Open Access

Update of in vitro, in vivo and ex vivo fluoro‑edenite effects on malignant mesothelioma: A systematic review (Review)

  • Authors:
    • Veronica Filetti
    • Ermanno Vitale
    • Giuseppe Broggi
    • Maria P. Hagnäs
    • Saverio Candido
    • Anna Spina
    • Claudia Lombardo
  • View Affiliations

  • Published online on: October 15, 2020     https://doi.org/10.3892/br.2020.1367
  • Article Number: 60
  • Copyright: © Filetti 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

Fluoro‑edenite (FE), asbestiform fiber found in Biancavilla (Sicily, Italy), presents various characteristics similar to the asbestos group, in particular two fibrous phases tremolite and actinolite. Indeed, epidemiological studies have shown that FE fibers have similar effects to those of asbestos fibers. Such studies have reported a high incidence of malignant mesothelioma (MM), an aggressive neoplasm of the serosal membranes lining the pleural cavity, in individuals residing there due to FE exposure in Biancavilla related to environmental contamination. Evidence has led to the classification of FE as a Group 1 human carcinogen by the International Agency for Research on Cancer (IARC). The aim of this systematic review is to compare the results achieved in in vitro, in vivo and ex vivo experimental studies involving FE in order to update the current knowledge on the pathogenesis and molecular mechanisms responsible for FE‑mediated MM development as well as the availability of effective biomarkers for MM prevention and diagnosis. This review is focused on the pathophysiological mechanisms mediated by inflammation induced by FE fiber exposure and which are responsible for MM development. This review also discusses the discovery of new diagnostic and prognostic biomarkers for the management of this pathology. It is known that the risk of cancer development increases with chronic inflammation, arising from enhanced reactive oxygen species (ROS) and NO production stimulated by the body to remove exogenous agents, causing DNA damage and enhanced signal transduction that may lead to activation of oncogenes. Studies concerning MM biomarker discovery indicate that several biomarkers have been proposed for MM, but mesothelin is the only Food and Drug Administration (FDA)‑approved biomarker for MM, with limitations. In recent studies, in silico analysis to identify selected miRNAs highly deregulated in cancer samples when compared with normal control have been developed. This in silico approach could represent an effort in the field of biomarker discovery for MM.

1. Introduction

It is well known that the general population may be exposed to ‘naturally occurring asbestos’ (NOA) (1-4). The term NOA refers to the mineral as a natural component of soils or rocks as opposed to asbestos in commercial products, mining or processing operations. NOA may be released as fibers into the air by human activities or natural weathering processes that represent a risk for human exposure (3).

A fiber is defined as a particle (length/diameter ratio 3:1) with certain characteristics that make it respirable, penetrating into the alveolar level and participating in gaseous exchanges (5). In vitro, in vivo and ex vivo studies have demonstrated that the dimension, surface property, shape, cristallinity, chemical composition, physical durability, exposure route, duration of exposure, dose (6) and genetic background of the host exposed (7) are all determinants for the biological activities of a certain fibrous element. Fiber dimensions are important because only fibers with diameter <0.4 µm and length <10 µm are respirable to the distal alveolar space. Dose is a crucial determinant for triggering inflammation. Indeed, high doses over short periods promote an acute neutrophil-predominant inflammation, whereas low doses over prolonged exposure periods promote alveolar macrophage (AM)-predominant chronic inflammation (7). In general, fibers greater than 20 µm in length are associated with asbestosis, and fibers greater than 10 µm in length are the most carcinogenic (5).

Carcinogenic mineral fibers are divided into asbestos and asbestiform fibers (8,9). The term ‘asbestos’ is used to identify silicate minerals belonging to two families: Amphiboles (amosite, anthophyllite, actinolite, crocidolite, tremolite, anthracite) and serpentine (crysotile) (6,10). Amphiboles are straight, rod-like fibers, whereas serpentines are curvilinear fibers. Asbestos has a significant industrial importance for its characteristics, its abundance and its low cost. Asbestos has sound-absorbing and sound-proofing properties, thermal stability at high temperatures, good mechanical resistance, good resistance to chemical and biological agents, and for this high versatility it has been widely used in different areas, especially in the building industry (11).

Exposure to asbestos fibers can cause several diseases such as asbestosis, lung and bronchus cancer, malignant mesothelioma (MM) of the pleura, peritoneum, pericardium and tunica vaginalis testis, neoplasms of the ovary, larynx and trachea carcinoma (11). There is evidence that the inhalation of asbestos fibers can provoke two types of inter-connected pathogenetic processes: Chronic inflammation and carcinogenesis, involving the lung after inhalation and deposition of asbestos fibers (12). Therefore, it has been established that cancer frequently arises in areas of chronic inflammation (13). Many lines of evidence have highlighted the ability of asbestos fibers to: Interfere with the mitotic apparatus; stimulate host cell proliferation; induce genetic and epigenetic alterations; induce cytotoxicity and fibrosis; produce oxidative stress by at least three sources including fiber surface reactivity, release from inflammatory cells especially AMs, and mitochondrial-derived ROS release from inflammatory and other target cells such as lung epithelial cells and mesothelial cells (7). The ROS production results in DNA damage, release of inflammatory cytokines and growth factors that collectively contribute to fiber pathogenicity (12,14) and H2O2 production in mediating asbestos pulmonary toxicity (7).

Several studies have reported a high incidence of MM due to asbestos exposure in: Finland (15), California, USA (16), China (17), New Caledonia (18), Corsica (19), Cyprus (20) and Greece (21). Yet, in many cases, it has been discovered that the cause of these MM cases has not been asbestos but asbestiform fibers.

The term ‘asbestiform fiber’ is commonly used to indicate erionite, winchite, magnesio-riebeckite, richterite, Libby asbestos, antigorite and fluoro-edenite (FE) fibers. Erionite is a mineral belonging to the zeolite family (22). Several studies have reported a high incidence of MM due to erionite exposure in rural regions of Turkey, Central Anatolia (23-27). Furthermore, individuals exposed to erionite may develop interstitial fibrosis and additional pulmonary pathology impacting lung function and patient survival (28).

Clark and Nye counties, in southern Nevada, USA, have shown a significantly high incidence of MM due to carcinogenic fibers including erionite, winchite, magnesio-riebeckite, and richterite (29). These are the same fibrous minerals present in Libby, Montana, USA, where they have been related to MM and other asbestos-related diseases (30).

Antigorite is a silicate mineral very similar in chemical composition to chrysotile and its asbestiform variant is present in serpentinite rocks associated with MM (31). Antigorite is found in the Western Alps (Piemonte, Italy) (32,33), in North America, Australia-Oceania, and Rowland Flat in South Australia (34,35).

Fluoro-edenite (FE), the amphibole of Biancavilla (Sicily, Italy), is a silicate mineral belonging to the amphibole family (36). This silicate mineral has been identified in the lavic products of Monte Calvario from stone quarries located in the southeast of Biancavilla (37), a small town of the Etnean volcanic complex, in Sicily. This silicate mineral presents some characteristics similar to the asbestos group (38,39); in particular it presents the same morphological and compositional aspect of the two fibrous phases tremolite and actinolite. The mineralization process led to the development of large prismatic crystals embedded in the matrix, small acicular crystals that line cavities or also fibrous and asbestiform (37). The salient feature, which nonetheless distinguishes the FE of Biancavilla not only from other fibrous minerals, but also from all the other known amphiboles, is the very anomalous composition characterized by high sodium, aluminum and fluorine contents, in comparison to other known oncogenic minerals (40).

Epidemiological studies have indeed confirmed that FE fibers have shown similar effects to those already reported after exposure to asbestos fibers (8,41-43) including cell necrosis with release of high mobility group protein B1 and activation of the Nalp3 inflammasone, leading to chronic inflammation, DNA damage and carcinogenesis (44).

Several studies have reported a high incidence of MM in Italy due to FE exposure in Biancavilla (45-48) concerning the time window 1980-2009. All of the data suggest that a mode of exposure to FE fibers is related to environmental contamination, rather than specific occupational activities (45). In fact, the stone material from the quarry of Monte Calvario has been used locally for about 50 years for building purposes (8,49,50) and none of the residents diagnosed with MM have been significantly exposed to asbestos during their professional lives (12).

Diseases related to erionite and FE fibers present with characteristics similar to asbestos-related pathologies. The underlying modes of action of asbestosis, lung cancer and MM seem to be different in regards to the fiber type, lung clearance, and genetics. Several lines of evidence have led to the classification of asbestos, erionite, and FE as Group 1 human carcinogens (51; IARC, 1987).

Therefore, NOA represents an important environmental concern. Thus, asbestos and asbestiform fibers continue to cause a high health concern due to the long latency period of related diseases.

2. Literature search methodology

This systematic review was carried out in accordance with PICo criteria (52). The review/research question was defined, using PICo criteria, by identifying: Population, interest, and context of research. What are the in vitro, in vivo and ex vivo studies correlated with MM due to FE exposure in Biancavilla? (Fig. 1). The research was performed by using the following search term: ‘Fluoro-edenite fibers’. A search of the research manuscripts suitable for inclusion in this systematic review, was carried out and the research papers of significance were collected and reviewed. The main topics and alternate terms from our PICo question that were used for the search were: Fluoro-edenite exposure, Fluoro-edenite, Fluoro-edenite fibers, Fluoro-edenite fibres, Biancavilla, Biancavilla's exposure, Malignant mesothelioma. The English language was used as a limit to our search. SCOPUS and Medline (using PubMed as the search engine) databases were used to search relevant research articles available from March 4 to August 4, 2020 (Fig. 1).

