Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

Journal of Toxicology and Environmental Health, Part A

Current Issues

ISSN: 1528-7394 (Print) 1087-2620 (Online) Journal homepage: https://www.tandfonline.com/loi/uteh20

Pleural mesothelioma and lung cancer: the role of


asbestos exposure and genetic variants in selected
iron metabolism and inflammation genes

F. Celsi, S. Crovella, R. R. Moura, M. Schneider, F. Vita, L. Finotto, G. Zabucchi,


P. Zacchi & V. Borelli

To cite this article: F. Celsi, S. Crovella, R. R. Moura, M. Schneider, F. Vita, L. Finotto, G.


Zabucchi, P. Zacchi & V. Borelli (2019): Pleural mesothelioma and lung cancer: the role of asbestos
exposure and genetic variants in selected iron metabolism and inflammation genes, Journal of
Toxicology and Environmental Health, Part A, DOI: 10.1080/15287394.2019.1694612

To link to this article: https://doi.org/10.1080/15287394.2019.1694612

View supplementary material

Published online: 22 Nov 2019.

Submit your article to this journal

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=uteh20
JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH, PART A
https://doi.org/10.1080/15287394.2019.1694612

Pleural mesothelioma and lung cancer: the role of asbestos exposure and
genetic variants in selected iron metabolism and inflammation genes
F. Celsia,b, S. Crovellac,d, R. R. Mourae, M. Schneiderf, F. Vitaf, L. Finottog, G. Zabucchib, P. Zacchib, and V. Borellib
a
Lega Italiana per la Lotta contro i Tumori (LILT), Italy; bDepartment of Life Sciences, University of Trieste, Trieste, Italy; cInstitute for Maternal
and Child Health, IRCCS Burlo Garofolo, Trieste, Italy; dDepartment of Medical, Surgical and Health Sciences, University of Trieste, Ospedale di
Cattinara, Trieste, Italy; eDepartment of Genetics, Federal University of Pernambuco, Recife, Brazil; fLaboratory of Pathological Anatomy,
AAS2 “Bassa Friulana-Isontina” - S. Polo General Hospital, Monfalcone, Italy; gWorkplace Safety and Prevention, AAS2 “Bassa Friulana-
Isontina” - S. Polo General Hospital, Monfalcone, Italy

ABSTRACT KEYWORDS
Two of the major cancerous diseases associated with asbestos exposure are malignant pleural Mesothelioma; lung cancer;
mesothelioma (MPM) and lung cancer (LC). In addition to asbestos exposure, genetic factors have formalin-fixed and paraffin-
been suggested to be associated with asbestos-related carcinogenesis and lung genotoxicity. embedded tissue samples;
inflammasome; hephaestin;
While genetic factors involved in the susceptibility to MPM were reported, to date the influence of
iron; polymorphisms;
individual genetic variations on asbestos-related lung cancer risk is still poorly understood. Since asbestos exposure
inflammation and disruption of iron (Fe) homeostasis are hallmarks of asbestos exposure affecting
the pulmonary tissue, this study aimed at investigating the association between Fe-metabolism
and inflammasome gene variants and susceptibility to develop LC or MPM, by comparing an
asbestos-exposed population affected by LC with an “asbestos-resistant exposed population”.
A retrospective approach similar to our previous autopsy-based pilot study was employed in
a novel cohort of autoptic samples, thus giving us the possibility to corroborate previous findings
obtained on MPM by repeating the analysis in a novel cohort of autoptic samples. The protective
role of HEPH coding SNP was further confirmed. In addition, the two non-coding SNPs, either in
FTH1 or in TF, emerged to exert a similar protective role in a new cohort of LC exposed individuals
from the same geographic area of MPM subjects. No association was found between NLRP1 and
NLRP3 polymorphisms with susceptibility to develop MPM and LC. Further research into a specific
MPM and LC “genetic signature” may be needed to broaden our knowledge of the genetic
landscape attributed to result in MPM and LC.

Introduction candidate genes approaches (Betti et al. 2017; Borelli


et al. 2015; Christiani 2000; Crovella et al. 2016, 2018;
Malignant pleural mesothelioma (MPM) and lung
Dianzani et al. 2006; Gemignani et al. 2009; Girardelli
cancer (LC) represent two types of diseases asso-
et al. 2012; Hirvonen et al. 1996; Landi et al. 2007;
ciated with asbestos exposure (Andujar et al. 2016;
London et al. 1995; Murakami et al. 2012; Neri et al.
Gilham et al. 2016; Lemen 2016). Although the
2008; Schabath et al. 2002; Schneider and Bernges
mechanisms underlying lung and pleural cell
2009; Tunesi et al. 2015; Wang, Neuberg, and
injury are not completely known, exposure to
Christiani 2004) or whole-genome association studies
these fibrous minerals appears not to be the sole
for both MPM and LC (Cadby et al. 2013; Kettunen
cause responsible for the development of these
et al. 2017; Matullo et al. 2013; Wei et al. 2012). While
debilitating diseases (Kamp and Weitzman 1999;
low- (Cadby et al. 2013; Matullo et al. 2013) and high-
Mossman 1990). In the last decade, many studies
risk (Betti et al. 2017; Ohar et al. 2016) genetic factors
unveiled that genetic factors are also involved in
for MPM were noted, defining germline variants in
promoting asbestos-related carcinogenesis and
BAP1 tumor suppressor gene as high-risk factor is still
lung genotoxicity.
not conclusive (Carbone et al. 2019, 2012, 2013; Ohar
A number of studies compared past asbestos expo-
et al. 2016); thus, the impact of individual genetic
sure and genetic polymorphisms using either

CONTACT V. Borelli borelliv@units.it Department of Life Sciences, University of Trieste, Trieste, Italy
Color versions of one or more of the figures in the article can be found online at www.tandfonline.com/uteh.
Supplemental data for this article can be accessed here.
© 2019 Taylor & Francis
2 F. CELSI ET AL.

