Induction of IL-17 production from human peripheral blood CD4+ cells by asbestos exposure

  • Authors:
    • Megumi Maeda
    • Ying Chen
    • Suni Lee
    • Naoko Kumagai-Takei
    • Kei Yoshitome
    • Hidenori Matsuzaki
    • Shoko Yamamoto
    • Tamayo Hatayama
    • Miho Ikeda
    • Yasumitsu Nishimura
    • Takemi Otsuki
  • View Affiliations

  • Published online on: May 9, 2017     https://doi.org/10.3892/ijo.2017.3991
  • Pages: 2024-2032
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

We have previously reported that chronic, recurrent and low-dose exposure to asbestos fibers causes a reduction in antitumor immunity. Investigation of natural killer (NK) cells using an in vitro cell line model and comprising in vitro activation using freshly isolated NK cells co-cultured with chrysotile fibers, as well as NK cells derived from asbestos-exposed patients with pleural plaque (PP) or malignant mesothelioma (MM), revealed decreased expression of NK cell activating receptors such as NKG2D, 2B4 and NKp46. An in vitro differentiation and clonal expansion model for CD8+ cytotoxic T lymphocytes (CTLs) showed reduced cytotoxicity with decreased levels of cytotoxic molecules such as granzyme B and perforin, as well as suppressed proliferation of CTLs. Additionally, analysis of T helper cells showed that surface CXCR3, chemokine receptor, and the productive potential of interferon (IFN)γ were reduced following asbestos exposure in an in vitro cell line model and in peripheral CD4+ cells of asbestos-exposed patients. Moreover, experiments revealed that asbestos exposure enhanced regulatory T cell (Treg) function. This study also focused on CXCR3 expression and the Th-17 cell fraction. Following activation with T-cell receptor and co-culture with various concentrations of chrysotile fibers using freshly isolated CD4+ surface CXCR3 positive and negative fractions, the intracellular expression of CXCR3, IFNγ and IL-17 remained unchanged when co-cultured with chrysotile. However, subsequent re-stimulation with phorbol 12-myristate 13-acetate (PMA) and ionomycin resulted in enhanced IL-17 production and expression, particularly in CD4+ surface CXCR3 positive cells. These results indicated that the balance and polarization between Treg and Th-17 fractions play an important role with respect to the immunological effects of asbestos and the associated reduction in antitumor immunity.

Introduction

Asbestos and silica are the most well-known causative, environmental and occupational substances that induce pneumoconiosis (13). The initial event following entry of these substances into the human body involves the action of immune competent cells such as alveolar macrophages to treat these foreign materials by activating the NOD-like receptor family, pyrin domain containing 3 (NLRP3: NALP3) inflammasome to produce interleukin (IL)-1β and attract fibroblasts (46).

Since the immunological effects of asbestos fibers have not been thoroughly investigated, we decided to study the various effects of asbestos on human immune competent cells. Investigation of natural killer (NK) cells using in vitro chronic exposure in an NK cell line and freshly isolated NK cells derived from healthy volunteers (HV) showed a reduction in NK cell activating receptors such as NKG2D and 2B4, as well as intracellular granzyme A, and decreased phosphorylation of ERK (7,8). In addition to these findings, studies of peripheral blood NK cells from asbestos-exposed patients with pleural plaque (PP) or malignant mesothelioma (MM) showed decreased expression of NKp46 activating receptor (79). Furthermore, the differentiation and proliferation of CD8+ cytotoxic T lymphocytes (CTLs) were also impaired by asbestos exposure. The cytotoxicity for allogeneic targets decreased in peripheral blood mononuclear cells (PBMCs) exposed to chrysotile B (CB) asbestos, but not in cells exposed to crocidolite (CR) asbestos, when compared with PBMCs without exposure during a mixed lymphocyte reaction (MLR) (10). Exposure to CB during the MLR resulted in suppression of an increase in granzyme B+ cells and interferon (IFN)γ positive cells. CB exposure also resulted in suppression of increases in CD45RO+ effector/memory cells and CD25+-activated cells in CD8+ lymphocytes, and a decrease in CD45RA+ cells. Furthermore, CB exposure suppressed the proliferation of CD8+ lymphocytes without yielding an increase in Annexin V+ apoptotic cells in CD8+ lymphocytes. Moreover, the production of IL-10, IFNγ and tumor necrosis factor (TNF)-α, but not IL-2, decreased in the presence of CB (1013).

Our research also established a model involving chronic and continuous exposure to asbestos for the investigation of CD4+ T helper (Th) cells. Transient and relatively high-dose exposure to asbestos caused apoptosis in a human T-cell leukemia virus type (HTLV)-1 immortalized human polyclonal T-cell line, MT-2, by the production of reactive oxygen spices (ROS). Activation of the pro-apoptotic MAK transduction signaling pathway such as p38 and JUN, and activation of the mitochondrial apoptotic pathway during continuous and relatively low-dose (in which the occurrence of apoptosis was <50%) exposure for more than one year resulted in MT-2 being more resistant to asbestos-induced apoptosis (14). Several independent sublines of MT-2 continuously exposed to asbestos showed upregulation of IL-10 and transforming growth factor (TGF)β, and activation of signal transducer and activator of transcription 3 (STAT3) and Bcl-2 (15). Additionally, these sublines showed alterations in cytoskeletal molecules such as β-actin and vimentin (16). The results obtained from these sublines continuously exposed to asbestos revealed reduced expression of C-X-C chemokine receptor type 3 (CXCR3) and a decreased potential for IFNγ production. These results were also observed in CD4+ cells derived from patients with PP or MM (17,18).