Inclusion and exclusion criteria

The following inclusion criterion was adopted: Experimental studies that assessed the effects of FE fiber exposure in vitro, in vivo and ex vivo models. The following exclusion criteria were applied: i) Scientific articles that were not published in the English language; ii) review or conference abstracts or letters to the editor; iii) experimental studies that did not concern in vitro, in vivo and ex vivo models. For duplicate studies, the article containing further detailed information was solely included.

Quality assessment and data extraction

Two reviewers (VF and CL) retrieved articles independently. The title, abstract and full text of each potentially pertinent study were reviewed. Any divergence on the eligibility of the studies was determined by debate. The following information was extracted from all qualified papers: Authors, year of publication, and study characteristics.

After a free search for scientific literature by reviewers, a total of 29 documents were collected. In conclusion, 5 studies were disqualified after review of the manuscript. A total of 24 studies satisfied the inclusion criteria and were included in this systematic review. A flow-chart depicting the choice of studies is documented in Fig. 2. The information concerning authors, year of publication, and characteristics of included studies have been included in Table I.

Table I

Characteristics of the studies included in the systematic review.

Table I

Characteristics of the studies included in the systematic review.

 (Refs.)Samples
In vitro N=11Travaglione et al (12)A549 cells
 Cardile et al (60)J774 cells
 Cardile et al (64)A549 cells
  J774 cells
  Human lung fibroblasts
 Travaglione et al (56)A549 cells
 Cardile et al (32)MeT-5A cells
  J774 cells
 Pugnaloni et al (54)MeT-5A cells
  A549 cells
  J774 cells
 Loreto et al (63)A549 cells
 Musumeci et al (69)MeT-5A cells
  A549 cells
 Rapisarda et al (74)MeT-5A cells
 Rapisarda et al (76)Human lung fibroblasts
 Filetti et al (77)MeT-5A cells JU77 cells
In vivo N=2Soffritti et al (36)Sprague-Dawley rats
 Belpoggi et al (88)Sprague-Dawley rats
Ex vivo N=11DeNardo et al (82)Lung tissue of sheep
 Martinez et al (43)Lung tissue of sheep
 Loreto et al (83)Lung tissue of sheep
 Musumeci et al (84)Lung tissue of sheep
 Musumeci et al (92)Lung tissue of sheep
 Musumeci et al (85)Lung tissue of sheep
 Loreto et al (86)Lung and lymph nodes of sheep
 Rapisarda et al (93)Tracheobronchial lymph nodes of sheep
 Ledda et al (87)Tracheobronchial lymph nodes of sheep
 Angelico et al (94)Human MM tissue
 Caltabiano et al (44)Human MM tissue

[i] MM, malignant mesothelioma. A549, human pulmonary epithelial cancer cells; MeT-5A, human pleural mesothelial cells; J774, mouse alveolar monocyte-macrophage cells; JU77, human MM cells.

3. In vitro studies concerned with MM due to FE exposure

Various studies have been conducted to investigate the effects of FE fibers on several cell lines which are commonly used to evaluate the cytotoxicity of various silica dusts (53): A549 (human pulmonary epithelial cancer cells), MeT-5A (human pleural mesothelial cells), J774 (mouse alveolar monocyte-macrophage cells), JU77 (human MM cells), and human lung fibroblasts. Epithelial cells are involved in proinflammatory effects and they are the cells of origin of bronchogenic carcinoma. In addition, the transformation of mesothelial cells leads to mesothelioma, thus they are suitable to determine the direct effects of fibers. Alveolar macrophages are the first defense mechanism against particulates and fibers entering the lower respiratory tract (54); and fibroblasts represents a cell type of central regulatory potential in lung diseases (55).

Prismatic vs. fibrous FE

Several studies (12,56) have reported the cellular effects of prismatic and fibrous FE by using the A549 cell line. These cells, upon contact with prismatic FE, develop actin-rich protrusions from the plasma membrane, namely ruffles and filopodia, that allow the capture and internalization of material into the cytoplasm of epithelial cells. This phagocytic-like behavior of cells exposed to prismatic FE occurs only after the cells have reached the confluent state. The organization of the actin cytoskeleton, which represents one of the key target for a huge number of toxicants, remains well organized both in control and in treated A549 cells. In contrast with the results obtained with prismatic FE (12), the fibrous mineral provokes dramatic changes in the actin network of A549 cells (56). In particular, the actin stress fibers completely disappeared and actin-rich membrane ruffles arise from the cell surface following FE fiber exposure. In addition to these effects, the results obtained show that fibrous FE promote multinucleation, cell spreading and a dramatic increase in cell size, but without interfering with the passage of the resulting multinucleated cells through the cell cycle and without condemning cells to death.

Cytokines and growth factors derived from alveolar macrophages are implicated as mediators of asbestos-induced patho-physiological responses (57). Indeed, inflammation also characterizes the response of epithelial cells to external danger, which produce an array of mediators transmitting cellular signals (58). Interleukin (IL)-6 and IL-8 concentrations, that are respectively a multifunctional cytokine with immunoregulatory and proinflammatory effects (56) and a chemotactic cytokine involved in the recruitment of polymorphonuclear granulocytes to the site of injury (59), have been determined by Travaglione et al (12,56) after treatment of A549 cells with prismatic and fibrous FE. The results revealed that although prismatic FE interferred with cell physiology, by reducing the proliferation rate and increasing the release of the proinflammatory cytokine IL-6, this did not perturb the cell cycle and there was no evidence of any particular effects correlated to cellular transformation (12). On the contrary, in the case of fibrous FE exposure, an increase in both IL-6 and IL-8 secretion in the surnatant, in a time dependent-manner, was demonstrated (56). Cytokines, produced by epithelial cells in response to a cellular damage, activate neutrophils and macrophages which accumulate in the injured area. The recruited cells then produce ROS (60) and additional cytokines in an attempt to remove the unsafe agent. With chronic inflammation, tissue fibrosis can occur concomitantly with an enhanced risk of cancer development, arising from enhanced ROS production leading to DNA mutations and enhanced signal transduction that may lead to activation of oncogenes.

Therefore, these research studies, which demonstrated a pro-mesotheliomatogenic effect of fibrous but not prismatic FE in lung epithelial cells, highlight a differential cell response ensued by prismatic or fibrous FE.

Fibrous FE vs. crocidolite and tremolite

In lung epithelial cancer A549 cells, the behavior of FE fibers is similar to crocidolite (56), whose link with chronic inflammation and lung cancer is renowned (61,62). Fibrous FE causes actin reorganization and multinucleation accompanied by a significant increase in cell size. While these effects became evident after 48 h of FE exposure, in the case of crocidolite exposure the effects are observable at 24 h (56). Loreto et al (63) demonstrated that also FE 27 (70% of Fe3+), FE 19 (50% Fe3+) and tremolite fibers are able to promote the formation of multinucleated cells, but the phenomenom is greater in cells following crocidolite exposure (56,63).

Furthermore, the incubation with fibrous FE and crocidolite causes a comparable decrease in the number of viable A549 cells (56), J774 cells and human lung fibroblasts (64). Pӓӓkkӧ et al (65) associated the observed reduction in A549 growth to the induction of the apoptotic process. The results reported that crocidolite is able to cause apoptosis in A549 cells, associating this phenomenom to the nuclear accumulation of the p53 protein, a transcription factor mainly involved in cell cycle arrest and apoptosis after DNA damage. In contrast, Travaglione et al (56) demonstrated that both fibrous FE and crocidolite were not able to influence the expression of p53 and of pro-Bax and anti-apoptotic Bcl-2 and Bcl-XL proteins. Furthermore, neither fibrous FE nor crocidolite were able to induce alterations in the passage of A549 cells throughout the cell cycle; the percentages of cells in the G0/G1, S, G2/M phases did not vary between the control and cells exposed to the fibers.