variations exerts on asbestos-related lung cancer risk initiating the inflammatory response (Martinon,
remains poorly understood. Burns, and Tschopp 2002), and susceptibility to
Candidate gene approach studies demonstrated MPM development (Borelli et al. 2015; Crovella
that polymorphisms in genes encoding for xenobio- et al. 2016).
tic metabolizing enzymes such as GSTM1, GSTT1, Since inflammation and Fe homeostasis disrup-
MPO, CYP1A1 and CYP2E1, and manganese super- tion are both hallmarks of lung damage attributed
oxide dismutase (SOD2) are associated with asbes- to asbestos exposure, the aim of this study was to
tos-related lung cancer risk (Schabath et al. 2002; examine the possible association between Fe-
Schneider and Bernges 2009). Although recent gen- metabolism and inflammasome gene variants and
ome-wide association studies (GWAS) indicated risk of development of LC by comparing an asbes-
novel loci for lung cancer risk, few investigators tos-exposed LC affected cohort with an “asbestos-
addressed genome–environment interactions. In resistant exposed population”. A retrospective
particular, Wei et al. (2012) suggested that immune approach was employed relying on our autopsy-
function regulation-related pathways (Fas pathway) based pilot study.
might be mechanistically involved in asbestos-
associated lung cancer risk. Liu et al. (2015) pro-
posed MIRLET7BHG (MicroRNA Let-7b Host Materials and methods
Gene) as a possible important predictive marker for
asbestos exposure-related lung cancer. Finally, A population-based autopsy study
Kettunen et al. (2017) identified novel DNA methy- Our tissue samples originated from Monfalcone area
lation changes associated with lung tumors and as described previously by Crovella et al. (2016). This
asbestos exposure. area is characterized by high incidence of asbestos-
Lung inflammation and disruption of iron (Fe) related mesothelioma (Bianchi et al. 1993) and by
homeostasis were observed both in animal models frequent presence of pleural plaques observed after
and in patients affected by asbestos-related lung dis- the necroscopic examination (Bianchi et al. 1991).
ease (Dostert et al. 2008; Ghio et al. 2008; Ghio, Tissue samples in this study were obtained from
Pavlisko, and Roggli 2015; Jiang et al. 2012; autopsies performed between 1980 and 2015 on
Mossman 1990; Mossman and Churg 1998; asbestos-exposed subjects. All necroscopic examina-
Mossman et al. 2013) and are considered important tions were conducted at Monfalcone Hospital and
mechanisms underlying pulmonary toxicity induced were initially reviewed for signs of asbestos exposure
by asbestos (Ather et al. 2014; Aust, Cook, and and then for asbestos-related neoplasms (pleural
Dodson 2011; Aust et al. 2000; Chew and Toyokuni mesothelioma and lung cancer) or for the absence
2015; Ghio et al. 2008; Ghio, Pavlisko, and Roggli of asbestos-related diseases to select a control popu-
2015; Liu et al. 2015; Shannahan et al. 2011). lation (see below). Asbestos exposure was objectively
Previously the association between selected Fe- established for all autopsies by evaluation, during the
metabolism and inflammation-associated genes necroscopic examination, of the presence of pleural
with susceptibility to develop MPM was examined plaques and/or Asbestos Bodies (AB) in routine lung
in a highly selected asbestos-exposed population sections obtained at necropsy. Pleural plaques were
employing postmortem paraffin-embedded tissues examined and classified in three stages, as previously
(Borelli et al. 2015; Crovella et al. 2016, 2018). described (Crovella et al. 2016). Asbestos bodies
Three SNPs, localized in the ferritin heavy chain, were also quantified, using the Smith–Naylor
transferrin, and hephaestin genes, resulted in protec- method (Smith and Naylor 1972), with AB lung
tion against the development of MPM. No marked burden expressed as the number of AB per gram
associations were found, however, between poly- dry lung tissue. A documented occupational history
morphisms in inflammation-associated genes: of asbestos exposure was collected for the majority of
NACHT, LRR, FIIND, CARD domain, and PYD subjects. On the basis of occupational history, indi-
domains-containing protein 1 and 3 (NLRP1 and viduals were subdivided into five categories: 1) mar-
NLRP3), both constituents of a multiprotein oligo- itime, having worked on merchant ships; 2) builder,
mer called “inflammasome” and responsible for being involved in construction/refurbishment of
JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH, PART A 3

houses/industrial warehouse; 3) shipyard, being Table 1. Characteristics of selected asbestos-exposed indivi-


employed either directly or indirectly in shipbuilding duals which developed pleural mesothelioma (AEM) in monfal-
cone area (2009–2015).
on Monfalcone shipyard; 4) domestic (take-home
AEM
exposure), through laundering work clothes of All cases, n 52
family members employed; 5) other, not pertaining Mean age (years ± S.D.) 74.2 ± 8.8
to aforementioned categories. Considering the dif- Gender, n(%)
Female 6 (11.6%)
ferent occupational histories, the potential exists for Male 46 (88.4%)
mixed asbestos exposure (chrysotile and Histology, n(%)
Epithelioid Mesothelioma 23 (44.2%)
amphiboles). Sarcomatoid Mesothelioma 10 (19.2%)
Biphasic Mesothelioma 16 (30.8%)
Study population: individuals exposed to asbestos N.A. 3 (5.8%)
Asbestos exposure (Asbestos bodies counts)
who developed mesothelioma or lung cancer (n/g dry tissue)
Autopsies were performed between 2009 and 2015. 0–999 9 (17.2%)
1000–9999 17 (33.2%)
Inclusion criteria were: subjects who presented objec- 10.000–99.999 14 (26.8%)
tive signs of asbestos exposure (presence of AB and/or 100.000–1.000.000 6 (11.4%)
pleural plaques during routine lung sections examina- N.A. 6 (11.4%)
Ialine plaques, n(%)
tion) and developed either pleural mesothelioma Absent 10 (19.2%)
(Asbestos Exposed with Mesothelioma = AEM, n = Grade 1 11 (21.2%)
Grade 2 26 (50%)
52) or lung cancer (Asbestos Exposed with Lung Grade 3 5 (9.6%)
Cancer = AELC, n = 57); subjects’ characteristics are Occupation type, n(%)
summarized in Tables 1 and 2. Diagnosis of pleural Maritime 4 (7.69%)
Builder 3 (5.77%)
mesothelioma or lung cancer was confirmed or Shipyard 34 (64.38%)
obtained during necropsy and assessed by histological Domestic 4 (7.69%)
Other 4 (7.69%)
examination. Information on histological type of neo- N.A. 3 (5.77%)
plasia was collected for each patient both mesothe-
lioma or lung cancer and subsequently, patients were
subdivided into categories based upon this informa- pleural plaques and/or of AB during routine lung
tion. The majority of malignant pleural mesothelioma sections examination), who did not develop asbestos-
cases were differentiated into three different histologi- related diseases (specifically mesothelioma or lung
cal types: 1) epithelioid, possessing mostly cells with cancer and also other types of asbestos-induced
epithelial morphology; 2) sarcomatoid, with cells hav- tumors, such as laryngeal, gastrointestinal and ovarian
ing a spindle morphology; 3) biphasic, with cells cancer (International Agency for Research on Cancer
belonging to both categories (spindle and epithelioid) 2012)) and died of other causes after 75 years of age
(Inai 2008). The majority of lung cancers were classi- (Table 3). The threshold of 75 years of age was
fied into two large histological categories: small cell selected on the basis of the latency period for asbestos-
lung cancer or non-small cell lung cancer, further related diseases, occupational exposure and age at the
subdivided into adenocarcinoma, squamous carci- time of death, which hinged on previously reported
noma, and large cell carcinoma (Travis et al. 2015). data from a series of mesothelioma cases in the same
In AELC patients, information regarding smoking geographical area (Bianchi e Bianchi, 2007). The study
status was obtained and used to classify subjects in was approved by the regional ethical committee for
nonsmokers (N) or smokers (S). Friuli Venezia Giulia.

