Elsevier

Lung Cancer

Volume 94, April 2016, Pages 61-67
Lung Cancer

Chemokines involved in the early inflammatory response and in pro-tumoral activity in asbestos-exposed workers from an Italian coastal area with territorial clusters of pleural malignant mesothelioma

https://doi.org/10.1016/j.lungcan.2016.01.020Get rights and content

Highlights

  • Specific growth factors were observed in sera of asbestos exposed workers.

  • The chemokines IP-10 and RANTES were associated with the severity of diseases.

  • RANTES showed the highest serum level.

  • RANTES showed a significant relationship between sera and pleural effusions.

  • Mesothelioma biopsies showed detectable levels for RANTES, VEGF, and IP-10.

Abstract

Objectives

Immune mediators are likely to be relevant for the biological response to asbestos exposure. The aim of this study was to investigate the association between immune mediators involved in inflammation, cell survival and angiogenesis, and asbestos-related diseases in workers from a coastal area of North-East Italy with a high incidence of pleural malignant mesothelioma (PMM).

Materials and methods

A selected custom set of 12 soluble mediators was evaluated with a Luminex platform in sera, pleural fluid and mesothelioma biopsies from 123 asbestos-exposed workers (38 free from pleural-pulmonary disorders, 46 with non-malignant asbestos diseases, 39 with PMM) and in sera from 33 healthy controls from the same territorial area.

Results

Increased immune mediator concentrations were observed in the sera of the asbestos-exposed workers compared to controls for human fibroblast growth factor (FGF-b), vascular endothelial growth factor (VEGF), CCL5 (RANTES), CXCL10 (IP-10), CLEC11A (SCGF-b), CCL27 (CTACK), CCL11 (EOTAXIN), IL-5 and IL-6 (p < 0.001). The chemokines IP-10 and RANTES were associated with the severity of asbestos-related diseases. In the workers with PMM, the immune proteins secreted by mesothelioma biopsies showed detectable levels of RANTES, VEGF, and IP-10. In the same workers with PMM, a significant relationship between serum and pleural fluid concentrations was found for RANTES alone.

Conclusions

Occupational exposure to asbestos seems to drive the production of specific growth factors dually involved in the early inflammatory response and in pro-tumoral activity before clinical evidence of related disorders, suggesting that their over-expression may precede the onset of asbestos-related diseases. These findings suggest that some chemokines may have a prognostic role in the progression of asbestos-related diseases and could be used for the health surveillance of either workers with an occupational history of asbestos exposure or patients affected by non-malignant asbestos-related diseases.

Introduction

Exposure to asbestos and the nature of the inhaled fibers are associated with a high risk of developing lung and mesothelial diseases including pleural malignant mesothelioma (PMM) [1], [2].

In mesothelial and lung epithelial cells, asbestos fibers initiate signaling and survival pathways which are often up-regulated in PMM and contribute to tumor pathogenesis [3]. Asbestos fibers deposited in the distal regions of the respiratory system cause early and sustained inflammation. The inflammatory process is sustained by interaction of the fibers with epithelial cells and alveolar macrophages which, in turn, may release chemical mediators up-regulating these pathways. The mentioned pathways may be activated by a direct interplay of asbestos fibers with receptors on the cell surface and with integrins or via elaboration of ROS generated catalytically on the fiber surface or after frustrated phagocytosis [4]. In addition, an increase in the expression of genes encoding cytokines/chemokines such as IL-8 and their receptors or ligands such as CXCL2 and CXCL3 is promoted, suggesting that mesothelial cells may orchestrate leukocyte chemotaxis and trafficking and pleural inflammation [5], [6], [7], [8].

Recent research has reported that cancer, including mesothelioma, subvert the normal pathway of inflammatory proteins, which become crucial constituents of the tumor microenvironment and influence its development and metastasis. Specifically, the supernatants of asbestos-exposed mesothelioma cell lines contained higher levels of vascular endothelial growth factor (VEGF) than the supernatants of unstimulated normal mesothelial cells, and such differences were also noticed for fibroblast growth factor (FGF). Similarly, the serum levels of both proteins were found to be significantly higher in mesothelioma patients than in controls or patients with other malignancies [9].

TNF-α release and cooperation to the activation of the Ras/MAPK/NF-kB pathway is one of the described mechanisms by which tumor cell-macrophage crosstalk may enhance tumor growth and influence the over-expression of both CC and CXC chemokines in amplifying paracrine signaling loops [10], [11]. Nevertheless, the important contribution of IL-1b and TNF-α to mesothelial cell malignant transformation may be partially due to activation of AP-1 – or NF-kB – dependent gene expression [12].