In order to investigate the proinflammatory potential of FE 19 and FE 27, tremolite, and crocidolite fibers, the secretion of IL-8(56), IL-6, IL-1β and TNF-α (63) were determined in A549 cells. All fibers increased the release of proinflammatory cytokines in a time-dependent manner, but crocidolite was found to promote a more consistent secretion of the analyzed cytokines (59,63); on the contrary, tremolite induced a reduced release of the same (63). Among cytokines, it has been demonstrated that IL-1β and tumor necrosis factor (TNF)-α act through the phosphoinositide-specific phospholipase C (PLC) pathway to activate protein kinase C (PKC) (66), essential for many cellular functions such as the processes of secretion, differentiation, proliferation and cell growth (67). However, it is known that asbestos stimulates ROS generation by interaction with cellular membranes or through protein tyrosine kinase (PTK), PLC and PKC pathway activation in a dose-response manner (68). Loreto et al (63) aimed to ascertain whether exposure to FE 27 and FE 19 fibers may induce cytokine increase related to PLC activation in A549 cells comparing these effects with those of tremolite and crocidolite. The results revealed that all fibers induced PLC expression in lung epithelial cells following exposure to 50 µg/ml for 48 h but with a different level of expression induced by each fiber. Tremolite showed the highest level of expression of isoform PLC-β1 while FE 19 showed the highest level of expression of isoform PLC-γ1. FE 27 showed values of expression of both the comparable isoforms, while crocidolite fibers induced lower expression of PLC. The possible induction of oxidative stress was also examined by Cardile et al (64) by evaluating the intracellular ROS production, the amount of nitrite/nitrate and the expression of inducible nitric oxide synthase (iNOS). In this case, the cell lines exposed to FE and crocidolite included A549, J774 and human lung fibroblasts. The results indicated that the increase in ROS generation was directly proportional to fiber concentration and exposure time. Moreover, in all experimental cultures, NO synthesis and iNOS expression increased after crocidolite fiber but not after FE fiber exposure. Cell cytotoxicity was also evaluated assessing lactic dehydrogenase (LDH) release in A549, J774, human lung fibroblasts exposed to FE and crocidolite. The presence of LDH in culture medium is a marker of membrane breakdown. The results revealed that LDH release was significantly increased when cell cultures, in particular A549 cells, were exposed to 50 µg/ml FE or crocidolite for at least 48 h, or the when the fiber concentration was increased to 100 µg/ml for 24 h (64).

The sensitivity to mineral fibers of A549 cells has also been compared with pleural mesothelial MeT-5A cells (54,69). The data demonstrated the critical role of epithelial and mesothelial cross-talk in FE fiber exposure and that these are able to induce functional modifications in a variety of parameters with crucial roles in cell cycle control, cancer development and progression. Pugnaloni et al (54) investigated the distribution of polymerized actin in A549 and MeT-5A cells exposed to FE fibers. Furthermore, the effects of FE exposure on the synthesis of vascular endothelial growth factor (VEGF), β-catenin (54) and retinoblastoma (Rb) (69), three critical steps of epithelial cell activation pathways, have been investigated. The results showed greater viability in A549 than in MeT-5A cells exposed to FE fibers (54,69) with consequent actin staining more irregular and granular than in the respective controls, suggesting that FE induces a dyregulated assembly of actin (54). The pathogenicity of FE fibers was demonstrated by the almost total arrest of cell movement at 48 h in both cell lines and by the flocculation of F-actin molecules. After a 48-h incubation, VEGF and β-catenin expression were evident in both cell lines (54), suggesting that FE fibers do not exert a primary toxic action inducing rapid cell death, but induce an abnormal cellular status with upregulated cell activities and a risk of cell transformation (54,70-72). In contrast to expectations, A549 and MeT-5A cells exposed to FE 19 fibers exhibited no change in Rb level, but overexpressed phospho-retinoblastoma (pRb) (69). The initial status of Rb changes with the cell cycle and it is regulated by the activity of cyclins D1, D2, D3 and CDK4 or CDK6 complexes in mid-G1 phase (73). Rb promotes cell cycle arrest and a return to the G1 phase; it has a central role in most instances of apoptosis, while pRb functions as a checkpoint in the G1 phase promoting cell growth. Since cyclin D1 is a sensor of cell division signals, it has been evaluated whether its expression is correlated with pRb expression; indeed, there was found a positive relationship in a dose-response manner in both cell lines (69). Furthermore, FE reduced the p27 expression both in A549 cells (69) and in MeT-5A cells (69,74), a tumor-suppressor gene due to its function as cell cycle regulator, that in cancer is often inactivated (75). The downregulation of p27 is associated with stathmin-1 upregulation in cancer, conferring an aggressive phenotype to cancer cells (74). In addition, fibulin-3 (Fb-3) overexpression may be responsible for the malignant transformation of MeT-5A cells after exposure to FE fibers (74). Fb-3 overexpression reflects a defensive response of the tissues after exogenous stimuli as FE exposure (76).

Functional in vitro experiments performed on MeT-5A and JU77 cells have been carried out in order to test the carcinogenetic effects and epigenetic modulation induced by FE exposure (77). The results showed that MeT-5A cells were more sensitive to FE fibers compared to JU77 tumor cells. The in silico analyses revealed a set of miRNAs strictly involved in MM and these have been used as in vitro experimental targets. The in vitro results showed that the expression levels of hsa-miR-323a-3p vared significantly in both supernatant- and cell-derived miRNAs derived from treated and untreated cells. Secreted and cellular hsa-miR-101-3p in MeT-5A cells exposed to FE fibers and JU77 cells showed different trends of expression. In regards to hsa-miR-20b-5p, there was no differential expression between secreted and cellular hsa-miR-20b-5p. This miRNA has shown a significant upregulation in JU77 cells vs. control and treated MeT-5A. Certainly, translational analyses will be performed on a subset of patients chronically exposed to FE fibers to further verify the clinical role of such miRNAs in high-risk individuals and their possible use as biomarkers of FE exposure or MM early onset (77).

The monocyte-macrophage J774 cells are more sensitive to FE than MeT-5A cells, suggesting that the primary site of the inflammatory response induced by mineral fibers could be the macrophage rather than the lung epithelium (78). The greatest sensitivity has been demonstrated in terms of heat shock protein 70 (Hsp70) induction, that was found to stimulate the formation of ROS and NO. The form 19 was found to have a markedly strong effect on NO biosynthesis while the form 27 had a stimulatory effect on ROS generation in J774 cells, in contrast to MeT-5A cells. In contrast, detection of LDH release, which is a marker of cell necrosis, was found to be amplified in MeT-5A cells compared to J774 cell line (60,64,78). In general, tremolite is less effective than FE in producing more biological alterations, while it is inactive at 5 µg/ml. In opposition, at higher concentrations FE 19 is stronger than the other particulate (78).

Alveolar macrophages have an important role in the fibrotic process involved in silicosis and in other lung diseases (79). They are mediators in the interaction between inhaled particulates and different types of cells, by the release of a variety of inflammatory and growth-mediating factors (80). Cyclooxygenase-2 (COX-2) catalyses the conversion of arachinodic acid to prostaglandins (PGs) and it is mainly induced in response to proinflammatory stimuli, cytokines, growth factors and mitogens. It is known that PGs have an important role in cancer pathogenesis (81); therefore, Pugnaloni et al (54) demonstrated a time-dependent COX-2 overexpression and a PGE2 increase in monocyte-macrophage J774 cells exposed to FE. PEG2 derived from COX-2 was found to be involved in solid tumor pathogenesis through inhibition of apoptosis, facilitation of tumor cell invasiveness and promotion of angiogenesis (54).

4. In vivo studies concerned with MM due to FE exposure

It is important to underline that FE fibers were identified in the lungs of a housewife of Biancavilla, who died subsequently to a diagnosis of MM (45), in lung tissue (43,82-86) and lymph nodes (86,87) of sheep living in the Biancavilla area. Indeed, several studies have been performed to evaluate the relationship between FE and MM and to prove the biopersistence of these fibers in tissue.

Induced exposure of prismatic vs. fibrous FE

Different authors (36,88) have tested the same concentration of two mineral forms, prismatic and fibrous FE, with two administration methods, single intraperitoneal and intrapleural injection, on groups of Sprague-Dawley rats, to acquire more information on the potential relationship between exposure to FE and MM. The results are concordant and in line with previous preliminary data (64), and there is evident confirmation regarding a mesotheliomatogenic potential of FE fibers. In contrast, prismatic FE failed to induce a mesotheliomatogenic response in the totality of the animals, in accordance to the controls (36,88).

The intraperitoneal injection of fibrous FE caused effects much stronger than those observable by intrapleural administration (36,88). In particular, 82.5% of the deceased rats treated by intraperitoneal injection died because of mesothelioma; only 14.3% of the deceased rats treated by intrapleural injection died due to mesothelioma induced by fibrous FE. The peritoneal tumors involved the abdominal cavity, with whitish and yellowish tissue on the surface of all the organs; furthermore a serosal effusion was present in almost all cases. Instead, the pleural tumors involved the visceral and/or parietal pleura and in 80% of the cases the diaphragm was largely involved with subsequent extension of the cancer into the peritoneal cavity (36).

Prismatic FE did not provoke mesothelioma in animal models. On the contrary, fibrous FE was found to cause a strong mesotheliomatogenic effect on the peritoneum and a milder extent on the pleura.

5. Ex vivo studies concerned with MM due to FE exposure

Environmental exposure of fibrous FE

Sheep lung is comparable to human lung in architecture, volume, and respiratory parameters (89); therefore, it is a suitable model for toxicological studies concerning exposure to environmental pollutants (89), such as asbestos (20,90,91) or FE fibers (43,83-87,92).