The overall findings concerning the immunological effects of asbestos on human immune competent cells indicated that chronic exposure to asbestos causes a reduction in antitumor immunity (1921). This suggests that asbestos-exposed individuals possess gradually reduced antitumor immunity and subsequent increased susceptibility to the onset of cancer. This may explain the long latency period observed in asbestos-exposed individuals before they develop malignant tumors following initial exposure to asbestos, and this sensitivity may lead to the development of lung cancer and MM, in addition to other malignancies such as those of the larynx, gastrointestinal tract and bladder (22,23).

The role of Th17 cells in the development of tumors has been investigated from various viewpoints, and consideration of the part played by Th17 cells in carcinogenesis has spawned various paradigms (2427). It appears that Th17 cells play a complex and controversial role in tumor immunity. A study of the effects of asbestos on Th17 cells in a murine model using erionite, which has similar chemical and physical properties to asbestos, demonstrated induced production of IL-17 (28). Similarly, a study utilizing amphibole asbestos, but not chrysotile asbestos, demonstrated induced production of IL-17 in a murine model (29). Both of these studies indicated that the recorded upregulation of IL-17 was related to the production of autoantibodies and assumed that autoimmune disease might be caused by asbestos exposure (2931). However, the precise manner by which asbestos exposure affects IL-17 production and alters Th cell-type commensuration is unclear. At the very least, we found that exposure of T cells to asbestos causes a reduction in CXCR3 and IFNγ under the experimental conditions employed and in asbestos-exposed patients with PP or MM (17,18). These findings were thought to be very important in assessing antitumor immunity in Th cells since CXCR3 expressing Th cells recruit IFNγ-producing antitumor Th into the area surrounding the tumor. However, the activity of both processes might be reduced in asbestos-exposed patients. Taken together, it might be of value to determine whether asbestos can induce inhibition of the cellular features of Th17 cells. The issue therefore is to determine the manner by which asbestos affects Th17 cells. In an effort to address this matter, freshly isolated human peripheral CD4+ cells were activated in vitro with chrysotile asbestos, since the use of chrysotile in industrial and commercial products is higher than that of amphibole asbestos, and the immunological effects were found to be similar for both forms of asbestos as we previously reported (32). We then investigated IL-17 expression and production in relation to CXCR3 expression. Since a reduction in CXCR3 expression was observed following asbestos exposure, the cellular roles of IL-17 production and expression in CXCR3 positive and negative cells were examined.

Materials and methods

Peripheral blood cell preparation and intracellular staining

Peripheral blood T cells were prepared from three HV. Peripheral blood was drawn from the vein with the aid of heparin. PBMCs were isolated using the Ficoll-Hypaque method. Cells were then stained with anti-CD4 monoclonal antibody (mAb) and anti-CXCR3 mAb. Both antibodies were purchased from BD Biosciences, Minneapolis, MN, USA. PBMCs stained by both antibodies were divided into two fractions comprising CD4+ and surface CXCR3 negative (CD4+sCXCR3) or positive (CD4+sCXCR3+) cells (Fig. 1A). Cells in two fractions (CD4+sCXCR3 or CD4+sCXCR3+, 1×106 cells) were stimulated with 1 µg/ml of plate-coated anti-CD3 mAb and 1 µg/ml of soluble anti-CD28 mAb with 10 µg/ml of IL-2 for four weeks (Fig. 1B). These cultures received 0, 10, 25 or 50 µg/ml of chrysotile asbestos.

The chrysotile asbestos was removed following cultivation using the Ficoll-Hypaque method and intracellular expression of CXCR3, IFNγ and IL-17 was analyzed using mAbs for these molecules (BD Biosciences). Intracellular staining was performed using a Cytofix/Cytoperm kit (BD Biosciences) in accordance with the manufacturer's instructions. Cells were stained with individual mAbs for 30 min and then subjected to FACSCalibur flow cytometry.

Additionally, cells cultured with various concentrations of chrysotile were re-activated using phorbol 12-myristate 13-acetate (PMA) and ionomycin (IM) for 4 h. Human PBMCs were obtained for this study from H.V. who provided written informed consent according to guidelines established by the ethics committee of the Kawasaki Medical School, Kurashiki, Japan.

CBA assay of culture supernatants

As shown in Fig. 1B, activated cells from two fractions (CD4+sCXCR3 and CD4+sCXCR3+) co-cultured for four weeks with various concentrations of chrysotile were re-stimulated by PMA and ionomycin for 4 h. These supernatants were subjected to CBA assays (BD Biosciences) to measure the concentration of IL-17 and IFNγ.