Control population: individuals exposed to Postmortem samples


asbestos without mesothelioma or lung cancer
The archives of the Department of Pathological
development (AE)
Anatomy of Monfalcone Hospital stored the histo-
Autopsies were performed between 1980 and 2000. logical samples from all autopsies. Myocardial tissue
Inclusion criteria were: subjects (n = 48) presenting was chosen as the starting material for DNA extrac-
objective signs of asbestos exposure (presence of tion, selected to be free from neoplastic cells and thus
4 F. CELSI ET AL.

Table 2. Characteristics of selected asbestos-exposed indivi- Table 3. Characteristics of selected asbestos-exposed indivi-
duals which developed lung cancer (AELC) in monfalcone area duals which did not develop asbestos-related disease (AE) in
(2009–2015). monfalcone area (1980–2000).
AELC AEM
All cases, n 57 All cases, n 48
Mean age (years ± S.D.) 73.9 ± 8.7 Mean age (years ± S.D.) 80.1 ± 0.6
Gender, n(%) Gender, n(%)
Female 0 (0%) Female 3 (6.25%)
Male 57 (100%) Male 45 (93.75%)
Histology, n(%) Death causes, n(%)
Adenocarcinoma 14 (24.6%) Non-tumoral 32 (66.7%)
Squamous cell carcinoma 7 (12.3)% Atherosclerosis 19 (39.6%)
Small cell carcinoma 3 (5.3%) Other 13 (27.1%)
Large cell carcinoma 4 (7.0%) Tumoral 16 (33.3%)
N.A. 29 (50.8%) Liver cancer 6 (12.5%)
Smoking Status, n (%) Other 10 (20.8%)
Smokers (S) 46 (80.7%) Asbestos exposure (Asbestos bodies counts)
Nonsmokers (N) 2 (3.5%) (n/g dry tissue)
N.A. 9 (15.8%) 0–999 0
Asbestos exposure (Asbestos bodies counts) 1000–9999 0
(n/g dry tissue) 10.000–99.999 30 (62.5%
0–999 13 (22.8%) 100.000–1.000.000 18 (37.5%)
1000–9999 20 (35.1%) N.A. 0
10.000–99.999 11 (19.3%) Ialine plaques, n(%)
100.000–1.000.000 5 (8.8%) Absent 0
NA 8 (14.0%) Grade 1 0
Ialine plaques, n (%) Grade 2 27 (56.25%)
Absent 2 (3.5%) Grade 3 21 (43.75%)
Grade 1 6 (10.5%) Occupation Type, n(%)
Grade 2 31 (54.4%) Shipyard 27 (56.25%)
Grade 3 12 (21.1%) Builder 2 (4.2%)
N.A. 6 (10.5%) Other 3 (6.25%)
Occupation Type, n(%) NA 16 (33.3%)
Maritime 4 (7.0%) Other 4 (7.69%)
Builder 3 (5.3%) N.A. 3 (5.77%)
Shipyard 44 (77.2%)
Other 1 (1.8%)
N.A. 5 (8.7%)
Scientific, Wilmington, DE) and agarose gel elec-
trophoresis; final concentration for genotyping was
50 ng/ml. Due to very aggressive fixation with 10%
without somatic alterations due to tumorigenic formalin, the quality of the DNA extracted was
transformation (Bisel, Wroblewski, and Ladue extremely poor with important degradation; this
1953). Mean age ± S.D. of the material at the time just allowed genetic analysis to be performed
of DNA extraction was 14 ± 11.69 years, ranging using Taqman probe technology, more suitable,
from 4 to 35 years. Fixation was made in 10% for- working with targets of limited size (i.e. 50 bp),
malin for all the samples; from the same paraffin for highly damaged DNA extracted from archive
block, 40–50 slices were cut with a 5–7 µm thickness samples (Supplementary Figure 1 illustrates an
and processed for DNA extraction. example of eight different samples; each of them
shows an extensive DNA degradation as indicated
by presence of intense staining at low molecular
DNA extraction from formalin-fixed weight).
paraffin-embedded (FFPE) samples
Genomic DNA was extracted using QIAamp DNA
HEPH, TF, FTH1, NLRP1, and NLRP3
FFPE Tissue Kit (Qiagen, Hilden Germany),
polymorphism analysis in AEM, AELC, and AE
according to manufacturer instructions, and eluted
population
in 30 µL final volume of Tris-EDTA buffer.
Extracted DNA was qualitatively and quantitatively Genetic variants previously analyzed in a population
evaluated using NanoDrop ND-1000 (Thermo from the same area were selected. These variants were
JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH, PART A 5