One of the first experimental study (43) that analyzed sheep exposed to FE fibers present in the surrounding environment of Biancavilla, demonstrated that the first pathological event seems to involve the alveolar epithelium, resulting in classic honeycombing (43,83), and subsequently the interstitial matrix (43,83,84). Matrix metalloproteinase (MMP)-13 is mainly overexpressed in fibroblasts and epithelial cells, while immunopositivity of TNF-related apoptosis-inducing ligand (TRAIL) and its receptor death receptor 5 (DR5) are detected on alveolar surfaces and in the vascular stroma. The triggering event at the level of type I pneumocytes seems to damage the cytoplasmic membrane, resulting in loss of cell elements and exposure of underlying capillaries, and eventually in a series of reactions including macrophage activation, possible release of growth factors, metaplasic reconstruction of lung alveoli, and fibrosis (43). Loreto et al (83) demonstrated epithelial and interstitial Bax overexpression and negative Bcl-2 immunoexpression. pRb overexpression was also detected in FE-exposed sheep lung, in particular in alveolar epithelium and the interstitium, while Rb expression was absent (84). Immunopositivity for TRAIL and MMP-13 receptor (43), the changes in Bax and Bcl-2(83), and the altered balance between Rb and pRb expression (84) can be considered a programmed response to protect the organism against uncontrolled cell proliferation, suggesting that apoptosis may be activated by FE fibers. A significant increase in the expression of CD68-positive macrophages, tryptase-positive mast cells, as well as a significant increase in microvascular density evaluated as CD31-positive areas in lung tissue of sheep exposed to FE fibers have been demonstrated. These data confirm the important role played by tumor microenvironment components in favor of angiogenesis in MM induced by FE exposure (85). Musumeci et al (92) investigated N-cadherin, ADAM-10 and aquaporin-1 (AQP1) expression in the lung tissue of sheep exposed to FE fibers, showing different patterns of immunolabeling. N-cadherin and ADAM-10 were more expressed in FE-exposed lung tissue, when compared with the control. On the contrary, AQP1 was more highly expressed in non-exposed lung tissue. These results suggest that N-cadherin, ADAM-10 and AQP1 are probably involved in different pathological processes induced by FE fiber exposure. The cellular and molecular toxicity mechanisms and the cellular response to FE fibers are still not well known, but these results highlight that molecules involved in carcinogenesis and in the inflammatory process participate in the network of events induced by exposure to FE fibers.

Loreto et al (86) demonstrated overexpression of MacroH2A.1, at the protein level, in lung epithelial cells and in lymph nodes of sheep exposed to FE fibers. The data suggest an involvement of MacroH2A.1 in the cellular response triggered by direct exposure to FE. The immunoreactions were detected in the areas where fibers were embedded and localized, in the lung, to the pulmonary and bronchial epithelium and not to the fibrotic interstitium. This significant research (86) seems to show a clear association between exposure to FE fibers and MacroH2A.1 expression view to identifying, in the future, a targeting epigenetics for cancer therapy.

Several studies have examined the lymph node draining pulmonary lobes of sheep grazing around Biancavilla (87,93). The results show a greater size of lymph nodes with signs of anthracosis. At the paracortical level, they show lymph-follicle hyperplasia with wide reactive corea and several macrophages containing grey-brownish particulate interspersed with elements with a fibril structure, forming nodules. Similar findings were detected in some peribronchiolar areas of the lung parenchyma. The FE fiber dimensions found in digested lymph nodes of sheep were similar to those found in the lung of a housewife from Biancavilla who died of MM as described by Paoletti et al (45). Indeed, sheep can be a biological indicator of environmental pollution by FE fibers, measuring these fibers in lymph node draining pulmonary lobes (87,93).

Several immunohistochemical investigations were conducted to demonstrate the implication of different compounds in MM due to FE fibers exposure and to investigate their potential role as diagnostic and prognostic markers. Angelico et al (94) demonstrated the prognostic role of AQP1 in FE-induced MM. In fact, the immunohistochemical overexpression of AQP1 was found to be associated with an increased median overall survival. Caltabiano et al (44) found high immunoexpression of Fb-3 in neoplastic cells with nuclear and cytoplasmic localization, demonstrating the implication of Fb-3 in MM due to FE exposure. Fb-3 could therefore have a potential role as a diagnostic and prognostic marker.

6. Discussion

The prevention of pathologies related to exposure to carcinogenic fibers such as asbestos and fluoro-edenite (FE), also includes the reduction of these fibers in the environment. Generally, this can be achieved in three ways: Reclamation, encapsulation and confinement. These interventions tend to eliminate airborne fibers to avoid exposure that can cause various diseases including cancer.

The observation of a significant incidence of MM, subsequently linked to the inhalation of FE fibers of Biancavilla, has been reported by epidemiological studies (45-48). Previous cross-sectional studies conducted on subjects exposed to FE fibers confirm the in vitro, in vivo and ex vivo data. In fact, it has been demonstrated that exposure to FE fibers may induce autoimmunity (95,96), and the involvement of the inflammosome (41).

This malignant cancer is a highly aggressive neoplasm of the serosal membranes lining the pleural cavity (97). Only 5% of MM patients are diagnosed at an early stage (98) and the median survival is approximately 6-12 months (97,99). Moreover, current treatment for MM, which is based on surgery and standard chemotherapy, has a modest effect on the overall survival (OS), which remains dismal (4).

The diagnosis of MM is always challenging as MM may appear in patients up to 30-40 years after exposure to carcinogenic fibers; the clinical and imaging signs of MM are non-specific; and a definitive diagnosis, which relies on histology, can sometimes be very difficult to achieve, even with the use of immunohistochemistry. To date, no single marker or panel of soluble biomarkers is available for a clear diagnosis of MM (100).

Many biomarkers have been proposed for screening and diagnosis of MM in subjects exposed, such as calretinin, cytokeratin 5 (CK5), podoplanin, mesothelin, osteopontin, hyaluronic acid, Fb-3(44), VEGF (101), AQP1(94), high mobility group box 1 (HMGB-1) (102), and MacroH2A.1(86). Mesothelin is the only Food and Drug Administration (FDA)-approved biomarker for MM (103-105), but with limitations (106). In fact, the poor sensitivity of mesothelin clearly limits the added value to the diagnosis of MM (100).

Some studies have been conducted to understand the link between common genetic variations in the molecular pathways and cancer risk with the final goal to develop novel therapeutic targets. Lim et al (107) reported mutations in SMO and SUFU and a novel multi-exonic deletion in PTCH1 in MM cell lines and tumors. These data suggest that aberrant activation of the Hedgehog (HH) signaling in MM is unlikely to be driven by mutations in the majority of tumors but instead activated through autocrine signaling (107,108). This pathway may represent a novel therapeutic target in MM for recently developed HH pathway inhibitors.

Several studies have demonstrated that microRNAs (miRNAs) may be used as valuable non-invasive diagnostic and prognostic biomarkers for various human diseases, including cancers (42,109,110). In the clinical setting, circulating cell-free miRNAs and fecal miRNAs are the main forms of RNA used as diagnostic biomarkers (111). In particular, a recent review of the literature by Ledda et al (112) indicates a list of miRNAs potentially involved in MM. Potential miRNA biomarkers for this malignant neoplasm include the following: miRNA-126-3p, miRNA-625-3p, miRNA-103a-3p, miRNA-16-5p, miRNA-143-3p, miRNA-145-5p, miRNA-192-5p, miRNA-193a-3p, miRNA-200b-3p, miRNA-203a-3p, and miRNA-652-3p. The scientific community has revealed that several miRNAs are involved in deregulation and in all molecular mechanisms associated with MM development (113,114) and constantly updates the miRNAs which can be associated with MM early diagnosis and prognosis.

In recent studies, several bioinformatics approaches to identify selected miRNAs highly deregulated in cancer samples when compared with normal control have been developed (77,115-118). The in silico study of the expression of certain miRNAs represents an effort in the field of biomarker discovery because in this way it is possible to analyze the data coming from multiple studies of miRNA profiling; in this way it is possible to have a large series of samples useful to obtain truthful expression data concerning miRNAs with a potential diagnostic and prognostic role in cancer. In addition, the development of new high-sensitivity technologies and the analysis of liquid biopsy samples and circulating tumor DNA are paving the way to new non-invasive validation studies aimed to discover new promising diagnostic and prognostic biomarkers for several pathologies, including MM (119-121).

Therefore, future studies will be conducted to understand the link between common genetic variations in the molecular pathways and cancer risk with the final goal to develop novel therapeutic targets. Research is needed in order to computationally select putative miRNAs involved in the development and progression of lung cancer or MM and to be validated in correspective in vitro tumor models and in a subset of patients chronically exposed to FE. It could also be particularly helpful to study and subsequently use a combination of several protein and molecular markers to improve diagnostic accuracy.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

All information included in this review has been documented by relevant references.

Authors' contributions

Conceptualization of the review was accomplished by VF and CL. The research methodology was designed and conducted by VF, EV and GB. Validation of the research data was conducted by VF and CL. Formal analysis was carried out by VF. Investigation of the data was carried out by VF, EV and GB. Data curation was conducted by VF, MPH and SC. Writing-original draft preparation was carried out by VF and CL. Writing-review and editing was accomplished by VF, MPH, SC and AS. Supervision was conducted by CL. All authors read and approved the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors report that they have no competing interests.