Real-time RT-PCR

Total RNA was extracted from cells re-stimulated by PMA and ionomycin and derived from two fractions (CD4+sCXCR3+ and CD4+sCXCR3) activated for four weeks and co-cultured with various concentrations of chrysotile using RNAzol. Following synthesis of the first strand of cDNA, real-time RT-PCR was performed using the SYBER Green method (Takara, Shiga, Japan) with the Mx3000P QPCR System (Agilent Technologies, Inc., Santa Clara, CA), as previously described (Maeda et al, 16–18,32). The following primers were used; for CXCR3: forward (F), 5′-ACACCTTCCTGCTCCACCTA-3′, reverse (R), 5′-GTTCAGGTAGCGGTCAAAGC-3′); for IL-17: F, 5′-ACCAATCCCAAAAGGTCCTC-3′, R, 5′-CCCACGGA CACCAGTATCTT-3′; for T-bet: F, 5′-AGGTGTCGGGG AAACTGAG-3′, R, 5′-ACCACGTCCACAAACATCCT-3′; and for RORγt: F, 5′-AAATCTGTGGGGACAAGTGG-3′, R, 5′-TCCCTCTGCTTCTTGGACAT-3′.

Statistical analysis

Statistical analyses were performed using SPSS version 21 (IBM Japan, Tokyo, Japan). Group comparisons in this study were performed as follows. The first comprised a comparison of intracellular CXCR3, IFNγ or IL-17 expression in CD4+CXCR3+ or CD4+CXCR3 fractions derived from cells cultured in the absence or presence of various concentrations of chrysotile fibers. The next involved a comparison of IFNγ and IL-17 levels in supernatants of CD4+CXCR3+ or CD4+CXCR3 fractions from cells cultured in the absence or presence of various concentrations of chrysotile fibers. Additionally, real-time RT-PCR was employed to examine mRNA expression levels of T-bet, IFNγ, RORγT or IL-17 of CD4+CXCR3+ or CD4+CXCR3 fractions from cells cultured in the absence or presence of various concentrations of chrysotile fibers. All statistical analyses were subjected to the one-way ANOVA test and subsequent post-hoc comparisons were made using the Mann-Whitney U test.

Results

Intracellular CXCR3 expression following cultivation with chrysotile

CD4+sCXCR3 and CD4+sCXCR3+ cells were cultured in the presence of various concentrations of chrysotile (0, 10, 25 or 50 µg/ml) for four weeks with T-cell receptor (TCR) stimulation and IL-2. Following this activation period, intracellular CXCR3 expression was analyzed by flow cytometry (Fig. 2). The intracellular expression of CXCR3 was significantly higher in CD4+sCXCR3+ cells compared with CD4+sCXCR3 cells in both R1 (totally positive cell fraction) and R2 (highly positive fraction) regions. The results showed that although intracellular CXCR3 expression remained unaltered in cells derived from CD4+sCXCR3+ fractions, the presence of chrysotile tended to reduce the highly positive intracellular expression of CXCR3 in cells that were initially CD4+sCXCR3.

Intracellular IFNγ expression following cultivation with chrysotile

Levels of IFNγ were examined in CD4+sCXCR3 and CD4+sCXCR3+ fractions derived from cells cultured with various concentrations of chrysotile (Fig. 3). The intracellular expression of IFNγ was significantly higher in CD4+sCXCR3+ cells compared with CD4+sCXCR3 cells in both R1 (totally positive cell fraction) and R2 (highly positive fraction) regions. The results revealed that the initial CD4+sCXCR3 cells tended to display reduced IFNγ expression when cells were co-cultured with relatively low doses of chrysotile, although exposure to chrysotile did not alter intracellular IFNγ expression in any of the three fractions.

Intracellular IL-17 expression following cultivation with chrysotile

Intracellular IL-17 expression was analyzed in CD4+sCXCR3 and CD4+sCXCR3+ fractions derived from cells following activation and cultivation with various concentrations of chrysotile (Fig. 4). The intracellular expression of IL-17 was significantly higher in CD4+sCXCR3+ cells compared with CD4+sCXCR3 cells in both R1 (totally positive cell fraction) and R2 (highly positive fraction) regions. The results showed that CD4+ fractions tended to display increased intracellular IL-17 expression, although there were no significant differences in intracellular IL-17 expression in any of the fractions.

IFNγ and IL-17 production following stimulation of cells cultured with chrysotile

Since analysis of the intracellular expression of IFNγ and IL-17 in cell fractions initially divided as CD4+sCXCR3 and CD4+sCXCR3+ following four weeks of cultivation with chrysotile asbestos did not show any marked alterations, these cells were re-stimulated by PMA and ionomycin for 4 h as shown in Fig. 1B. The concentration of IFNγ and IL-17 in culture supernatants was then measured.

As shown in Fig. 5, high levels of both IFNγ and IL-17 were found in CD4+sCXCR3+ fractions. In contrast, IFNγ production remained unaltered in cells cultured with chrysotile (Fig. 5A). However, IL-17 production was enhanced in cells cultured with chrysotile in a dose-dependent manner (Fig. 5B), although large variations were found.

