSE treatment alone does not induce differentiation of naive CD4+ T cells (15). Under specific differentiation conditions, however, CSE substantially interfered with Treg development and concomitantly promoted the differentiation of Th17 cells. Results from the present study demonstrated that cigarette smoke-induced dysfunction in pro/anti‑inflammation may be associated with Smad-mediated canonical TGF-β signalling pathway and functionally expressed BAMBI levels. BAMBI functions as a molecular switch to regulate the balance between the development of Tregs and Th17 cells (5). These data may aid in guiding future clinical strategies for maintaining immune tolerance and for the treatment of smoking-induced immune disorders.

Although considerable progress has been made in understanding the mechanisms by which tobacco smoking exerts immunomodulatory functions in inflammation and autoimmunity (8), these mechanisms are still not fully understood. A balance of Tregs and Th17 cells is crucial for the treatment of smoking-induced pathological conditions. To determine the potential mechanisms for immune response following cigarette smoke exposure, the present study examined the role of CSE in freshly isolated naive CD4+ T cells from healthy donors. In line with a previous study (15), in vitro CSE treatment alone did not induce the development and differentiation of naive CD4+ T cells, which indicated that the development of Tregs and Th17 cells may be regulated by the cytokine milieu in addition to cigarette smoke alone.

To better understand the induction process of Treg development, the expression of CD25 and FOXP3 in CD4+ T cells induced by specific-Treg differentiation conditions, treatment with TGF-β1 were analysed. This induction was blocked by a specific Smad3 inhibitor, SIS3. Notably, under Treg‑polarizing conditions, the higher concentration of CSE remarkably inhibited Treg differentiation. In keeping with the observed trend in Treg generation, CD25 induction was also inhibited by SIS3 and by CSE during Treg cell differentiation. In this regard, it has been demonstrated previously that TGF-β1 collaborates with TCR signalling to upregulate CD25 expression and that CD25 gene regulatory region contains a Smad3 binding site that is required for TGF-β1-mediated upregulation (16). Thus, the effects of CSE on Treg differentiation depend not only on the CSE concentration, but also on the differentiation milieu and activation status.

Th17 cells were also successfully induced using two different in vitro Th17 cell differentiation protocols: One used TGF-β1 and IL-6, and the other used a combination of IL-1β, IL-6 and IL-23. Notably, low levels of endogenous TGF-β1 are still indispensable for the second Th17 cell-polarizing condition (17). Smad3 inhibition by SIS3 can promote Th17 cell differentiation in the presence of exogenous TGF-β1 (the first protocol) but not endogenous TGF‑β1 (the second protocol). However, CSE induced Th17 cell generation under both Th17 cell-polarizing conditions, and induced stronger Th17 expression compared with SIS3 treatment in the second protocol, which suggested that cigarette smoke may affect Th17 cells by unknown mechanisms (such as by reactive oxygen species) in addition to Smad signalling. Thus, although CSE itself does not induce the development of naive T cells, under Th subtype‑specific stimulating conditions CD4+ T cells had a decreased capacity to differentiate into Tregs, and a corresponding increased capacity to differentiate into Th17 cells upon CSE treatment.

A previous study reported that phosphorylation status induces opposing roles of Smad2/Smad3 as cofactors of signal transducer and activator of transcription 3 in Th17 differentiation (18); however, the molecular mechanisms by which context regulates phosphorylation status remain unclear. Flow cytometric analysis suggested that Smad2/Smad3 is phosphorylated following TCR stimulation with exogenous TGF-β1. The results also demonstrated that higher concentrations of CSE were able to partially inhibit TCR- and CD28-induced Smad2/Smad3 phosphorylation. However, CSE did not completely inhibit p-Smad2/Smad3 to the level of non-activated cells, which may partially be due to TCR-stimulated cells producing biologically active TGF-β1 that was responsible for Smad2/Smad3 phosphorylation (19). The effects of CSE on p-Smad2/Smad3 in T cells were inconsistent with previous studies in human bronchial epithelial cells (20) and foetal lung fibroblasts (21), which reported that exposure to CSE increased Smad3 phosphorylation. This discrepancy may be due to differences of CSE doses and cell types.