References

1 

Hansen J, De Klerk NH, Eccles JL, Musk AW and Hobbs MS: Malignant mesothelioma after environmental exposure to blue asbestos. Int J Cancer. 54:578–581. 1993.PubMed/NCBI View Article : Google Scholar

2 

Kanarek MS: Mesothelioma from chrysotile asbestos: Update. Ann Epidemiol. 21:688–697. 2011.PubMed/NCBI View Article : Google Scholar

3 

Bayram M, Dongel I, Bakan ND, Yalççn H, Cevit R, Dumortier P and Nemery B: High risk of malignant mesothelioma and pleural plaques in subjects born close to ophiolites. Chest. 143:164–171. 2013.PubMed/NCBI View Article : Google Scholar

4 

Carbone M, Kanodia S, Chao A, Miller A, Wali A, Weissman D, Adjei A, Baumann F, Boffetta P, Buck B, et al: Consensus report of the 2015 weinman international conference on mesothelioma. J Thorac Oncol. 11:1246–1262. 2016.PubMed/NCBI View Article : Google Scholar

5 

Case BW, Abraham JL, Meeker G, Pooley FD and Pinkerton KE: Applying definitions of ‘asbestos’ to environmental and ‘low-dose’ exposure levels and health effects, particularly malignant mesothelioma. J Toxicol Environ Health B Crit Rev. 14:3–39. 2011.PubMed/NCBI View Article : Google Scholar

6 

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.PubMed/NCBI View Article : Google Scholar

7 

Liu G, Cheresh P and Kamp DW: Molecular basis of asbestos-induced lung disease. Annu Rev Pathol. 8:161–187. 2013.PubMed/NCBI View Article : Google Scholar

8 

Ledda C, Pomara C, Bracci M, Mangano D, Ricceri V, Musumeci A, Ferrante M, Musumeci G, Loreto C, Fenga C, et al: Natural carcinogenic fiber and pleural plaques assessment in a general population: A cross-sectional study. Environ Res. 150:23–29. 2016.PubMed/NCBI View Article : Google Scholar

9 

Falzone L, Marconi A, Loreto C, Franco S, Spandidos DA and Libra M: Occupational exposure to carcinogens: Benzene, pesticides and fibers (Review). Mol Med Rep. 14:4467–4474. 2016.PubMed/NCBI View Article : Google Scholar

10 

Gangemi S, Rapisarda V, Minciullo PL, Di Pasquale G, Lombardo G, Valentino M and Fenga C: Circulating levels of interleukin-18 in asbestos-exposed workers. Toxicol Ind Health. 21:125–129. 2005.

11 

Aitio A, Cantor KP, Attfield MD, Demers PA, Fowler BA, Grandjean P, Fubini B, Hartwig A, Gérin M, et al: A review of human carcinogens: Arsenic, metals, fibres and dusts. Vol 100 C. IARC, Lyon, pp219-224, 2012.

12 

Travaglione S, Bruni B, Falzano I, Paoletti I and Fiorentini C: Effects of the new-identified amphibole FE in lung epithelial cells. Toxicol In Vitro. 17:547–552. 2003.

13 

Pikarsky E, Porat RM, Stein I, Abramovitch R, Admit S, Kasem S, Gutkovich-Pyest E, Urieli-Shoval S, Galun E and Ben-Neriah Y: NF-kappaB function as a tumor promoter in inflammation-associated cancer. Nature. 431:461–466. 2004.PubMed/NCBI View Article : Google Scholar

14 

Tokokuni S: Mechanisms of asbestos-induced carcinogenesis. Nagoya J Med Sci. 71:1–10. 2009.PubMed/NCBI

15 

Koskinen K, Rinne JP, Zitting A, Tossavainen A, Kivekäs J, Reijula K, Roto P and Huuskonen MS: Screening for asbestos-induced diseases in Finland. Am J Ind Med. 30:241–251. 1996.PubMed/NCBI View Article : Google Scholar

16 

Pan XL, Day HW, Wang W, Beckett LA and Schenker MB: Residential proximity to naturally occurring asbestos and mesothelioma risk in California. Am J Respir Crit Care Med. 172:1019–1025. 2005.PubMed/NCBI View Article : Google Scholar

17 

Luo S, Liu X, Mu S, Tsai SP and Wen CP: Asbestos related diseases from environmental exposure to crocidolite in Da-yao, China. I. Review of exposure and epidemiological data. Occup Environ Med. 60:35–41. 2003.PubMed/NCBI View Article : Google Scholar

18 

Baumann F, Maurizot P, Mangeas M, Ambrosi JP, Douwes J and Robineau B: Pleural mesothelioma in New Caledonia: Associations with environmental risk factors. Environ Health Perspect. 119:695–700. 2011.PubMed/NCBI View Article : Google Scholar

19 

Rey F, Boutin C, Steinbauer J, Viallat JR, Alessandroni P, Jutisz P, Di Giambattista D, Billon-Galland MA, Hereng P, Dumortier P, et al: Environmental pleural plaques in an asbestos exposed population of northeast Corsica. Eur Respir J. 6:978–982. 1993.PubMed/NCBI

20 

McConnochie K, Simonato L, Mavrides P, Christofides P, Pooley FD and Wagner JC: Mesothelioma in Cyprus: The role of tremolite. Thorax. 42:342–347. 1987.PubMed/NCBI View Article : Google Scholar

21 

Constantopoulos SH: Environmental mesothelioma associated with tremolite asbestos: Lessons from the experiences of Turkey, Greece, Corsica, New Caledonia and Cyprus. Regul Toxicol Pharmacol. 52 (1 Suppl):S110–S115. 2008.PubMed/NCBI View Article : Google Scholar

22 

Baris YI, Artvinli M, Sahin AA, Sebastien P and Gaudichet A: Diffuse lung fibrosis due fibrous zeolite (erionite) exposure. Eur J Respir Dis. 70:122–125. 1987.PubMed/NCBI

23 

Yazicioglu S, Ilçayto R, Balci K, Sayli BS and Yorulmaz B: Pļeural calcification, pleural mesotheliomas, and bronchial cancers caused by tremolite dust. Thorax. 35:564–569. 1980.PubMed/NCBI View Article : Google Scholar

24 

Dumortier P, Coplü L, de Maertelaer V, Emri S, Baris I and De Vuyst P: Assessment of environmental asbestos exposure in Turkey by bronchoalveolar lavage. Am J Respir Crit Care Med. 158:1815–1824. 1998.PubMed/NCBI View Article : Google Scholar

25 

Senyigit A, Dalgic A, Kavak O and Tanrikulu AC: Determination of environmental exposure to asbestos (tremolite) and mesothelioma risks in the southeastern region of Turkey. Arch Environ Health. 59:658–662. 2004.PubMed/NCBI View Article : Google Scholar

26 

Metintas M, Metintas S, Hillerdal G, Ucgun I, Erginel S, Alatas F and Yildirim H: Non malignant pleural lesions due to environmental exposure to asbestos: A field-based, cross-sectional study. Eur Respir J. 26:875–880. 2005.PubMed/NCBI View Article : Google Scholar

27 

Döngel I, Bayram M, Bakan ND, Yalçın H and Gültürk S: Is living close to ophiolites related to asbestos related diseases? Cross-sectional study. Respir Med. 107:870–874. 2013.PubMed/NCBI View Article : Google Scholar

28 

Kliment CR, Clemens K and Oury TD: North American erionite-associated mesothelioma with pleural plaques and pulmonary fibrosis: A case report. Int J Clin Exp Pathol. 2:407–410. 2009.PubMed/NCBI

29 

Baumann F, Buck BJ, Metcalf RV, McLaurin BT, Merkler DJ and Carbone M: The presence of asbestos in the natural environment is likely related to mesothelioma in young individuals and women from southern Nevada. J Thorac Oncol. 10:731–737. 2015.PubMed/NCBI View Article : Google Scholar

30 

Konen T, Johnson JE, Lindgren P and Williams A: Cancer incidence and mortality associated with non-occupational and low dose exposure to Libby vermiculite in Minnesota. Environ Res. 175:449–456. 2019.PubMed/NCBI View Article : Google Scholar

31 

Baur X: Review on the adverse health effects of asbestiform antigorite, a non-regulated asbestiform serpentine mineral. Am J Ind Med. 61:625–630. 2018.PubMed/NCBI View Article : Google Scholar

32 

Cardile V, Lombardo L, Belluso E, Panico A, Capella S and Balazy M: Toxicity and carcinogenicity mechanisms of fibrous antigorite. Int J Environ Res Public Health. 4:1–9. 2007.PubMed/NCBI View Article : Google Scholar

33 

Groppo C and Compagnoni R: Ubiquitous fibrous antigorite veins from the Lanzo Ultramafic Massif, Internal Western Alps (Italy): Characterisation and genetic conditions. Per Mineral. 76:169–181. 2007.

34 

FitzGerald J, Eggleton R and Keeling J: Antigorite from Rowland flat, South Australia: Asbestiform character. Eur J Mineral. 22:525–533. 2010.

35 

Fitzgerald SM and Harty EA: Antigorite: Is it the forgotten asbestos? Prof Saf. 59:43–48. 2014.

36 

Soffritti M, Minardi F, Bua L, Degli Esposti D and Belpoggi F: First experimental evidence of peritoneal and pleural mesotheliomas induced by FE fibres present in Etnean volcanic material from Biancavilla (Sicily, Italy). Eur J Oncol. 9:169–715. 2004.

37 

Gianfagna A and Oberti R: Fluoro-edenite from Biancavilla (Catania, Sicily, Italy): Crystal chemistry of a new amphibole end-member. Am Mineral. 86:1489–1493. 2001.