located in three Fe-metabolism-related genes, namely FractionalO:R:numerator



hephaestin (HEPH), transferrin (TF), and ferritin ðFractionalO:R:numeratorÞ þ ðFractionalO:R:denominatorÞ
heavy polypeptide 1 (FTH1) as in Crovella et al. with P as probability.
(2016), and in two inflammation-related genes,
namely nucleotide-binding domain, leucine-rich-
containing family, pyrin domain-containing-protein Results
3 (NLRP3), and NLRP1 as in Borelli et al. (2015). Study and population characteristics
Genotyping was performed on an ABI7900HT
Fast Real-time PCR (polymerase chain reaction) The study was performed on autopsy samples
instrument, using fluorescent TaqMan SNPs geno- derived from individuals exposed to asbestos
typing assay and TaqMan Genotyping Master Mix who developed either pleural mesothelioma
(Applied Biosystems-Life Technologies, Carlsbad, (AEM, n = 52) or lung cancer (AELC, n = 57)
CA) following manufacturer’s instructions. Samples using, as a reference, another set of autopsy sam-
were run in duplicate. For HEPH and TF poly- ples from asbestos-exposed individuals which
morphisms, rs3747359 and rs2715631, pre- died of asbestos-unrelated diseases (AE, n = 48).
designed SNP genotyping assay were used AEM and AELC populations were in Hardy–
(C__27476246_20 and C____148065_20, respec- Weinberg equilibrium (HWE) for all the genetic
tively); for FTH1 rs76059597 a custom TaqMan variants analyzed. However, the AE population
SNP assay (Life Technologies, Foster City, CA) was was not in HWE for FTH1 SNP, while for all
employed. For NLRP1 polymorphism, rs12150220 the other variants considered it was in HWE.
and rs6502867 pre-designed SNP genotyping assay
were used (C___1600653_10 and C__29222211_20). Asbestos-exposed individuals who developed
For NLRP3 polymorphism, rs35829419 and mesothelioma (AEM)
rs10754558, pre-designed SNP genotyping assays The AEM population included 6 women and 46
were used (C__25648615_10 and men, with a mean age of 74.2 ± 8.8 years at the
C__26052028_10). Data analysis was performed time of death, all exhibiting evidence of pleural pla-
both manually and automatically using SDS software ques, the majority of samples presenting grade 2 or
version 2.1 (Life Technologies, Foster City, CA). above (59.6%). Further, AB lung burden was deter-
mined for all cases, ranging from 108 to 950,000 AB/
g of dry tissue, median 6400 AB/g, which exceeds the
Statistical analysis threshold for occupational exposure (1000 AB/g)
Test for Hardy–Weinberg equilibrium (HWE) (Casali et al. 2015). Occupational data were available
either on autosomal or sex chromosomes was per- for 49 out of 52 cases (94.3%) and validated asbestos
formed with the HardyWeinberg R package exposure due to working conditions: 34 cases were
(Graffelman 2015). Allelic and genotypic differ- employed in the shipyard while additional 11 cases
ences between the groups were determined using in shipyard-related professions. Domestic (take-
the Fisher exact test whereas odds ratio (OR) and home) exposure was also observed in four cases,
95% confidence interval were estimated using being wives of shipyard workers.
Woolf’s method. Calculations were conducted for
different genetic models as indicated in Lewis and Asbestos-exposed individuals who developed lung
Knight (2012) Statistical calculation was per- cancer (AELC)
formed using R-studio (Anonymous 2015) and Only men belonged to the AELC population with
dedicated packages. Power analysis was performed a mean age of 73.9 ± 8.7 years at the time of death;
using GPower (G*Power, version 3.1.9.4, pleural plaques were present in 51 out of 57 cases
Universitat Kiel, Germany) (Faul et al. 2009). (89.5%) with the majority of samples showing grade
Probability calculation was made using guidelines 2 or above (75.5%). In addition, in this population, AB
from Andrade (2015) and Norton, Dowd, and lung burden was determined in the majority of sam-
Maciejewski (2018) and performed with the fol- ples (48 out of 57) demonstrating exposure exceeding
lowing formula the occupational threshold, with a median of 3600
6 F. CELSI ET AL.