IFNγ, T-bet, RORγT and IL-17 mRNA expression

Although IFNγ production remained unaltered in CD4+sCXCR3 and CD4+sCXCR3+ cells cultured with various concentrations of chrysotile asbestos, the production of IL-17 increased as shown in Fig. 6. Consequently, the expression of IFNγ, T-bet, RORγT and IL-17 mRNA was analyzed.

The results showed that cultivation with chrysotile did not modify the expression of T-bet, whereas IFNγ expression was reduced in cells initially categorized as CD4+sCXCR3. These results indicated that although cultivation with chrysotile did not appear to alter the Th1 population, Th1 function was reduced and depended in particular on the initial expression of CXCR3.

The Th17 status in these three fractions derived from cells treated with PMA and ionomycin and followed by T-cell receptor stimulation with IL-2 and cultivation with various concentrations of chrysotile asbestos was then examined. IL-17 mRNA expression was enhanced in both CD4+sCXCR3 and CD4+sCXCR3+ fractions, although the expression of RORγT increased slightly in cells co-cultured with chrysotile.

Discussion

The causative mechanisms of asbestos-induced cancer are thought to include 1) DNA damage due to ROS production by iron found in asbestos fibers, 2) direct chromosomal/DNA damage by physical attack of cells near the inhaled asbestos fibers, and 3) adsorption of various carcinogenic substances around the fiber itself (3336). The effect of these processes results in the generation of mesothelioma cells containing various genetic changes such as deletion of p16/p15 cyclin-dependent kinase-inhibitors (CDK-Is), deletion of NF2/merlin, and alteration (deletions and mutations) of BRCA1-associated protein 1/ubiquitin carboxyl-terminal hydrolase (BAP1) (3739). Mesothelioma is thought to result after a 30 to 40-year latency period following initial exposure to asbestos. During these long latency periods, and under conditions leading to the occurrence of other malignancies such as cancers of the larynx, gastrointestinal tract and bladder (22,23), it is thought that individuals exposed to asbestos and possessing asbestos fibers in their body might have reduced antitumor immunity due to asbestos exposure and recurrent and chronic encounters between intra-body fibers and immune competent cells. It is on this basis that we have been investigating the immunological effects of asbestos on immune competent cells, particularly in regard to antitumor immunity (1921,40).

Investigation of Th cells showed a reduction in CXCR3 surface expression and impaired IFNγ production capacity (17,18). The T-cell line model utilized in vitro stimulated freshly isolated human peripheral CD4+ cells and CD4+ cells derived from PP and MM (17,18). Since CXCR3 is thought to operate with IFNγ producing cells and functions to attack tumor cells, these findings also suggest that individuals exposed to asbestos possess reduced antitumor immunity (40).

We then investigated Th17 cells since they were considered to be important in the development of dysregulation of autoimmunity and cancer (2427). Some human cancers have been shown to possess increased levels of Th17 cells in the tumor, such as melanoma, ovarian and prostate cancers (2427). Furthermore, the polarization of Th17 cells was considered to be opposite to that of CD4+CD25+ and Forkhead box protein P3 (FoxP3) positive regulatory T cells (Treg) (41,42). Since enhancement of the volume or function of Treg may cause a reduction in antitumor immunity that inhibits tumor-recognizing T cells, the peripheral balance and polarizing conditions defined by cytokine conditions among IL-6 and TGFβ and between Treg and Th17 (2427) may be altered, and an increase in one population may cause a decrease in the other population. Therefore, an increase in number of cells or enhancement of Th17 function may reflect augmentation of antitumor immunity (2427).

A consideration of the overall results and our previous data suggests that asbestos exposure reduces CXCR3 expression and decreases IFNγ production capacity in Th1 cells, and increases IL-17 production capacity in CD4+ and surface CXCR3+ fractions. This production capacity appears to be strongly related to the CXCR3 positive cell fraction (Fig. 5), and reduction in the surface CXCR3 positive fraction during asbestos exposure was associated with a reduction in IL-17 production capacity in peripheral blood derived from asbestos-exposed patients. These findings also indicated that the immunological effects of asbestos lead to a reduction in antitumor immunity, and that these mechanisms may play a role in the subsequent occurrence of mesothelioma and other cancers in asbestos-exposed individuals.

The status of Treg in asbestos-exposed patients and the effect of asbestos on Treg require investigation. Recently, we indicated that asbestos enhances Treg function through a cell-cell contact pathway, and increases production of typical soluble factors such as IL-10 and TGFβ. These investigations also showed a reduction in antitumor immunity in asbestos-exposed patients. However, it remains unclear whether chronic, recurrent and continuous exposure of T cells to asbestos influences the polarization of Th subpopulations such as Th1, Treg and Th17.