Although a recent study defined the cytokine TGF-β1 to serve a crucial role in governing whether CD4+ T cells differentiate into tolerogenic Tregs or pro‑inflammatory Th17 cells (22), the molecular mechanisms underlying the mutually exclusive differentiation of the two lineages remain incompletely understood. Using a BAMBI-knockout mouse model of collagen‑induced arthritis, BAMBI was identified as part of a rheostat-like mechanism for the control of TGF-β availability and signalling strength (5). However, the response of human T cells to polyclonal mitogens is more complicated compared with that of mouse T cells (23). For instance, human CD4+ cells activated without exogenous TGF-β1 will still express FOXP3 through endogenous TGF-β1 activation by reactive oxygen species (19). In this regard, although previous studies have demonstrated that BAMBI expression may be involved in human lung inflammation associated with COPD and bacterial infection (7), the present study is the first, to the best of our knowledge, to report the in vitro expression and role of BAMBI in human immune cells.

BAMBI is a 260-amino-acid-long transmembrane glyco-protein that is structurally related to TGF-βRI, but lacking the intracellular kinase domain (6). Although BAMBI expression may be upregulated by TGF-β1 (24,25), bone morphogenetic protein (BMP)-4 (26) and Wnt/β-catenin (27) in various cell types, these effects have not been observed in human preadipocytes (28), which indicated that the modulation of BAMBI expression is cell type-dependent. Consistent with previous reports using C57BL/6 mouse T cells (5), increased expression of BAMBI was observed in activated CD4+ T cells from human peripheral blood. Endogenous TGF-β1 produced by TCR-activated T cells may serve a crucial role in BAMBI induction in human CD4+CD25- T cells, and the addition of exogenous TGF-β1 further enhanced this effect. Taken together, these data demonstrated that endogenous and/or exogenous TGF-β1 may serve as a link in BAMBI induction in naive human CD4+ T cells following TCR stimulation. Additionally, we have provided the first evidence that CSE needs to cooperate with TGF-β1 to provoke induction of BAMBI expression. In summary, the effects of CSE on BAMBI induction and expression depend not only on cell activation status but also on local inflammatory stimuli, such as endogenous/exogenous TGF-β1 and the concentration of CSE.

Although BAMBI has been reported to negatively regulate TGF-β-family signalling mainly through Smad-dependent pathways in various cells lines (2931), it serves a positive role in triggering Smad2/Smad3 phosphorylation in human preadipocytes (28), which suggested cell type‑specific differences. Given its high expression in activated CD4+ T cells, the present study hypothesized that BAMBI also serves a crucial haemostatic role by controlling TGF-β signalling in human T cells. The results demonstrated that CSE caused a enhancement of BAMBI expression under stimulation by anti-CD3/28 antibodies and exogenous TGF-β1, which may result in a concomitant inhibition of p-Smad2/Smad3. Additionally, BAMBI upregulation by CSE may lead to the suppression of Treg differentiation and the promotion of Th17 generation in response to TGF-β1 stimulation, partially via Smad-dependent pathways. Thus, as in mouse cells, BAMBI may function as a crucial regulator of Th17/Treg plasticity through TGF-β/Smad-dependent pathways in human CD4+ T cells. It was proposed that at the steady state or in the absence of any inflammatory insult, TGF‑β1 produced in the immune system will facilitate the generation of Tregs, which keep effector T cells in check (Fig. 7). However, upon inflammation or CSE exposure, BAMBI overexpression by the activated immune system in susceptible individuals may suppress the generation of TGF-β-induced Tregs and evoke pro‑inflammatory responses dominated by Th17 cells.

BAMBI is an inhibitor or regulator of TGF-β superfamily members, including BMP and activin, in addition to TGF-β (6). The present study focussed solely on TGF-β, as activin-A or BMP-2/4 seem to have only synergistic effects on endogenous and/or exogenous TGF-β-induced Treg generation (32,33). However, the present study has several limitations that warrant mention; the focus was primarily on TGF-superfamily signalling without considering the Wnt/β-catenin pathway, although BAMBI also functions as a positive regulator of Wnt signalling (28,34), which further negatively modulates Treg function (35). In addition, further work is needed to demonstrate the supplemental role of non-Smad TGF-β signals by mitogen-activated protein kinases, including c-Jun N-terminal kinase, p38 and, in particular, extracellular signal-regulated kinase (ERK) (36), as previous studies have demonstrated that BAMBI is able to facilitate ERK1/2 phosphorylation (37) and ERK inactivation mediates the TGF-β1-induced expression of FOXP3 (38). Furthermore, functional studies such as BAMBI knockdown and/or overexpression in human T cells in vitro are warranted to clarify the mechanism by which CSE exerts its effects.

In conclusion, results from the present study confirmed that BAMBI may be able to regulate the balance between protective Tregs and pathogenic Th17 cells through a rheostat-like mechanism in the human immune system. CSE, together with TGF-β1, was demonstrated to effect BAMBI expression and reduce Smad3 phosphorylation, which may act as a master regulator of Th17 induction and Treg suppression. These results may be beneficial in improving our understanding of the reciprocal regulation of these two cell lineages, and may aid in the development of better approaches for the treatment of smoking‑associated inflammation or other T cell‑mediated autoimmune diseases.