AB/gr dry tissue (min = 60, max = 275,000), a result reference, subjects exposed to asbestos who did
confirmed by occupational data, available in 52 out of not develop asbestos-related diseases (AE). The
57 cases, showing that the majority of cases were aim was to determine if a common pathogenic
employed in shipyard (44 cases) and shipyard- mechanism existed, shared by AEM and AELC.
related professions (7 cases), while for only one case
employment was not directly linked to occupational
Iron(Fe)-related genes
asbestos exposure, being the patient a carpenter, not
directly working on the shipyard. HEPH gene
Smoking habit was available for 48 cases out of The first genetic variant examined in this study was
57 (84.2%): 46 cases were smokers (S) and only 2 rs3747359 in the HEPH gene. It is a coding SNP
cases were nonsmokers (N). Due to the small (cSNP) with a minimum allele frequency
number of nonsmoker samples, it was not possible (MAF) < 0.01, as reported by 1000 genomes data
to subdivide the AELC population based on smok- (http://www.internationalgenome.org/); HEPH gene
ing habits. Similarly, due to the small number of is located on X chromosome. The protein encoded
samples, it was not possible to subdivide both by this gene, hephaestin, is a multi-copper oxidase,
AEM and AELC population based on pleural/ involved in Fe transport across epithelial cells into
lung cancer histological subtype, an interesting the circulatory system. As HEPH is localized on the
aspect that requires further investigation. X chromosome and not in the pseudoautosomal
region, allele frequencies were compared by count-
Asbestos-exposed individuals (AE) ing the chromosomes, according to Clayton (2009),
The reference AE population is composed of 45 where males contribute for one observation and
men and 3 women, mean age 80.1 ± 0.6 years at females for two. For the analysis to be considered
the time of death; cause of death is not asbestos- statistically solid, HWE must be valid, as it is in our
related. These subjects were also selected based samples (p = .14 for AE and p = .76 for AEM with χ2
upon the presence of AB and of pleural plaques test). Further, to avoid sex bias effects, allele frequen-
of class 2 or above. Quantitation of AB lung bur- cies between male and female need to be equal, as
den in all cases indicated that asbestos exposure was the case for our samples (p = .06 for AE and p = 1
exceeds the threshold for occupational exposure for AEM with Fisher’s exact test comparing the
(AB/g dry lung tissue from 16,000 to 994,000). frequencies). Taking into account these considera-
Occupational data were available for only 32 out tions, the allelic frequencies were compared: the
of 48 individuals with 27 patients being employed C allele was statistically more frequent in AE popula-
in shipyard and 5 in shipyard-related professions. tion than in AEM thus possibly being associated with
Exposure and longevity data indicate that AE, even protection against mesothelioma (OR = 0.06 95% CI
though exposed to asbestos, did not develop = 0.003–0.3; p = 8.5x10−5, Fisher Exact test, power β
mesothelioma or lung cancer, and thus were = 0.99), increasing the probability by 5.6% of not to
enrolled as a control for genetic testing on AEM develop this disease. In addition, assessing the
and AELC, as previously described in Crovella genetic frequencies, a statistically significant differ-
et al. (2016). ence in C hemizygotes in AE population was
observed, where this genotype was detected more
frequently in this population compared to AEM.
Genetic analysis
The C/C genotype was also more frequent in the
Initially, an attempt was made to replicate the control population (Figure 1) (OR = 0.01 95%
findings obtained in Crovella et al. (2016) for Fe- CI = 0.00–0.05 p = 2.68x10−18 Fisher’s Exact test,
related genes and in Borelli et al. (2015) for power β = 0.99). Genetic model of inheritance was
inflammasome-related genes in a new population also analyzed, considering males as homozygote as in
of asbestos-exposed individuals who developed Clayton (2009), and statistical analysis indicates
mesothelioma (AEM). Subsequently, a population a dominant model of inheritance (Supplementary
of exposed individuals was assessed who developed Table 1) (G/G vs G/C + C/C OR = 0.04 95%
lung cancer (AELC) and compared using, as CI = 0.0004–0.2 p = 2.6 X10−7, power β = 0.99).
JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH, PART A 7

TF gene
One variant in the TF gene, which encodes transferrin,
a protein involved in Fe transport was also assessed.
The SNP rs2715631, situated in an intronic region,
was reported to be involved in the protection from
mesothelioma, as previously shown in Crovella et al.
(2016). Allele G was significantly more frequent in AE
population in comparison with AEM or AELC
(OR = 0.22 95% CI = 0.12–0.47 p = 3.5 X 10−6,
Fisher’s Exact test for AEM vs AE, power β = 0.93
Figure 1. rs3747359 – HEPH gene – genetic frequencies in AE Supplementary Table 3) (OR = 0.15 95%
(control) and AEM (mesothelioma-affected) populations. CI = 0.07–0.31 p = 3.9 X 10−9, Fisher Exact test for
AELC vs AE, power β = 0.99 Supplementary Table 4).
Therefore, the presence of the G allele increases the
probability of not developing pleural mesothelioma by
18.1% and 13.01% considering lung cancer. Analyzing
genotype distribution in AEM vs AE (Figure 3)
a major frequency of G/G individuals in AE control
population was noted, suggesting a protective role of
this genotype against mesothelioma. Statistical analy-
sis showed a dominant model of inheritance for pro-
tective alleles (T/T vs T/G + G/G OR = 0.22; 95%
CI = 0.09–0.52 p = 3.3 x10−4; Fisher’s Exact test,
power β = 0.99) (Supplementary Table 3).
Figure 2. rs3747359 – HEPH gene – genetic frequencies in AE Further, genotype frequencies in AELC vs AE
(control) and AELC (lung carcinoma-affected) populations.
populations demonstrated a major frequency of G/
G individuals in the control population (Figure 4).
We decided to further expand our findings examin- Statistical analysis indicated a co-dominant model of
ing the AELC population in comparison to AE (con- inheritance for the protective alleles (T/T vs T/G
trol). As the AELC population composed only of OR = 0.21 95% CI = 0.08–0.56 p = 1.7 X 10−3
males, it was not possible to verify HWE and equal Fisher’s Exact test, power β = 0.99, and T/T vs G/G
allelic frequency between genders. Indeed, since the OR = 0.06 95% CI = 0.01–0.22 p = 2.9 X 10−6 Fisher’s
AE population fulfilled those criteria, assessment was Exact test, power β = 0.99) (Supplementary Table 4).
undertaken to determine allelic distribution in these
samples, finding a significant difference of C allele FTH1 gene
presence in AE population, associating with protec- The third gene examined in this study was FTH1,
tion against LC (Figure 2) (OR = 0.03 95% selected since one SNP, rs76059597, located in an
CI 0.005 − 0.12, p = 6.1x10−12, Fisher Exact test,
power β = 0.99). The presence of the C allele elevated
2.9% the probability of not developing lung cancer.
Observing the genetic distribution between the two
population it is also possible to note the higher
frequency of C allele and C/C genotypes in AE with
respect to AELC (Figure 2) (OR: = 0.03 95% CI
0.01–0.13, p = 2.69x10−18, Fisher’s Exact test, power
β = 0.99) (Figure 2). Analysis of inheritance models
indicated in this case is a dominant model (G/G vs
G/C + C/C OR = 0.04 95% CI = 0.01–0.16; p = 6.3 Figure 3. rs2715631 – TF gene – genetic frequencies in AE
X10−8, power β = 0.99) (Supplementary table 2). (control) and AEM (mesothelioma-affected) populations.
8 F. CELSI ET AL.