Additionally, studies of Treg and Th17 should determine whether there is an increase in the number and function of tumors surrounding these subpopulations. However, such investigations must collect all of the immune competent cells from rejected tumor specimens. These issues need to be examined in future studies in an effort to address unresolved questions such as the alteration of antitumor immunity in mesothelioma patients. However, our focus is to consider the gradual decrease in antitumor immunity in asbestos-exposed patients following initial exposure to asbestos. Therefore, it would be better to analyze a population that has a significant exposure history to asbestos, but which does not show any significant physical alterations or the presence of certain cancers or PP.

The overall results suggest that Treg and Th17 conditions should be analyzed with respect to antitumor immunity not only in PP and MM patients, but also in individuals who have a significant history of asbestos exposure without any changes in health and body-related issues.

An experimental model should be explored in order to confirm the reduction in antitumor immunity. For example, as previously reported, continuous exposure of the human NK cell line YT-A1 to chrysotile asbestos for more than five months resulted in reduced antitumor killing activity against the human erythroblastic leukemia cell line K562, a commonly used target cell line to assay NK cell activity and compared with the original YT-A1 cell line, which has never been exposed to asbestos fibers (79). Regarding our previous reports showing that asbestos fibers enhance Treg function (43) and increase its volume by acceleration of the cell cycle (44), the construction of a cell co-culture model using human cell lines such as a Treg cell model or responder T cell model with mesothelioma (or other cancer) model might be difficult. As we previously reported, subjecting a human Treg model comprising the MT-2 cell line to continuous exposure to asbestos resulted in increased production of IL-10 and TGFβ (43). The effects of soluble factors such as these cytokines can be assayed using a transwell culture model. However, these soluble factors act to reduce antitumor immunity by inhibiting the attack of tumor cells by T cells. Thus, the model should comprise two cell types such as tumor cells and tumor-attacking responder T cells. Of course, it would be possible to match the HLA of some mesothelioma cell line with HV who could be used to provide PBMCs, including tumor attacking T cells. Thereafter, PBMCs derived from HLA-matched HV can be activated in vitro in the absence or presence of asbestos fibers. The tumor killing activity may then be examined in vitro with or without cell culture supernatants derived from a human Treg cell line model cultured in the absence or continuous presence of asbestos fibers. Animal models transplanting human mesothelioma cells can also be employed with the induction of immune cells in the absence or continuous presence of asbestos. Furthermore, since antibodies are produced against tumor cells, human myeloma cell lines can be employed and we have successfully established many of these cell lines (4547). These experiments should be performed in an effort to confirm our hypothesis that asbestos exposure causes a reduction in antitumor immunity.

In conclusion, we found that in vitro asbestos exposure of freshly isolated human T-cells induces IL-17 production. Further investigations regarding Th17 status in asbestos-exposed patients with PP or MM, and in individuals who do not exhibit any abnormalities, should be conducted to confirm the status of antitumor immunity in these asbestos-exposed individuals. Following confirmation of these hypotheses, certain kinds of food compositions or physiologically active substances derived from plants or other materials should be tested for their ability to neutralize the altered antitumor immunity in individuals exposed to asbestos in an effort to thwart the development of tumors.

Abbreviations:

NLRP3/NALP3

NOD-like receptor family, pyrin domain containing 3

IL

interleukin

NK

natural killer

HV

healthy volunteers

PP

pleural plaque

MM

malignant mesothelioma

CTL

cytotoxic T lymphocyte

PBMC

peripheral blood mononuclear cell

CB

chrysotile B

CR

crocidolite

MLR

mixed lymphocyte reaction

IFN

interferon

TNF

tumor necrosis factor

HTLV

human T-cell leukemia virus type

ROS

reactive oxygen species

TGF

transforming growth factor

STAT3

signal transducer and activator of transcription 3

CXCR3

C-X-C chemokine receptor type 3

mAb

monoclonal antibody

PMA

phorbol 12-myristate 13-acetate

IM

ionomycin

F

forward

R

reverse

TCR

T-cell receptor

CDK-I

cyclin-dependent kinase-inhibitor

BAP1

BRCA1-associated protein 1/ubiquitin carboxyl-terminal hydrolase

Th

T helper

FoxP3

Forkhead box protein P3

Treg

regulatory T cells

R

region

Acknowledgments

We thank Ms. Naomi Miyahara, Minako Katoh, Misao Kuroki, Keiko Kimura, Yoshiko Yamashita and Tomoko Sueishi for their technical assistance. This study was supported by Special Coordination Funds for Promoting Science and Technology grant H18-1-3-3-1 (Comprehensive Approach on Asbestos-Related Diseases), grants from the Ministry of Education, Culture, Sports, Science and Technology of Japan (18390186, 1965153, 19790411, 20390178 and 22700933), and Kawasaki Medical School Project Grants (18-209T, 19-205Y and 20-210O). This research was also partially supported by the Translational Research Network Program from the Japan Agency for Medical Research and Development (AMED).