Funding

The present study was supported by The National Natural Science Foundation of China (grant nos. 81370146, 81570032 and 81500031). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Availability of data and materials

The datasets used and/or analysed during the current study are available from the corresponding author on reasonable request.

Authors’ contributions

HJL and GC contributed to data analysis and interpretation. LC and MZ contributed to cell isolation and culture. XZX contributed to the design of the study. XZX and XNT were involved in volunteer recruitment. HJL, GC, ZJM and SWS wrote the manuscript and critically revised the paper. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The study was approved by the Ethics Committee of Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, and informed written consent was obtained from each subject.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Abbreviations:

BAMBI

BMP and activin membrane-bound inhibitor

COPD

chronic obstructive pulmonary disease

CSE

cigarette smoke extract

TGF-β

transforming growth factor β

Th

T helper cell

Treg

regulatory T cell

Acknowledgments

Not applicable.

References

1 

Nicholson LB and Kuchroo VK: Manipulation of the Th1/Th2 balance in autoimmune disease. Curr Opin Immunol. 8:837–842. 1996. View Article : Google Scholar : PubMed/NCBI

2 

Tran EH, Prince EN and Owens T: IFN-gamma shapes immune invasion of the central nervous system via regulation of chemokines. J Immunol. 164:2759–2768. 2000. View Article : Google Scholar : PubMed/NCBI

3 

Korn T, Bettelli E, Oukka M and Kuchroo VK: IL-17 and Th17 cells. Annu Rev Immunol. 27:485–517. 2009. View Article : Google Scholar : PubMed/NCBI

4 

Sakaguchi S, Yamaguchi T, Nomura T and Ono M: Regulatory T cells and immune tolerance. Cell. 133:775–787. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Postigo J, Iglesias M, Alvarez P, Jesús Augustin J, Buelta L, Merino J and Merino R: Bone morphogenetic protein and activin membrane-bound inhibitor, a transforming growth factor β rheostat that controls murine treg Cell/Th17 cell differentiation and the development of autoimmune arthritis by reducing interleukin-2 signaling. Arthritis Rheumatol. 68:1551–1562. 2016. View Article : Google Scholar

6 

Onichtchouk D, Chen YG, Dosch R, Gawantka V, Delius H, Massagué J and Niehrs C: Silencing of TGF-beta signalling by the pseudoreceptor BAMBI. Nature. 401:480–485. 1999. View Article : Google Scholar : PubMed/NCBI

7 

Dromann D, Rupp J, Rohmann K, Osbahr S, Ulmer AJ, Marwitz S, Röschmann K, Abdullah M, Schultz H, Vollmer E, et al: The TGF-beta-pseudoreceptor BAMBI is strongly expressed in COPD lungs and regulated by nontypeable haemophilus influenzae. Respir Res. 11:672010. View Article : Google Scholar : PubMed/NCBI

8 

Arnson Y, Shoenfeld Y and Amital H: Effects of tobacco smoke on immunity, inflammation and autoimmunity. J Autoimmun. 34:J258–J265. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Chen G, Zhou M, Chen L, Meng ZJ, Xiong XZ, Liu HJ, Xin JB and Zhang JC: Cigarette smoke disturbs the survival of CD8+ Tc/Tregs partially through muscarinic receptors-dependent mechanisms in chronic obstructive pulmonary disease. PLoS One. 11:e1472322016.

10 

Zhang MQ, Wan Y, Jin Y, Xin JB, Zhang JC, Xiong XZ, Chen L and Chen G: Cigarette smoking promotes inflammation in patients with COPD by affecting the polarization and survival of Th/Tregs through Up-regulation of muscarinic receptor 3 and 5 expression. PLoS One. 9:e1123502014. View Article : Google Scholar : PubMed/NCBI

11 

Zhang JC, Chen G, Chen L, Meng ZJ, Xiong XZ, Liu HJ, Jin Y, Tao XN, Wu JH and Sun SW: TGF-β/BAMBI pathway dysfunction contributes to peripheral Th17/Treg imbalance in chronic obstructive pulmonary disease. Sci Rep. 6:319112016. View Article : Google Scholar

12 

Jinnin M, Ihn H and Tamaki K: Characterization of SIS3, a novel specific inhibitor of Smad3, and its effect on transforming growth factor-beta1-induced extracellular matrix expression. Mol Pharmacol. 69:597–607. 2006. View Article : Google Scholar