Figure 4. rs2715631 – TF gene – genetic frequencies in AE Figure 6. rs76059597 – FTH1 gene – genetic frequencies in AE
(control) and AELC (lung carcinoma-affected) populations. (control) and AELC (lung carcinoma-affected) populations.

intronic region, was described as involved in protec- 0.15, 95% CI = 0.06–0.35, p-value from Fisher’s Exact
tion against mesothelioma in asbestos-exposed indi- test = 1.4 X 10−5, power β = 0.99) (Supplementary
viduals (Crovella et al. 2016). FTH1 encodes ferritin Table 5).
heavy subunit, a protein involved in Fe-storage and Similarly, for the other genes, genotypes distribu-
one of the main components of AB coating (Borelli tion in AELC vs AE populations demonstrated
et al. 2007). In AEM population allele C was signifi- a higher prevalence of C/C individuals in AE control
cantly less frequent in comparison with AE, confirm- population, again suggesting a protective role of this
ing previous results (Crovella et al. 2016)(OR = 0.1 genotype (Figure 6). Statistical analysis indicated
95% CI = 0.05–0.22 p = 2.5X 10−11, Fisher’s Exact test, a dominant model of transmission (T/T vs C/T + C/
power β = 0.99; Supplementary Table 5). Further, in C, OR = 0.22, 95% CI = 0.09–0.47, p-value for Fisher’s
AELC population, the C allele was less present in Exact test = 3.1 x10−4, power β = 0.99) (Supplementary
affected individuals (OR = 0.13 95% CI = 0.07–0.25 Table 6)
p = 2.3 X 10−10, Fisher’s Exact test, power β = 0.99;
Supplementary Table 6). These results indicated that
with the presence of this allele, the probability of Inflammation-related genes
developing either mesothelioma or lung cancer was
Following replication and expansion of previous
reduced by 9.1% and 11.5%, respectively.
findings regarding Fe-signature genes, it was decided
Genotypes distribution in AEM vs AE population
to test if genetic variants in inflammasome genes
also exhibited the major prevalence of the C/C geno-
might influence the risk of developing mesothelioma
type in AE (control) with respect to the AEM popula-
or lung cancer. The proteins coded by NLRP1 and
tion, confirming a potential protective role (Figure 5).
NLRP3 genes are involved in innate immunity and
Indeed, statistical analysis of genotype distribution
inflammation. In response to damage-associated sig-
suggested a dominant model of inheritance further
nals and pathogens, these proteins catalyze the
indicating a protective role (T/T vs T/C + C/C OR =
assembly of inflammasome complex, which activates
and cleaves caspase-1, leading to secretion of IL-1β,
a key mediator of inflammation (Hayward et al.
2018). Previously Borelli et al. (2015) examined the
role of different SNPs in NLRP1 and NLRP3 genes
and noted no significant difference in allelic distri-
bution between the AEM and AE.

NLRP1 gene
When analyzing rs12150220 alleles distribution in
the AEM population, no marked differences were
Figure 5. rs76059597 – FTH1 gene – genetic frequencies in AE found in comparison to AE subjects, thus indicating
(control) and AEM (mesothelioma-affected) populations. no apparent involvement of this NLRP1
JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH, PART A 9

inflammasome SNP in mesothelioma development NLRP3 protein was proposed as the key mediator
(Table 4, top). When considering the AELC group in in this process (Thompson et al. 2017). Previously
the entire population, no marked differences were Borelli et al. (2015) found that two NLRP3 gene
observed in the distribution of NLRP1 SNP SNPs (rs10754558 and rs35829419) did not associ-
rs12150220 A and T allele when compared with AE ate with mesothelioma predisposition. It was thus
reference (Table 4, top). of interest to replicate our previous findings by
To complete our analysis, allelic distribution of analyzing both NLRP3 SNPs which were formerly
another NLRP1 SNP, rs6502867, was then assessed assessed. As indicated in Table 5, both NLRP3 SNP
as described by Borelli et al. (2015), pertaining to alleles exhibited no significant differences consid-
a different haploblock than rs12150220. Even on ering either AEM vs AE and AELC vs AE groups.
this occasion, no statistical differences in allele
distribution were detected both between AEM
and AE and between AELC and AE populations, Discussion
further confirming previous Borelli et al. (2015)
findings for AEM group and expanded our obser- The presence of genetic risk factors affecting
vation to AELC patients (Table 4, bottom) mesothelioma development is demonstrated by
epidemiological analysis, where it was evident
that only a minority of asbestos-exposed subjects
NLRP3 gene develop such pleural neoplasm (5–17% of heavily
The involvement of inflammasome in susceptibil- exposed individuals) (Neri et al. 2008). Further,
ity to develop malignant pleural mesothelioma subjects only mildly exposed to asbestos might
(MPM), considering its capacity to interact with develop the disease whereas others, heavily
asbestos fibers was previously reported (Dostert exposed, apparently fail to become ill (Betti et al.
et al. 2008; Hillegass et al. 2013; Mossman et al. 2018; Carbone and Yang 2012). However, as MPM
2013). Activation of inflammation appears to be is a rare disease with a long latency period, the
crucial for the transition from mesothelial cell to design of genetic susceptibility studies might be
fibroblast cell, induced by asbestos fibers, and affected by these two factors. Moreover, asbestos

Table 4. Distribution of genotype frequencies for NLRP1 polymorphism rs12150220 and rs6502867 in asbestos-
exposed individuals who developed mesothelioma (AEM) or lung cancer (AELC) vs asbestos-exposed indivi-
duals (AE) OR (CI 95%): Odds ratio with values covering 95% confidence interval. P value: p value from fisher
exact tests, asterisk indicates statistical significance.
NLRP1
AEM AE OR (CI 95%) P-value
rs12150220 n = 52 (Freq.) n = 48 (Freq.)
A/A 18 (0.35) 14 (0.29) Ref.
A/T 23 (0.44) 21 (0.44) 0.85 (0.33–2.15) 0.82
T/T 11 (0.21) 13 (0.27) 0.66 (0.22–1.94) 0.59
AELC AE OR (CI 95%) P-value
rs12150220 n = 57 (Freq.) n = 48 (Freq.)
A/A 20 (0.35) 14 (0.29) Ref.
A/T 24 (0.43) 21 (0.44) 0.8 (0.32–1.98) 0.65
T/T 13 (0.22) 13 (0.27) 0.7(0.24–1.99) 0.6
AEM AE OR (CI 95%) P-value
rs6502867 n = 52 (Freq.) n = 48 (Freq.)
T/T 26 (0.5) 27 (0.57) Ref.
T/C 17 (0.33) 16 (0.33) 1.1 (0.45–2.66) 1
C/C 9 (0.17) 5 (0.10) 1.83 (0.54–6.84) 0.37
AELC AE OR (CI 95%) P-value
rs6502867 n = 57 (Freq.) n = 48 (Freq.)
T/T 36 (0.63) 27 (0.57) Ref.
T/C 15 (0.26) 16 (0.33) 0.7 (0.29–1.68) 0.51
C/C 6 (0.11) 5 (0.10) 0.89 (0.24–3.52) 1.0
10 F. CELSI ET AL.