References

1 

Fujimura N: Pathology and pathophysiology of pneumoconiosis. Curr Opin Pulm Med. 6:140–144. 2000. View Article : Google Scholar : PubMed/NCBI

2 

Mossman BT and Churg A: Mechanisms in the pathogenesis of asbestosis and silicosis. Am J Respir Crit Care Med. 157:1666–1680. 1998. View Article : Google Scholar : PubMed/NCBI

3 

Seaman DM, Meyer CA and Kanne JP: Occupational and environmental lung disease. Clin Chest Med. 36:249–268. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Cassel SL, Eisenbarth SC, Iyer SS, Sadler JJ, Colegio OR, Tephly LA, Carter AB, Rothman PB, Flavell RA and Sutterwala FS: The Nalp3 inflammasome is essential for the development of silicosis. Proc Natl Acad Sci USA. 105:9035–9040. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Dostert C, Pétrilli V, Van Bruggen R, Steele C, Mossman BT and Tschopp J: Innate immune activation through Nalp3 inflammasome sensing of asbestos and silica. Science. 320:674–677. 2008. View Article : Google Scholar : PubMed/NCBI

6 

Hornung V, Bauernfeind F, Halle A, Samstad EO, Kono H, Rock KL, Fitzgerald KA and Latz E: Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization. Nat Immunol. 9:847–856. 2008. View Article : Google Scholar : PubMed/NCBI

7 

Nishimura Y, Miura Y, Maeda M, Kumagai N, Murakami S, Hayashi H, Fukuoka K, Nakano T and Otsuki T: Impairment in cytotoxicity and expression of NK cell- activating receptors on human NK cells following exposure to asbestos fibers. Int J Immunopathol Pharmacol. 22:579–590. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Nishimura Y, Maeda M, Kumagai N, Hayashi H, Miura Y and Otsuki T: Decrease in phosphorylation of ERK following decreased expression of NK cell-activating receptors in human NK cell line exposed to asbestos. Int J Immunopathol Pharmacol. 22:879–888. 2009. View Article : Google Scholar

9 

Nishimura Y, Maeda M, Kumagai-Takei N, Lee S, Matsuzaki H, Wada Y, Nishiike-Wada T, Iguchi H and Otsuki T: Altered functions of alveolar macrophages and NK cells involved in asbestos-related diseases. Environ Health Prev Med. 18:198–204. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Kumagai-Takei N, Nishimura Y, Maeda M, Hayashi H, Matsuzaki H, Lee S, Hiratsuka J and Otsuki T: Effect of asbestos exposure on differentiation of cytotoxic T lymphocytes in mixed lymphocyte reaction of human peripheral blood mononuclear cells. Am J Respir Cell Mol Biol. 49:28–36. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Kumagai-Takei N, Nishimura Y, Maeda M, Hayashi H, Matsuzaki H, Lee S, Kishimoto T, Fukuoka K, Nakano T and Otsuki T: Functional properties of CD8(+) lymphocytes in patients with pleural plaque and malignant mesothelioma. J Immunol Res. 2014:6701402014. View Article : Google Scholar : PubMed/NCBI

12 

Kumagai-Takei N, Nishimura Y, Matsuzaki H, Maeda M, Lee S, Yoshitome K and Otsuki T: Effects of asbestos fibers on human cytotoxic T cells. Biological Effects of Fibrous and Particulate Substances (Series Title: Current Topics in Environmental Health and Preventive Medicine). Otsuki T, Holian A and Yoshioka Y: Springer-Japan; pp. 211–221. 2015

13 

Kumagai-Takei N, Nishimura Y, Matsuzaki H, Lee S, Yoshitome K, Hayashi H and Otsuki T: The suppressed induction of human mature cytotoxic T lymphocytes caused by asbestos is not due to Interleukin-2 insufficiency. J Immunol Res. 2016:74848722016. View Article : Google Scholar : PubMed/NCBI

14 

Hyodoh F, Takata-Tomokuni A, Miura Y, Sakaguchi H, Hatayama T, Hatada S, Katsuyama H, Matsuo Y and Otsuki T: Inhibitory effects of anti-oxidants on apoptosis of a human poly-clonal T-cell line, MT-2, induced by an asbestos, chrysotile-A. Scand J Immunol. 61:442–448. 2005. View Article : Google Scholar : PubMed/NCBI

15 

Miura Y, Nishimura Y, Katsuyama H, Maeda M, Hayashi H, Dong M, Hyodoh F, Tomita M, Matsuo Y, Uesaka A, et al: Involvement of IL-10 and Bcl-2 in resistance against an asbestos-induced apoptosis of T cells. Apoptosis. 11:1825–1835. 2006. View Article : Google Scholar : PubMed/NCBI

16 

Maeda M, Chen Y, Kumagai-Takei N, Hayashi H, Matsuzaki H, Lee S, Hiratsuka J, Nishimura Y, Kimura Y and Otsuki T: Alteration of cytoskeletal molecules in a human T cell line caused by continuous exposure to chrysotile asbestos. Immunobiology. 218:1184–1191. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Maeda M, Nishimura Y, Hayashi H, Kumagai N, Chen Y, Murakami S, Miura Y, Hiratsuka J, Kishimoto T and Otsuki T: Reduction of CXC chemokine receptor 3 in an in vitro model of continuous exposure to asbestos in a human T-cell line, MT-2. Am J Respir Cell Mol Biol. 45:470–479. 2011. View Article : Google Scholar