13 

Sekiya T and Yoshimura A: In vitro Th differentiation protocol. Methods Mol Biol. 1344:183–191. 2016. View Article : Google Scholar

14 

Fantini MC, Dominitzki S, Rizzo A, Neurath MF and Becker C: In vitro generation of CD4+ CD25+ regulatory cells from murine naive T cells. Nat Protoc. 2:1789–1794. 2007. View Article : Google Scholar

15 

Garn H, Turowska A, Weissmann N and Baumgartl N: Effects of cigarette smoke extract-conditioned medium on T helper cell development and differentiation in vitro. Am J Respir Crit Care Med. 191:A27332015.

16 

Kim HP, Kim BG, Letterio J and Leonard WJ: Smad-dependent cooperative regulation of interleukin 2 receptor alpha chain gene expression by T cell receptor and transforming growth factor-beta. J Biol Chem. 280:34042–34047. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Chung Y, Chang SH, Martinez GJ, Yang XO, Nurieva R, Kang HS, Ma L, Watowich SS, Jetten AM, Tian Q, et al: Critical regulation of early Th17 cell differentiation by interleukin-1 signaling. Immunity. 30:576–587. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Yoon JH, Sudo K, Kuroda M, Kato M, Lee IK, Han JS, Nakae S, Imamura T, Kim J, Ju JH, et al: Phosphorylation status determines the opposing functions of Smad2/Smad3 as STAT3 cofactors in TH17 differentiation. Nat Commun. 6:76002015. View Article : Google Scholar : PubMed/NCBI

19 

Amarnath S, Dong L, Li J, Wu Y and Chen W: Endogenous TGF-beta activation by reactive oxygen species is key to Foxp3 induction in TCR-stimulated and HIV-1-infected human CD4+CD25- T cells. Retrovirology. 4:572007. View Article : Google Scholar

20 

Milara J, Peiro T, Serrano A and Cortijo J: Epithelial to mesenchymal transition is increased in patients with COPD and induced by cigarette smoke. Thorax. 68:410–420. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Farid M, Kanaji N, Nakanishi M, Gunji Y, Michalski J, Iwasawa S, Ikari J, Wang X, Basma H, Nelson AJ, et al: Smad3 mediates cigarette smoke extract (CSE) induction of VEGF release by human fetal lung fibroblasts. Toxicol Lett. 220:1261342013. View Article : Google Scholar : PubMed/NCBI

22 

Li MO and Flavell RA: TGF-beta: A master of all T cell trades. Cell. 134:392–404. 2008. View Article : Google Scholar : PubMed/NCBI

23 

Horwitz DA, Zheng SG, Wang J and Gray JD: Critical role of IL-2 and TGF-beta in generation, function and stabilization of Foxp3+CD4+ Treg. Eur J Immunol. 38:912–915. 2008. View Article : Google Scholar : PubMed/NCBI

24 

Sekiya T, Oda T, Matsuura K and Akiyama T: Transcriptional regulation of the TGF-beta pseudoreceptor BAMBI by TGF-beta signaling. Biochem Biophys Res Commun. 320:680–684. 2004. View Article : Google Scholar : PubMed/NCBI

25 

Xavier S, Gilbert V, Rastaldi MP, Krick S, Kollins D, Reddy A, Bottinger E, Cohen CD and Schlondorff D: BAMBI is expressed in endothelial cells and is regulated by lysosomal/autolysosomal degradation. PLoS One. 5:e129952010. View Article : Google Scholar : PubMed/NCBI

26 

Karaulanov E, Knochel W and Niehrs C: Transcriptional regulation of BMP4 synexpression in transgeni Xenopus. EMBO J. 23:844–856. 2004. View Article : Google Scholar : PubMed/NCBI

27 

Sekiya T, Adachi S, Kohu K, Yamada T, Higuchi O, Furukawa Y, Nakamura Y, Nakamura T, Tashiro K, Kuhara S, et al: Identification of BMP and activin membrane‑bound inhibitor (BAMBI), an inhibitor of transforming growth factor-beta signaling, as a target of the beta-catenin pathway in colorectal tumor cells. J Biol Chem. 279:6840–6846. 2004. View Article : Google Scholar

28 

Luo X, Hutley LJ, Webster JA, Kim YH, Liu DF, Newell FS, Widberg CH, Bachmann A, Turner N, Schmitz-Peiffer C, et al: Identification of BMP and activin membrane‑bound inhibitor (BAMBI) as a potent negative regulator of adipogenesis and modulator of autocrine/paracrine adipogenic factors. Diabetes. 61:124–136. 2012. View Article : Google Scholar