Table 5. Distribution of genotype frequencies for NLRP3 polymorphism rs10754558 and rs35829419 in
asbestos-exposed individuals who developed mesothelioma (AEM) or lung cancer (AELC) vs. asbestos-
exposed individuals (AE) OR (CI 95%): Odds ratio with values covering 95% confidence interval. P-value:
p-value from fisher exact tests, asterisk indicates statistical significance.
NLRP3
AEM AE OR (CI 95%) P-value
rs10754558 n = 52 (Freq.) n = 48 (Freq.)
G/G 14 (0.27) 16 (0.34) Ref.
G/C 27 (0.52) 21 (0.44) 1.45 (0.58–3.72) 0.48
C/C 11 (0.21) 11 (0.22) 1.13 (0.37–3.51) 1
AELC AE OR (CI 95%) P-value
rs10754558 n = 57 (Freq.) n = 48 (Freq.)
G/G 16 (0.28) 16 (0.34) Ref.
G/C 28 (0.49) 21 (0.44) 1.32 (0.53–3.29) 0.64
C/C 13 (0.23) 11 (0.22) 1.17 (0.4–3.48) 0.79
AEM AE OR (CI 95%) P-value
rs35829419 n = 52 (Freq.) n = 48 (Freq.)
C/C 46 (0.88) 46 (0.96)
C/A 6 (0.12) 2 (0.04) 2.83 (0.59–22.32) 0.27
A/A 0 (0) 0 (0) NA 1
AELC AE OR (CI 95%) P-value
rs35829419 n = 57 (Freq.) n = 48 (Freq.)
C/C 52 (0.91) 46 (0.96) Ref.
C/A 5 (0.09) 2 (0.04) 2.1 (0.41–17.01) 0.44
A/A 0 (0) 0 (0) NA 1

exposure is not routinely quantitatively evaluated. imbb.forth.gr/SSCprofiler.html) to check for miRNA


Postmortem FFPE tissue samples represent an genes, without reaching conclusive results. Since this
important tool for uncovering genetic susceptibil- finding, in spite of being replicated in a group of
ity to mesothelioma since these enable the selec- patients and controls coming from the same geo-
tion of individuals with similar exposure features graphic area (Monfalcone, Italy), has not been pub-
and are well characterized as far as clinicopatho- lished even in large GWAS studies, functional
logical characteristics and occupational history are studies are strongly needed to confirm the impact
concerned. of these genes and variation on susceptibility to
Previously Crovella et al. (2016) identified in mesothelioma.
a population-based autopsy study three Fe metabo- Data demonstrated that NLRP1 gene is not
lism-associated genes, significantly associated with involved in mesothelioma development, as no associa-
protection against MPM while no association was tion was found between the pathology and rs12150220
found for NLRP1 and NLRP3 polymorphisms SNP, in contrast to previous results from Girardelli
(Borelli et al. 2015). Our previous published results et al. (2012). Allelic frequencies obtained in the present
were confirmed (Crovella et al. 2016) by employing study (A = 0.56 and T = 0.44) are in accordance with
a different set of samples. The protective role of Borelli et al. (2015) (A = 0.50 and T = 0.50) and
HEPH coding SNP (rs3747359) was confirmed as Crovella et al. (2018) (A = 0.54 and T = 0.46), together
well as of the two non-coding SNPs, either in FTH with genotype frequencies, further strengthening our
or in TF (rs2715631 and rs76059597, respectively). conclusions. Allelic frequencies obtained in the pre-
Forgoing in silico analysis (Crovella et al. 2016) sug- sent and previous studies are similar to those reported
gested a damaging effect of C substitution in HEPH by 1000 Genomes project (http://www.international
gene which might impair hephaestin protein func- genome.org/home) for the European (A = 0.56 and
tion; however, it was not possible to determine the T = 0.44) and Tuscan (A = 0.55 and T = 0.45) popula-
putative functional impact related to the non-coding tions, indicating significant concordance of our ana-
SNPs. Indeed, other databases (such as SROOGLE; lysis with the general Italian population. Evidence
http://sroogle.tau.ac.il/) were also examined for indicated that polymorphisms in another inflamma-
change in splice sites or SSC profiler (http://mirna. tion-related gene, NLRP3, are not linked to MPM
JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH, PART A 11