18 

Maeda M, Nishimura Y, Hayashi H, Kumagai N, Chen Y, Murakami S, Miura Y, Hiratsuka J, Kishimoto T and Otsuki T: Decreased CXCR3 expression in CD4+ T cells exposed to asbestos or derived from asbestos-exposed patients. Am J Respir Cell Mol Biol. 45:795–803. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Kumagai-Takei N, Maeda M, Chen Y, Matsuzaki H, Lee S, Nishimura Y, Hiratsuka J and Otsuki T: Asbestos induces reduction of tumor immunity. Clin Dev Immunol. 2011:4814392011. View Article : Google Scholar : PubMed/NCBI

20 

Matsuzaki H, Maeda M, Lee S, Nishimura Y, Kumagai-Takei N, Hayashi H, Yamamoto S, Hatayama T, Kojima Y, Tabata R, et al: Asbestos-induced cellular and molecular alteration of immunocompetent cells and their relationship with chronic inflammation and carcinogenesis. J Biomed Biotechnol. 2012:4926082012. View Article : Google Scholar : PubMed/NCBI

21 

Otsuki T, Matsuzaki H, Lee S, Kumagai-Takei N, Yamamoto S, Hatayama T, Yoshitome K and Nishimura Y: Environmental factors and human health: Fibrous and particulate substance-induced immunological disorders and construction of a health-promoting living environment. Environ Health Prev Med. 21:71–81. 2016. View Article : Google Scholar :

22 

Craighead JE: Nonthoracic cancers possibly resulting from asbestos exposure. Asbestos and its Diseases. Craighead JE and Gibbs AR: Oxford University Press; New York: pp. 230–252. 2008, View Article : Google Scholar

23 

Freidman GK: Malignant diseases attributed to asbestos exposure. Asbestos. Risk assessment, epidemiology, and health effects. Dodson RF and Hammar SP: CRC Press; Boca Raton: pp. 561–592. 2011, View Article : Google Scholar

24 

Alizadeh D, Katsanis E and Larmonier N: The multifaceted role of Th17 lymphocytes and their associated cytokines in cancer. Clin Dev Immunol. 2013:9578782013. View Article : Google Scholar

25 

Bailey SR, Nelson MH, Himes RA, Li Z, Mehrotra S and Paulos CM: Th17 cells in cancer: The ultimate identity crisis. Front Immunol. 5:2762014. View Article : Google Scholar : PubMed/NCBI

26 

Lakshmi Narendra B, Eshvendar Reddy K, Shantikumar S and Ramakrishna S: Immune system: A double-edged sword in cancer. Inflamm Res. 62:823–834. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Zou W and Restifo NP: T(H)17 cells in tumour immunity and immunotherapy. Nat Rev Immunol. 10:248–256. 2010. View Article : Google Scholar : PubMed/NCBI

28 

Ferro A, Zebedeo CN, Davis C, Ng KW and Pfau JC: Amphibole, but not chrysotile, asbestos induces anti-nuclear autoantibodies and IL-17 in C57BL/6 mice. J Immunotoxicol. 11:283–290. 2014. View Article : Google Scholar

29 

Serve KM, Black B, Szeinuk J and Pfau JC: Asbestos-associated mesothelial cell autoantibodies promote collagen deposition in vitro. Inhal Toxicol. 25:774–784. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Pfau JC, Serve KM and Noonan CW: Autoimmunity and asbestos exposure. Autoimmune Dis. 2014:7820452014.PubMed/NCBI

31 

Zebedeo CN, Davis C, Peña C, Ng KW and Pfau JC: Erionite induces production of autoantibodies and IL-17 in C57BL/6 mice. Toxicol Appl Pharmacol. 275:257–264. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Maeda M, Yamamoto S, Chen Y, Kumagai-Takei N, Hayashi H, Matsuzaki H, Lee S, Hatayama T, Miyahara N, Katoh M, et al: Resistance to asbestos-induced apoptosis with continuous exposure to crocidolite on a human T cell. Sci Total Environ. 429:174–182. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Huang SX, Jaurand MC, Kamp DW, Whysner J and Hei TK: Role of mutagenicity in asbestos fiber-induced carcinogenicity and other diseases. J Toxicol Environ Health B Crit Rev. 14:179–245. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Mossman BT and Craighead JE: Mechanisms of asbestos carcinogenesis. Environ Res. 25:269–280. 1981. View Article : Google Scholar : PubMed/NCBI

35 

Moyer VD, Cistulli CA, Vaslet CA and Kane AB: Oxygen radicals and asbestos carcinogenesis. Environ Health Perspect. 102(Suppl 10): 131–136. 1994. View Article : Google Scholar : PubMed/NCBI

36 

Toyokuni S: Iron overload as a major targetable pathogenesis of asbestos-induced mesothelial carcinogenesis. Redox Rep. 19:1–7. 2014. View Article : Google Scholar