38 

Biggeri A, Pasetto R, Belli S, Bruno C, Di Maria G, Mastrantonio M, Trinca S, Uccelli R and Comba P: Mortality from chronic obstructive pulmonary disease and pleural mesothelioma in an area contaminated by natural fiber (fluoro-edenite). Scand J Work Environ Health. 30:249–252. 2004.PubMed/NCBI View Article : Google Scholar

39 

Comba P, Gianfagna A and Paoletti L: Pleural mesothelioma cases in Biancavilla are related to a new fluoro-edenite fibrous amphibole. Arch Environ Health. 58:229–232. 2003.PubMed/NCBI View Article : Google Scholar

40 

Gianfagna A, Ballirano P, Bellatreccia F, Bruni B, Paoletti L and Oberti R: Characterization of amphibole fibres linked to mesothelioma in the area of Biancavilla, Eastern Sicily, Italy. Mineral Mag. 67:1221–1229. 2003.

41 

Ledda C, Costa C, Matera S, Puglisi B, Costanzo V, Bracci M, Fenga C, Rapisarda V and Loreto C: Immunomodulatory effects in workers exposed to naturally occurring asbestos fibers. Mol Med Rep. 15:3372–3378. 2017.PubMed/NCBI View Article : Google Scholar

42 

Ledda C and Rapisarda V: Malignant pleural mesothelioma: The need to move from research to clinical practice. Arch Med Res. 47(407)2016.PubMed/NCBI View Article : Google Scholar

43 

Martinez G, Loreto C, Rapisarda V, Musumeci G, Valentino M and Carnazza ML: Effects of exposure to fluoro-edenite fibre pollution on the respiratory system: An in vivo model. Histol Histopathol. 21:595–601. 2006.PubMed/NCBI View Article : Google Scholar

44 

Caltabiano R, Loreto C, Vitale E, Matera S, Miozzi E, Migliore M, Angelico G, Tumino R, Ledda C and Rapisarda V: Fibulin-3 immunoexpression in malignant mesothelioma due to fluoro-edenite: A preliminary report. Future Oncol. 14 (6 Suppl):S53–S57. 2018.PubMed/NCBI View Article : Google Scholar

45 

Paoletti L, Batisti D, Bruno C, Di Paola M, Gianfagna A, Mastrantonio M, Nesti M and Comba P: Unusually high incidence of malignant pleural mesothelioma in a town of eastern Sicily: An epidemiological and environmental study. Arch Environ Health. 55:392–398. 2000.PubMed/NCBI View Article : Google Scholar

46 

Di Paola M, Mastrantonio M, Carboni M, Belli S, Grignoli M, Comba P and Nesti M: Mortality from malignant pleural neoplasms in Italy in the years 1988-1992. Rapporti ISTISAN, 96/40. Istituto Superiore di Sanità, Rome, 1996.

47 

Fazzo L, De Santis M, Minelli G, Bruno C, Zona A, Marinaccio A, Conti S and Comba P: Pleural mesothelioma mortality and asbestos exposure mapping in Italy. Am J Industr Med. 55:11–24. 2012.PubMed/NCBI View Article : Google Scholar

48 

Fazzo L, Minelli G, De Santis M, Bruno C, Zona A, Marinaccio A, Conti S, Pirastu R and Comba P: Mesothelioma mortality surveillance and asbestos exposure tracking in Italy. Ann Ist Super Sanita. 48:300–310. 2012.PubMed/NCBI View Article : Google Scholar

49 

Rapisarda V, Ledda C, Ricceri V, Arena F, Musumeci A, Marconi A, Fago L, Bracci M, Santarelli L and Ferrante M: Detection of pleural plaques in workers exposed to inhalation of natural fluoro-edenite fibres. Oncol Lett. 9:2046–2052. 2015.PubMed/NCBI View Article : Google Scholar

50 

Ledda C, Loreto C, Matera S, Massimino N, Cannizzaro E, Musumeci A, Migliore M, Fenga C, Pomara C and Rapisarda V: Early effects of fluoro-edenite: Correlation between IL-18 serum levels and pleural and parenchymal abnormalities. Future Oncol. 12 (23 Suppl):S59–S62. 2016.PubMed/NCBI View Article : Google Scholar

51 

Grosse Y, Loomis D, Guyton KZ, Lauby-Secretan B, El Ghissassi F, Bouvard V, Benbrahim-Tallaa L, Guha N, Scoccianti C, Mattock H and Straif K: Carcinogenicity of fluoro-edenite, silicon carbide fibres and whiskers, and carbon nanotubes. Lancet Oncol. 15:1427–1428. 2014.PubMed/NCBI View Article : Google Scholar

52 

Miller SA: PICO worksheet and search strategy. National Center for Dental Hygiene Research, 2001.

53 

Fenoglio I, Croce A, Di Renzo F, Tiozzo R and Fubini B: Pure-silica zeolites (Porosils) as model solids for the evaluation of the physicochemical features determining silica toxicity to macrophages. Chem Res Toxicol. 13:489–500. 2000.PubMed/NCBI View Article : Google Scholar

54 

Pugnaloni A, Lucarini G, Giantomassi F, Lombardo L, Capella S, Belluso E, Zizzi A, Panico AM, Biagini G and Cardile V: In vitro study of biofunctional indicators after exposure to asbestos-like fluoro-edenite fibres. Cell Mol Biol (Noisy-le-grand). 53 (Suppl):OL965–OL980. 2007.PubMed/NCBI

55 

Tremblay GM, Jordana M, Gauldie J and Särnstrand B: Fibroblasts as effector cells in fibrosis. In: Pulmonary fibrosis. Phan SH and Thrall RS (eds). Marcel Dekker Inc., New York, NY, pp541-577, 1995.

56 

Travaglione S, Bruni BM, Falzano L, Filippini P, Fabbri A, Paoletti L and Fiorentini C: Multinucleation and pro-inflammatory cytokine release promoted by fibrous fluoro-edenite in lung epithelial A549 cells. Toxicol in Vitro. 20:841–850. 2006.PubMed/NCBI View Article : Google Scholar

57 

Li XY, Lamb D and Donaldson K: The production of TNF-alpha and IL-1-like activity by bronchoalveolar leucocytes after intratracheal instillation of crocidolite asbestos. Int J Exp Pathol. 74:403–410. 1993.PubMed/NCBI

58 

Trimblin C, BeruBe K, Churg A, Driscoll K, Gordon T, Hemenway D, Walsh E, Cummins AB, Vacek P and Mossman B: Ambient particulate matter causes activation of the c-jun kinase/stress-activated protein kinase cascade and DNA synthesis in lung epithelial cells. Cancer Res. 58:4543–4547. 1998.PubMed/NCBI

59 

Hedges S, Svensson M, Agace W and Svanborg C: Cytokines induce an epithelial cell cytokine response. Adv Exp Med Biol. 371A:189–193. 1995.PubMed/NCBI View Article : Google Scholar

60 

Cardile V, Proietti L, Panico A and Lombardo L: Nitric oxide production in fluoro-edenite treated mouse monocyte-macrophage cultures. Oncol Rep. 12:1209–1215. 2004.PubMed/NCBI

61 

Hedenborg M and Klockars M: Production of reactive oxygen metabolites induced by asbestos fibres in human polymorphonuclear leucocytes. J Clin Pathol. 40:1189–1193. 1987.PubMed/NCBI View Article : Google Scholar

62 

Hamilton RF, Iyer LL and Holian A: Asbestos induces apoptosis in human alveolar macrophages. Am J Physiol. 271:L813–L819. 1996.PubMed/NCBI View Article : Google Scholar

63 

Loreto C, Carnazza ML, Cardile V, Libra M, Lombardo L, Malaponte G, Martinez G, Musumeci G, Papa V and Cocco L: Mineral fiber-mediated activation of phosphoinositide-specific phospholipase c in human bronchoalveolar carcinoma-derived alveolar epithelial A549 cells. Int J Oncol. 34:371–376. 2009.PubMed/NCBI

64 

Cardile V, Renis M, Scifo C, Lombardo L, Gulino R, Mancari B and Panico A: Behaviour of the new asbestos amphibole fluor-edenite in different lung cell systems. Int J Biochem Cell Biol. 36:849–860. 2004.PubMed/NCBI View Article : Google Scholar

65 

Pӓӓkkӧ P, Rämet M, Vähäkangas K, Korpela N, Soini Y, Turunen S, Jaworska M and Gillissen A: Crocidolite asbestos causes an induction of p53 and apoptosis in cultured A-549 lung carcinoma cells. Apoptosis. 3:203–212. 1998.PubMed/NCBI View Article : Google Scholar

66 

Yang CM, Luo SF, Wang CC, Chiu CT, Chien CS, Lin CC and Hsiao LD: Tumour necrosis factor-alpha- and interleukin-1beta-stimulated cell proliferation through activation of mitogen-activated protein kinase in canine tracheal smooth muscle cells. Br J Pharmac. 130:891–899. 2000.PubMed/NCBI View Article : Google Scholar

67 

Rana Rs and Hokin LE: Role of phosphoinositides in trans-membrane signaling. Physiol Rev. 70:115–164. 1990.PubMed/NCBI View Article : Google Scholar

68 

Lim Y, Kim SH, Kim KA, Oh MW and Lee KH: Involvement of protein kinase C, phospholipase C, and protein tyrosine kinase pathways in oxygen radical generation by asbestos-stimulated alveolar macrophage. Environ Health Perspect. 105 (Suppl 5):S1325–S1327. 1997.PubMed/NCBI View Article : Google Scholar