29 

Shangguan L, Ti X, Krause U, Hai B, Zhao Y, Yang Z and Liu F: Inhibition of TGF-/β/Smad signaling by BAMBI blocks differentiation of human mesenchymal stem cells to carcinoma‑associated fibroblasts and abolishes their protumor effects. Stem Cells. 30:2810–2819. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Yan X, Lin Z, Chen F, Zhao X, Chen H, Ning Y and Chen YG: Human BAMBI cooperates with Smad7 to inhibit transforming growth factor-beta signaling. J Biol Chem. 284:30097–30104. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Villar AV, García R, Llano M, Cobo M, Merino D, Lantero A, Tramullas M, Hurlé JM, Hurlé MA and Nistal JF: BAMBI (BMP and activin membrane-bound inhibitor) protects the murine heart from pressure-overload biomechanical stress by restraining TGF-β signaling. Biochim Biophys Acta. 1832.323–335. 2013.

32 

Huber S, Stahl FR, Schrader J, Lüth S, Presser K, Carambia A, Flavell RA, Werner S, Blessing M, Herkel J and Schramm C: Activin a promotes the TGF-beta-induced conversion of CD4+CD25- T cells into Foxp3+ induced regulatory T cells. J Immunol. 182:4633–4640. 2009. View Article : Google Scholar : PubMed/NCBI

33 

Lu L, Ma J, Wang X, Wang J, Zhang F, Yu J, He G, Xu B, Brand DD, Horwitz DA, et al: Synergistic effect of TGF-beta superfamily members on the induction of Foxp3+ Treg. Eur J Immunol. 40:142–152. 2010. View Article : Google Scholar :

34 

Lin Z, Gao C, Ning Y, He X, Wu W and Chen YG: The pseudo-receptor BMP and activin membrane-bound inhibitor positively modulates Wnt/beta-catenin signaling. J Biol Chem. 283:33053–33058. 2008. View Article : Google Scholar : PubMed/NCBI

35 

van Loosdregt J, Fleskens V, Tiemessen MM, Mokry M, van Boxtel R, Meerding J, Pals CE, Kurek D, Baert MR, Delemarre EM, et al: Canonical Wnt signaling negatively modulates regulatory T cell function. Immunity. 39:298–310. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Derynck R and Zhang YE: Smad-dependent and Smad- independent pathways in TGF-beta family signalling. Nature. 425:577–584. 2003. View Article : Google Scholar : PubMed/NCBI

37 

Mai Y, Zhang Z, Dong P, Yang H, Yang G and Sun S: BAMBI inhibits porcine preadipocyte differentiation by facilitating ERK1/2 phosphorylation. Sheng Wu Gong Cheng Xue Bao. 30:1531–1540. 2014.In Chinese.

38 

Luo X, Zhang Q, Liu V, Xia Z, Pothoven KL and Lee C: Cutting edge: TGF-beta-induced expression of Foxp3 in T cells is mediated through inactivation of ERK. J Immunol. 180:2757–2761. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2018
Volume 42 Issue 6

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liu HJ, Chen G, Chen L, Zhou M, Xiong XZ, Meng ZJ, Sun SW and Tao XN: Cytokine-induced alterations of BAMBI mediate the reciprocal regulation of human Th17/Treg cells in response to cigarette smoke extract. Int J Mol Med 42: 3404-3414, 2018
APA
Liu, H., Chen, G., Chen, L., Zhou, M., Xiong, X., Meng, Z. ... Tao, X. (2018). Cytokine-induced alterations of BAMBI mediate the reciprocal regulation of human Th17/Treg cells in response to cigarette smoke extract. International Journal of Molecular Medicine, 42, 3404-3414. https://doi.org/10.3892/ijmm.2018.3919
MLA
Liu, H., Chen, G., Chen, L., Zhou, M., Xiong, X., Meng, Z., Sun, S., Tao, X."Cytokine-induced alterations of BAMBI mediate the reciprocal regulation of human Th17/Treg cells in response to cigarette smoke extract". International Journal of Molecular Medicine 42.6 (2018): 3404-3414.
Chicago
Liu, H., Chen, G., Chen, L., Zhou, M., Xiong, X., Meng, Z., Sun, S., Tao, X."Cytokine-induced alterations of BAMBI mediate the reciprocal regulation of human Th17/Treg cells in response to cigarette smoke extract". International Journal of Molecular Medicine 42, no. 6 (2018): 3404-3414. https://doi.org/10.3892/ijmm.2018.3919