The chronic inflammatory response to the asbestos cellular damage probably perpetrated by the Nod-like receptor protein 3 (NLRP3) inflammasome [13], [14] represents an important aspect of the pulmonary microenvironment, characterized by a strong interplay between high levels of soluble mediators, immune cells, airway epithelial cell proliferation and angiogenesis [3], [15], [16], [17]. Notably, independently of asbestos fiber deposition, an array of pro-angiogenic molecules, growth factors and pro-inflammatory cytokines has been detected during the abnormal growth and differentiation of mesothelial cells in visceral pleura. It is plausible that such changes, which precede tumor development, may be linked causally to PMM [18], [19], [20].

Malignant pleural mesothelioma is a clinically aggressive tumor and no single-modality therapy has proven effective in curing it, presumably because of the multiplicity of survival and chemoresistance pathways in this tumor. PMM seems to respond to immunotherapy and patients are known to mount an anti-tumor immune response [21]. Careful surveillance of patients exposed to asbestos is a key issue in controlling the development of asbestos-associated diseases and the identification of healthy, asymptomatic persons exposed to asbestos could be an important result.

Since the production of immune mediators is likely to be relevant for the biological response to asbestos exposure and PMM seems to be responsive to immunotherapy, we undertook the present study with the aim of investigating the associations between asbestos exposure, asbestos-related diseases and candidate immune mediators involved in multiple cellular pathways affecting the inflammatory response, cell survival and angiogenesis [22], [23], [24], [25], in a cohort of workers with a history of occupational asbestos exposure in a coastal area of North-East Italy with a high incidence of PMM [26].

Section snippets

Study population

For this cross-sectional study, 123 male workers with a history of definite occupational exposure to asbestos (amphibole types) were consecutively enrolled during either a health surveillance survey (n = 84) or during hospital attendance for diagnostic procedures at a Thoracic Surgery Unit (n = 39). These subjects had a past occupational history of asbestos use in dockyards (asbestos handling and transport) and shipyards (naval construction and/or repair activities) located in a coastal area of the

Characteristics of the study population

Table 2 reports the characteristics of the groups studied. The PMM workers were older (median: 69 y) and had greater length of occupational asbestos exposure (median: 25 y) than the other worker groups (p < 0.001). These findings are consistent with those of previous studies based on the local Mesothelioma Register [26], [27], [28]: in the 1022 PMM cases occurred over the calendar period 1995–2013, the age at diagnosis averaged 69 y and the duration of occupational exposure to asbestos varied, on

Discussion

The prognostic role of immune mediators in tumorigenesis has been investigated in several neoplastic diseases, including PMM of all subtypes [15], [48], [49], [50].

In the present study, a custom set of soluble immune mediators reported to be involved in PMM pathogenesis (Table 1) was evaluated in biological samples of asbestos-exposed workers with the aim of assessing their potential prognostic relevance for the development of asbestos-related diseases.

An important finding of our study was the

Conclusion

This in vivo study confirms that the damage to the mesothelium by prolonged exposure to asbestos fibers does not impair the capacity of mesothelial cells to regulate the level of expression of specific chemokines, dually involved in the early inflammatory response and in pro-tumoral activity. Moreover, the PMM microenvironment indicated that (i) PMM cells secrete these cytokines, (ii) the same proteins produced in response to asbestos fibers can be measured in the sera of patients, and (iii)

Conflict of interest

The authors declare no conflicts of interest.

Acknowledgment

This work was supported by a grant from Regione Friuli Venezia Giulia, L.R. 22/2001/decree 889/Area Prevenzione (APREV).

References (64)

  • R. Gaafar et al.

    Tissue and serum EGFR as prognostic factors in malignant pleural mesothelioma

    Lung Cancer

    (2010)
  • A. Yasumitsu et al.

    Clinical significance of serum vascular endothelial growth factor in malignant pleural mesothelioma

    J. Thorac. Oncol.

    (2010)
  • M.C. Hartmann et al.

    Relationship between CCL5 and transforming growth factor-β1 (TGFβ1) in breast cancer

    Eur. J. Cancer

    (2011)
  • S.C. Kao et al.

    The predictive role of serum VEGF in an advanced malignant mesothelioma patient cohort treated with thalidomide alone or combined with cisplatin/gemcitabine

    Lung Cancer

    (2012)
  • K.A. Oldham et al.

    T lymphocyte recruitment into renal cell carcinoma tissue: a role for chemokine receptors CXCR3, CXCR6, CCR5, and CCR6

    Eur. Urol.

    (2012)
  • B.T. Mossman et al.

    New insights into understanding the mechanisms, pathogenesis, and management of malignant mesotheliomas

    Am. J. Pathol.