69 

Musumeci G, Cardile V, Fenga C, Caggia S and Loreto C: Mineral fibre toxicity: Expression of retinoblastoma (Rb) and phospho-retinoblastoma (pRb) protein in alveolar epithelial and mesothelial cell lines exposed to fluoro-edenite fibres. Cell Biol Toxicol. 27:217–225. 2011.PubMed/NCBI View Article : Google Scholar

70 

Beachy PA, Karhadkar SS and Berman DM: Tissue repair and stem cell renewal in carcinogenesis. Nature. 432:324–331. 2004.PubMed/NCBI View Article : Google Scholar

71 

Chen S, Guttrige DC, You Z, Zhang Z, Fribley A, Mayo MW, Kitajewski J and Wang CY: Wnt-1 signaling inhibits apoptosis by activating beta-catenin/T cell factor-mediated transcription. J Cell Biol. 152:87–96. 2001.PubMed/NCBI View Article : Google Scholar

72 

Zheng R, Yano S, Matsumori Y, Nakataki E, Muguruma H, Yoshizumi M and Sone S: SRC tyrosine kinase inhibitor, m475271, suppresses subcutaneous growth and production of lung metastasis via inhibition of proliferation, invasion, and vascularization of human lung adenocarcinoma cells. Clin Exp Metastasis. 22:195–204. 2005.PubMed/NCBI View Article : Google Scholar

73 

Yamanouchi H, Furihata M, Fujita JJ, Murakami H, Yoshinouchi T, Takahara J and Ohtsuki Y: Expression of cyclin E and cyclin D1 in non-small cell lung cancers. Lung Cancer. 31:3–8. 2001.PubMed/NCBI View Article : Google Scholar

74 

Rapisarda V, Salemi R, Marconi A, Loreto C, Graziano AC, Cardile V, Basile MS, Candido S, Falzone L, Spandidos DA, et al: Fluoro-edenite induces fibulin-3 overexpression in non-malignant human mesothelial cells. Oncol Lett. 12:3363–3367. 2016.PubMed/NCBI View Article : Google Scholar

75 

Chu IM, Hengst L and Slingerland JM: The Cdk inhibitor p27 in human cancer: Prognostic potential and relevance to anticancer therapy. Nat Rev Cancer. 8:253–267. 2008.PubMed/NCBI View Article : Google Scholar

76 

Rapisarda V, Caltabiano R, Musumeci G, Castrogiovanni P, Ferrante M, Ledda C, Lombardo C, Graziano ACE, Cardile V and Loreto C: Analysis of fibulin-3 after exposure to asbestos-like fibers. Environ Res. 156:381–387. 2017.PubMed/NCBI View Article : Google Scholar

77 

Filetti V, Falzone L, Rapisarda V, Caltabiano R, Graziano ACE, Ledda C and Loreto C: Modulation of microRNA expression levels after naturally occurring asbestiform fibers exposure as a diagnostic biomarker of mesothelial neoplastic transformation. Ecotoxicol Environ Saf. 198(110640)2020.PubMed/NCBI View Article : Google Scholar

78 

Cardile V, Lombardo L, Belluso E, Panico A, Renis M, Gianfagna A and Balazy M: Fluoro-edenite fibers induce expression of Hsp70 and inflammatory response. Int J Environ Res Public Health. 4:195–202. 2007.PubMed/NCBI View Article : Google Scholar

79 

Shukla A, Gulumian M, Hei TK, Kamp D, Rahman Q and Mossman B: Multiple roles of oxidants in the pathogenesis of asbestos-induced diseases. Free radical Biol Med. 34:1117–1129. 2003.PubMed/NCBI View Article : Google Scholar

80 

Holian A, Kelley K and Hamilton RF Jr: Mechanisms associated with human alveolar macrophage stimulation by particulates. Environ Health Perspect. 102 (Suppl 10):S69–S74. 1994.PubMed/NCBI View Article : Google Scholar

81 

De Witt DL: Prostaglandin endoperoxide synthase: Regulation of enzyme expression. Biochim Byophys Acta. 1083:121–134. 1991.PubMed/NCBI View Article : Google Scholar

82 

DeNardo P, Bruni B, Paoletti L, Pasetto R and Sirianni B: Pulmonary fibre burden in sheep living in the Biancavilla area (Sicily): Preliminary results. Sci Total Environ. 325:51–58. 2004.PubMed/NCBI View Article : Google Scholar

83 

Loreto C, Rapisarda V, Carnazza ML, Musumeci G, Valentino M, Fenga C and Martinez G: Fluoro-edenite fibres induce lung cell apoptosis: An in vivo study. Histol Histopathol. 23:319–326. 2008.PubMed/NCBI View Article : Google Scholar

84 

Musumeci G, Loreto C, Cardile V, Carnazza ML and Martinez G: Immunohistochemical expression of retinoblastoma and phospho-retinoblastoma protein in sheep lung exposed to fluoro-edenite fibers. Anat Sci Int. 85:74–78. 2010.PubMed/NCBI View Article : Google Scholar

85 

Musumeci G, Loreto C, Giunta S, Rapisarda V, Szychlinska MA, Imbesi R, Castorina A, Annese T, Castorina S, Castrogiovanni P and Ribatti D: Angiogenesis correlates with macrophage and mast cell infiltration in lung tissue of animals exposed to fluoro-edenite fibers. Exp Cell Res. 346:91–98. 2016.PubMed/NCBI View Article : Google Scholar

86 

Loreto C, Lombardo C, Caltabiano R, Ledda C, Hagnas M, Filetti V and Rapisarda V: An in vivo immunohistochemical study on MacroH2A.1 in lung and lymph-node tissues exposed to an asbestiform fiber. Curr Mol Med: Feb 20, 2020 (Epub ahead of print).

87 

Ledda C, Loreto C, Pomara C, Rapisarda G, Fiore M, Ferrante M, Bracci M, Santarelli L, Fenga C and Venerando R: Sheep lymph-nodes as a biological indicator of environmental exposure to fluoro-edenite. Environ Res. 147:97–101. 2016.PubMed/NCBI View Article : Google Scholar

88 

Belpoggi F, Tibaldi E, Lauriola M, Bua L, Falcioni L, Chiozzotto D, Manservisi F, Manservigi M and Soffritti M: The efficacy of long-term bioassays in predicting human risks: Mesotheliomas induced by fluoro-edenite fibres present in lava stone from the Etna volcano in Biancavilla, Italy. Eur J Oncol. 16:185–196. 2011.

89 

Begin R, Rola-Pleszczynski M, Sirois P, Masse S, Nadeau D and Bureau MA: Sequential analysis of the bronchoalveolar milieu in conscious sheep. J Appl Physiol Resp Envir Exerc Physiol. 50:665–671. 1981.PubMed/NCBI View Article : Google Scholar

90 

Schlesinger RB: Clearance from the respiratory tract. Fundam Appl Toxicol. 5:435–450. 1985.PubMed/NCBI View Article : Google Scholar

91 

Dumortier P, Rey F, Viallat JR, Broucke I, Boutin C and De Vuyst P: Chrysotile and tremolite asbestos fibres in the lungs and parietal pleura of Corsican goats. Occup Env Med. 59:643–646. 2002.PubMed/NCBI View Article : Google Scholar

92 

Musumeci G, Loreto C, Szychlinska MA, Imbesi R, Rapisarda V, Aiello FC, Castorina S and Castrogiovanni P: N-Cadherin, ADAM-10 and Aquaporin 1 expression in lung tissue exposed to fluoro-edenite fibers: An immunohistochemical study. Histol Histopathol. 30:987–999. 2015.PubMed/NCBI View Article : Google Scholar

93 

Rapisarda V, Rapisarda G, Vico CD, Gobbi L, Loreto C and Valentino M: Monitoring of fluoro-edenite fibre pollution through the study of sheep lymph nodes as a model of a biological indicator. Occup Environ Med. 62(656)2005.PubMed/NCBI View Article : Google Scholar

94 

Angelico G, Caltabiano R, Loreto C, Ieni A, Tuccari G, Ledda C and Rapisarda V: Immunohistochemical expression of aquaporin-1 in fluoro-edenite-induced malignant mesothelioma: A preliminary report. Int J Mol Sci. 19(685)2018.PubMed/NCBI View Article : Google Scholar

95 

Ledda C, Caltabiano R, Loreto C, Cinà D, Senia P, Musumeci A, Ricceri V, Pomara C and Rapisarda V: Prevalence of anti-nuclear autoantibodies in subjects exposed to natural asbestiform fibers: A cross-sectional study. J Immunotoxicol. 15:24–28. 2018.PubMed/NCBI View Article : Google Scholar

96 

Rapisarda V, Loreto C, Castorina S, Romano G, Garozzo SF, Musumeci A, Migliore M, Avola R, Cinà D, Pomara C and Ledda C: Occupational exposure to fluoro-edenite and prevalence of anti-nuclear autoantibodies. Future Oncol. 14 (6 Suppl):S59–S62. 2018.PubMed/NCBI View Article : Google Scholar

97 

Baas P, Fennell D, Kerr KM, van Schil PE, Haas RL and Peters S: ESMO Guidelines Committee. Malignant pleural mesothelioma: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 26 (Suppl 5):v31–v39. 2015.PubMed/NCBI View Article : Google Scholar