37 

LaFave LM, Béguelin W, Koche R, Teater M, Spitzer B, Chramiec A, Papalexi E, Keller MD, Hricik T, Konstantinoff K, et al: Loss of BAP1 function leads to EZH2-dependent transformation. Nat Med. 21:1344–1349. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Sekido Y: Molecular pathogenesis of malignant mesothelioma. Carcinogenesis. 34:1413–1419. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Singhi AD, Krasinskas AM, Choudry HA, Bartlett DL, Pingpank JF, Zeh HJ, Luvison A, Fuhrer K, Bahary N, Seethala RR, et al: The prognostic significance of BAP1, NF2, and CDKN2A in malignant peritoneal mesothelioma. Mod Pathol. 29:14–24. 2016. View Article : Google Scholar

40 

Maeda M, Yamamoto S, Hatayama T, Matsuzaki H, Lee S, Kumagai-Takei N, Yoshitome K, Nishimura Y, Kimura Y and Otsuki T: T cell alteration caused by exposure to asbestos. Biological Effects of Fibrous and Particulate Substances, (Series Title: Current Topics in Environmental Health and Preventive Medicine). Otsuki T, Holian A and Yoshioka Y: Springer; Tokyo: pp. 195–210. 2015

41 

O'Connor RA, Taams LS and Anderton SM: Translational mini-review series on Th17 cells: CD4 T helper cells: functional plasticity and differential sensitivity to regulatory T cell-mediated regulation. Clin Exp Immunol. 159:137–147. 2010. View Article : Google Scholar :

42 

Olson NC, Sallam R, Doyle MF, Tracy RP and Huber SA: T helper cell polarization in healthy people: Implications for cardiovascular disease. J Cardiovasc Transl Res. 6:772–786. 2013. View Article : Google Scholar : PubMed/NCBI

43 

Ying C, Maeda M, Nishimura Y, Kumagai-Takei N, Hayashi H, Matsuzaki H, Lee S, Yoshitome K, Yamamoto S, Hatayama T, et al: Enhancement of regulatory T cell-like suppressive function in MT-2 by long-term and low-dose exposure to asbestos. Toxicology. 338:86–94. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Lee S, Matsuzaki H, Maeda M, Yamamoto S, Kumagai-Takei N, Hatayama T, Ikeda M, Yoshitome K, Nishimura Y and Otsuki T: Accelerated cell cycle progression of human regulatory T cell-like cell line caused by continuous exposure to asbestos fibers. Int J Oncol. 50:66–74. 2017.

45 

Tsujioka T, Miura Y, Otsuki T, Nishimura Y, Hyodoh F, Wada H and Sugihara T: The mechanisms of vitamin K2-induced apoptosis of myeloma cells. Haematologica. 91:613–619. 2006.PubMed/NCBI

46 

Otsuki T, Yata K, Takata-Tomokuni A, Hyodoh F, Miura Y, Sakaguchi H, Hatayama T, Hatada S, Tsujioka T, Sato Y, et al: Expression of protein gene product 9.5 (PGP9.5)/ubiquitin-C-terminal hydrolase 1 (UCHL-1) in human myeloma cells. Br J Haematol. 127:292–298. 2004. View Article : Google Scholar : PubMed/NCBI

47 

Otsuki T, Yamada O, Kurebayashi J, Moriya T, Sakaguchi H, Kunisue H, Yata K, Uno M, Yawata Y and Ueki A: Estrogen receptors in human myeloma cells. Cancer Res. 60:1434–1441. 2000.PubMed/NCBI

Related Articles

Journal Cover

June-2017
Volume 50 Issue 6

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Maeda M, Chen Y, Lee S, Kumagai-Takei N, Yoshitome K, Matsuzaki H, Yamamoto S, Hatayama T, Ikeda M, Nishimura Y, Nishimura Y, et al: Induction of IL-17 production from human peripheral blood CD4+ cells by asbestos exposure. Int J Oncol 50: 2024-2032, 2017
APA
Maeda, M., Chen, Y., Lee, S., Kumagai-Takei, N., Yoshitome, K., Matsuzaki, H. ... Otsuki, T. (2017). Induction of IL-17 production from human peripheral blood CD4+ cells by asbestos exposure. International Journal of Oncology, 50, 2024-2032. https://doi.org/10.3892/ijo.2017.3991
MLA
Maeda, M., Chen, Y., Lee, S., Kumagai-Takei, N., Yoshitome, K., Matsuzaki, H., Yamamoto, S., Hatayama, T., Ikeda, M., Nishimura, Y., Otsuki, T."Induction of IL-17 production from human peripheral blood CD4+ cells by asbestos exposure". International Journal of Oncology 50.6 (2017): 2024-2032.
Chicago
Maeda, M., Chen, Y., Lee, S., Kumagai-Takei, N., Yoshitome, K., Matsuzaki, H., Yamamoto, S., Hatayama, T., Ikeda, M., Nishimura, Y., Otsuki, T."Induction of IL-17 production from human peripheral blood CD4+ cells by asbestos exposure". International Journal of Oncology 50, no. 6 (2017): 2024-2032. https://doi.org/10.3892/ijo.2017.3991