    (2013)
  • B.T. Mossman et al.

    Pulmonary endpoints (lung carcinomas and asbestosis) following inhalation exposure to asbestos

    J. Toxicol. Environ. Health B Crit. Rev.

    (2011)
  • C. Dostert et al.

    Innate immune activation through Nalp3 inflammasome sensing of asbestos and silica

    Science

    (2008)
  • Nicholas H. Heintz et al.

    Asbestos, lung cancers, and mesotheliomas. From molecular approaches to targeting tumor survival pathways

    Am. J. Respir. Cell Mol. Biol.

    (2010)
  • G. Galffy et al.

    Inhibition of interleukin-8 reduces human malignant pleural mesothelioma propagation in nude mouse model

    Oncol. Res.

    (1999)
  • A. Shukla et al.

    Alterations in gene expression in human mesothelial cells correlate with mineral pathogenicity

    Am. J. Respir. Cell Mol. Biol.

    (2009)
  • G.D. Hill et al.

    Soluble ICAM-1, MCP-1, and MIP-2 protein secretion by rat pleural mesothelial cells following exposure to amosite asbestos

    Exp. Lung Res.

    (2003)
  • Y.M. Janssen-Heininger et al.

    Cooperativity between oxidants and tumor necrosis factor in the activation of nuclear factor (NF)-kappaB: requirement of Ras/mitogen-activated protein kinases in the activation of NF-kappaB by oxidants

    Am. J. Respir. Cell Mol. Biol.

    (1999)
  • Y.M. Janssen et al.

    Asbestos induces nuclear factor kappa B (NF-kappa B) DNA-binding activity and NF-kappa B-dependent gene expression in tracheal epithelial cells

    Proc. Natl. Acad. Sci. U. S. A.

    (1995)
  • Y. Wang et al.

    Interleukin-1beta and tumour necrosis factor-alpha promote the transformation of human immortalised mesothelial cells by erionite

    Int. J. Oncol.

    (2004)
  • J.M. Hillegass et al.

    Asbestos and erionite prime and activate the NLRP3 inflammasome that stimulates autocrine cytokine release in human mesothelial cells

    Part. Fibre Toxicol.

    (2013)
  • M.T. Chow et al.

    NLRP3 promotes inflammation-induced skin cancer but is dispensable for asbestos-induced mesothelioma

    Immunol. Cell Biol.

    (2012)
  • H. Yang et al.

    TNF-alpha inhibits asbestos-induced cytotoxicity via a NF-kappaB-dependent pathway, a possible mechanism for asbestos-induced oncogenesis

    Proc. Natl. Acad. Sci. U. S. A.

    (2006)
  • H. Yang et al.

    Programmed necrosis induced by asbestos in human mesothelial cells causes high-mobility group box 1 protein release and resultant inflammation

    Proc. Natl. Acad. Sci. U. S. A.

    (2010)
  • J.M. Hillegass et al.

    Inflammation precedes the development of human malignant mesotheliomas in a SCID mouse xenograft model

    Ann. N. Y. Acad. Sci.

    (2010)
  • Y. Wang et al.

    Interleukin-1 beta and tumour necrosis factor-alpha promote the transformation of human immortalised mesothelial cells by erionite

    Int. J. Oncol.

    (2004)
  • B.W. Robinson et al.

    Localised spontaneous regression in mesothelioma-possible immunological mechanism

    Lung Cancer

    (2001)
  • Cited by (19)

    • Journey to the centre of the lung. The perspective of a mineralogist on the carcinogenic effects of mineral fibres in the lungs

      2023, Journal of Hazardous Materials
      Citation Excerpt :

      Activated M1 macrophages (but also lung epithelial cells) release pro-inflammatory cytokines (like TNF-α, IL-1β, IL-6, IL-12, IL-15), and promote the production of reactive oxygen species (ROS), proteolytic enzymes and bioactive lipids. TNF-α, in turn, stimulates the release of α-chemokines (IL-8 and IP-10 for example) or β-chemokines (like MCP-1, MIP and RANTES) mediators of inflammation via paracrine and autocrine pathways (Driscoll et al., 1997; Comar et al., 2016). M2 macrophages, which can be polarized by IL-4 and/or IL-13 stimuli (Rhee, 2016), may appear later at the site of inflammation and counteract the pro-inflammatory activity of the M1 phenotype by secretion of anti-inflammatory mediators (e.g., IL-10, CCL18, CCL22) and tissue repair promoting factors, such as TGF-β, VEGF and FGF (Martinez et al., 2009).

    • The effects of asbestos fibers on human T cells

      2020, International Journal of Molecular Sciences
    View all citing articles on Scopus
    View full text