98 

Ying S, Jiang Z, He X, Yu M, Chen R, Chen J, Ru G, Chen Y, Chen W, Zhu L, et al: Serum HMGB1 as a potential biomarker for patients with asbestos-related diseases. Dis Markers. 2017(5756102)2017.PubMed/NCBI View Article : Google Scholar

99 

Tomasson K, Gudmundsson G, Briem H and Rafnsson V: Malignant mesothelioma incidence by nation-wide cancer registry: A population-based study. J Occup Med Toxicol. 11(37)2016.PubMed/NCBI View Article : Google Scholar

100 

Cui A, Jin XG, Zhai K, Tong ZH and Shi HZ: Diagnostic values of soluble mesothelin-related peptides for malignant pleural mesothelioma: Updated meta-analysis. BMJ Open. 4(e004145)2014.PubMed/NCBI View Article : Google Scholar

101 

Arnold DT and Maskell NA: Biomarkers in mesothelioma. Ann Clin Biochem. 55:49–58. 2018.PubMed/NCBI View Article : Google Scholar

102 

Wu T, Zhang W, Yang G, Li H, Chen Q, Song R and Zhao L: HMGB1 overexpression as a prognostic factor for survival in cancer: A meta-analysis and systematic review. Oncotarget. 7:50417–50427. 2016.PubMed/NCBI View Article : Google Scholar

103 

Creaney J, Sneddon S, Dick IM, Dare H, Boudville N, Musk AW, Skates SJ and Robinson BW: Comparison of the diagnostic accuracy of the MSLN gene products, mesothelin and megakaryocyte potentiating factor, as biomarkers for mesothelioma in pleural effusions and serum. Dis Markers. 35:119–127. 2013.PubMed/NCBI View Article : Google Scholar

104 

Creaney J, Dick IM, Meniawy TM, Leong SL, Leon JS, Demelker Y, Segal A, Musk AW, Lee YC, Skates SJ, et al: Comparison of fibulin-3 and mesothelin as markers in malignant mesothelioma. Thorax. 69:895–902. 2014.PubMed/NCBI View Article : Google Scholar

105 

Chen Z, Gaudino G, Pass HI, Carbone M and Yang H: Diagnostic and prognostic biomarkers for malignant mesothelioma: An update. Transl Lung Cancer Res. 6:259–269. 2017.PubMed/NCBI View Article : Google Scholar

106 

Boudville N, Paul R, Robinson BW and Creaney J: Mesothelin and kidney function-analysis of relationship and implications for mesothelioma screening. Lung Cancer. 73:320–324. 2011.PubMed/NCBI View Article : Google Scholar

107 

Lim CB, Prêle CM, Cheah HM, Cheng YY, Klebe S, Reid G, Watkins DN, Baltic S, Thompson PJ and Mutsaers SE: Mutational analysis of hedgehog signaling pathway genes in human malignant mesothelioma. PLoS One. 8(e66685)2013.PubMed/NCBI View Article : Google Scholar

108 

Shi Y, Moura U, Opitz I, Soltermann A, Rehrauer H, Thies S, Weder W, Stahel RA and Felley-Bosco E: Role of hedgehog signaling in malignant pleural mesothelioma. Clin Cancer Res. 18:4646–4656. 2012.PubMed/NCBI View Article : Google Scholar

109 

Candido S, Lupo G, Pennisi M, Basile MS, Anfuso CD, Petralia MC, Gattuso G, Vivarelli S, Spandidos DA, Libra M and Falzone L: The analysis of miRNA expression profiling datasets reveals inverse microRNA patterns in glioblastoma and Alzheimer's disease. Oncol Rep. 42:911–922. 2019.PubMed/NCBI View Article : Google Scholar

110 

Falzone L, Romano GL, Salemi R, Bucolo C, Tomasello B, Lupo G, Anfuso CD, Spandidos DA, Libra M and Candido S: Prognostic significance of deregulated microRNAs in uveal melanomas. Mol Med Rep. 19:2599–2610. 2019.PubMed/NCBI View Article : Google Scholar

111 

Nikolouzakis TK, Vassilopoulou L, Fragkiadaki P, Mariolis Sapsakos T, Papadakis GZ, Spandidos DA, Tsatsakis AM and Tsiaoussis J: Improving diagnosis, prognosis and prediction by using biomarkers in CRC patients (Review). Oncol Rep. 39:2455–2472. 2018.PubMed/NCBI View Article : Google Scholar

112 

Ledda C, Senia P and Rapisarda V: Biomarkers for early diagnosis and prognosis of malignant pleural mesothelioma: The quest goes on. Cancers (Basel). 10(203)2018.PubMed/NCBI View Article : Google Scholar

113 

De Santi C, Melaiu O, Bonotti A, Cascione L, Di Leva G, Foddis R, Cristaudo A, Lucchi M, Mora M, Truini A, et al: Deregulation of miRNAs in malignant pleural mesothelioma is associated with prognosis and suggests an alteration of cell metabolism. Sci Rep. 7(3140)2017.PubMed/NCBI View Article : Google Scholar

114 

Martínez-Rivera V, Negrete-García MC, Ávila-Moreno F and Ortiz-Quintero B: Secreted and tissue miRNAs as diagnosis biomarkers of malignant pleural mesothelioma. Int J Mol Sci. 19(595)2018.PubMed/NCBI View Article : Google Scholar

115 

Falzone L, Candido S, Salemi R, Basile MS, Scalisi A, McCubrey JA, Torino F, Signorelli SS and Montella M: Computational identification of microRNAs associated to both epithelial to mesenchymal transition and NGAL/MMP-9 pathways in bladder cancer. Oncotarget. 7:72758–72766. 2016.PubMed/NCBI View Article : Google Scholar

116 

Falzone L, Scola L, Zanghì A, Biondi A, Di Cataldo A, Libra M and Candido S: Integrated analysis of colorectal cancer microRNA datasets: Identification of microRNAs associated with tumor development. Aging (Albany NY). 10:1000–1014. 2018.PubMed/NCBI View Article : Google Scholar

117 

Hafsi S, Candido S, Maestro R, Falzone L, Soua Z, Bonavida B, Spandidos DA and Libra M: Correlation between the overexpression of Yin Yang 1 and the expression levels of miRNAs in Burkitt's lymphoma: A computational study. Oncol Lett. 11:1021–1025. 2016.PubMed/NCBI View Article : Google Scholar

118 

Polo A, Crispo A, Cerino P, Falzone L, Candido S, Giudice A, De Petro G, Ciliberto G, Montella M, Budillon A and Costantini S: Environment and bladder cancer: Molecular analysis by interaction networks. Oncotarget. 8:65240–65252. 2017.PubMed/NCBI View Article : Google Scholar

119 

Salemi R, Falzone L, Madonna G, Polesel J, Cinà D, Mallardo D, Ascierto PA, Libra M and Candido S: MMP-9 as a candidate marker of response to BRAF inhibitors in melanoma patients with BRAFV600E mutation detected in circulating-free DNA. Front Pharmacol. 9(856)2018.PubMed/NCBI View Article : Google Scholar

120 

Cavallari I, Urso L, Sharova E, Pasello G and Ciminale V: Liquid biopsy in malignant pleural mesothelioma: State of the art, pitfalls, and perspectives. Front Oncol. 9(740)2019.PubMed/NCBI View Article : Google Scholar

121 

Tuaeva NO, Falzone L, Porozov YB, Nosyrev AE, Trukhan VM, Kovatsi L, Spandidos DA, Drakoulis N, Kalogeraki A, Mamoulakis C, et al: Translational application of circulating DNA in oncology: Review of the last decades achievements. Cells. 8(1251)2019.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

December-2020
Volume 13 Issue 6

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Filetti V, Vitale E, Broggi G, Hagnäs MP, Candido S, Spina A and Lombardo C: Update of in&nbsp;vitro, in&nbsp;vivo and ex&nbsp;vivo fluoro‑edenite effects on malignant mesothelioma: A systematic review (Review). Biomed Rep 13: 60, 2020.
APA
Filetti, V., Vitale, E., Broggi, G., Hagnäs, M.P., Candido, S., Spina, A., & Lombardo, C. (2020). Update of in&nbsp;vitro, in&nbsp;vivo and ex&nbsp;vivo fluoro‑edenite effects on malignant mesothelioma: A systematic review (Review). Biomedical Reports, 13, 60. https://doi.org/10.3892/br.2020.1367
MLA
Filetti, V., Vitale, E., Broggi, G., Hagnäs, M. P., Candido, S., Spina, A., Lombardo, C."Update of in&nbsp;vitro, in&nbsp;vivo and ex&nbsp;vivo fluoro‑edenite effects on malignant mesothelioma: A systematic review (Review)". Biomedical Reports 13.6 (2020): 60.
Chicago
Filetti, V., Vitale, E., Broggi, G., Hagnäs, M. P., Candido, S., Spina, A., Lombardo, C."Update of in&nbsp;vitro, in&nbsp;vivo and ex&nbsp;vivo fluoro‑edenite effects on malignant mesothelioma: A systematic review (Review)". Biomedical Reports 13, no. 6 (2020): 60. https://doi.org/10.3892/br.2020.1367