development, corroborating previous results from showed the presence of BAP1 (BRCA associated
Borelli et al. (2015). The availability of novel samples protein 1) mutations in a minority of patients
was not limited to mesothelioma patients but also (Ohar et al. 2016), a result further confirmed by
included individuals who died of lung cancer, another two whole-genome investigations accomplished in
asbestos-related disease for which, to date, the impact different geographic areas (Betti et al. 2017; Bueno
of individual genetic variations is still poorly under- et al. 2016). Mutations in the tumor suppressor
stood and also less studied. gene BAP1 (truncation, deletions, somatic variants
Inflammation and disruption of Fe homeostasis including large deletions, and/or chromosomal
are hallmarks of asbestos exposure affecting speci- loss) associated with different types of cancer,
fically lung tissue and inflammation is a key player including MPM, reviewed in Betti et al. (2019),
in lung cancer development which is mediated by and LC (Carbone et al. 2013), supporting the con-
activation of both inflammasome platforms cept that a common mutation might trigger
mediated by NLRP1 and NLRP3, expressed and diverse malignancies. BAP1 plays an important
activated in various lung cancer cell lines although role in chromatin modulation, transcriptional reg-
at different levels (Kong et al. 2015). However, ulation, cell proliferation, DNA repair, cell death,
data demonstrated that, as for MPM, SNPs at and glucidic metabolism. Interestingly, Zhang
NLRP1 and NLRP3 genes appeared not to be et al. (2018) reported that cells with reduced
involved in LC development. Interestingly, for all BAP1 activity also display impaired ferroptosis,
tested Fe related-SNPs, similar results were providing a possible link between Fe and BAP1
obtained as with mesothelioma samples, suggest- pathways in the development of asbestos-related
ing that a common protection mechanism might malignancies. Many other genes were identified as
operate. Iron metabolism is crucial for cells and involved in predisposition to MPM, which are
the whole organism survival, and precise home- predominantly involved in DNA repair (Betti
ostasis of this element is required. et al. 2019). These findings suggest that the con-
Evidence exists that cigarette smoke, the major tribution of the BAP1 failure (and also that of the
risk factor for lung cancer (Le Calvez et al. 2005), other genes) may follow the damage initiated by
produces Fe dysregulation (Zhang, Butler, and other pathways triggered by asbestos, such as those
Cloonan 2019) as well as asbestos, a lower risk factor Fe-mediated. It is likely that it is the synergic
(Kamp 2009). Asbestos exposure induced lung can- (rare) combination of these pathways to determine
cer independently or synergistically, with smoking the predisposition. External pollutants, such as
(International Agency for Research on Cancer and asbestos and cigarette smoke, increase Fe loads in
Weltgesundheits Organisation 2012) but the inter- the lungs (Ghio et al. 2008; Pascolo et al. 2016),
action between asbestos and smoking was found to induce oxidative stress and inflammation, tipping
be approximately multiplicative (Markowitz et al. the point toward cancer development. In this con-
2013). Unfortunately, due to the very small number text, alterations of genes involved in Fe-
of nonsmoker samples in the AELC population (2 of metabolism might either elevate or decrease the
57), it was not possible to subdivide it based upon toxicity of this metal thus possibly either inducing
smoking habits. Anyway, since our AELC study or protecting from the neoplastic transformation.
population was almost composed by smokers, it Our study confirms the previous results obtained
was postulated that asbestos and smoking exert in individuals exposed to asbestos which developed
a multiplier effect in inducing Fe dysregulation. mesothelioma and extends these findings to indivi-
Iron may represent the link between exposure to duals who developed lung cancer. As indicated
pollutants such as cigarette smoke, asbestos and above, these results led us to hypothesize the pre-
others, and related lung diseases. This interesting sence of a common mechanism in developing an
topic will need further investigation in a novel popu- asbestos-related disease that needs further investiga-
lation of AELC with participants equally distributed tion. The Fe-metabolism pathway might be poten-
between never, former, and current smokers. tially involved in pleural mesothelioma and lung
Recently, a screening of a cohort of mesothe- cancer development, with hephaestin playing a key
lioma patients with a family history of cancer role; expression of this protein was described as
12 F. CELSI ET AL.

a prognostic marker in renal cancer and glioma, comprehensive NGS study. A functional validation of
attributed to low expression correlating with the lung cell model is also needed to reaffirm and
increased survival (Uhlen et al. 2017)(The Human confirm preliminary genetic findings. Further
protein Atlas: www.proteinatlas.org). A possible sce- research into a specific “iron genetic signature” is
nario is conceivable in which the hypofunctional needed to broaden our knowledge of the genetic land-
hephaestin is correlated with protection against can- scape causative of pleural mesothelioma and lung
cer development; thus, its ferroxidase activity may be cancer. In addition, this research may be extended
correlated with neoplastic transformation, i.e. via also to other types of “non asbestos” (non-regulated)
ROS increase, modulated by the other proteins elongated mineral particles found in the natural envir-
linked to Fe metabolism; this scenario still needs onment, which, because of their ion-exchange proper-
a robust functional confirmation. The availability of ties, might acquire Fe after inhalation and whose
well-characterized populations in regard to asbestos exposure was also associated with mesothelioma
exposure that has been objectively quantitated dur- (e.g. erionite) (Ruda and Dutta 2005; Carbone et al.
ing necroscopic examination, and availability of 2011; Baumann, Ambrosi, and Carbone 2013).
occupational data, enabled us to precisely select indi-
viduals with comparable levels of asbestos exposure.
Numerous genetic investigations such as Wei et al. Acknowledgments
(2012) lack an exact quantitation of asbestos expo- We thank all the staff of the Monfalcone’s Hospital
sure, referring only to self-reported exposure or Pathological Anatomy Unit for their valuable technical assis-
occupational data. Our study, in spite of the power tance. We thank also the Council for the management of
analysis confirming the distribution in AE popula- Fondo del Lascito Carrara for the technical support.
tion of possible “protective” alleles in Fe-metabolism
genes, with respect to both AEM and AELC popula- Declaration of interest
tions, suffers limitations, due to the analysis of
All authors declare to have no conflict of interest.
a limited number of genetic variants on a small-size
sample instead of NGS approach as in most of the
recent published studies. Nevertheless, DNA quality Funding
of many samples was too poor, due to significant
degradation after aggressive fixation methods, to The authors acknowledge a grant from Regione Autonoma
Friuli-Venezia Giulia, Assessorato alla Salute e Protezione
perform Whole Exome Sequencing or targeted re-
Sociale, LR 22/2001 (decree 1124/SPS, 09/20/2016, N° 1299)
sequencing focusing on sets of genes already consid- and from Italian League for the Fight Against Cancer (LILT),
ered in previous studies, as experienced when trying Gorizia section (Bando di Ricerca sanitaria 2017-programma
to perform NGS experiments. Therefore, in our 5 per mille anno 2015);Regione Autonoma Friuli-Venezia
experimental design, it was decided to use the robust Giulia, Assessorato alla Salute e Protezione Sociale, LR 22/
Taqman technology, which is capable of working on 2001 [decree 1124/SPS, 09/20/2016, N° 1299];Italian League
for the Fight Against Cancer Lega Italiana per la Lotta contro
extremely fragmented DNA thus allowing us to
i Tumori (LILT) [Bando di Ricerca sanitaria 2017-
replicate previous findings obtained on another programma 5 per mille];
group of mesothelioma patients from the same geo-
graphic region (Monfalcone, North-East Italy). Of
course, this approach does not permit any compar- References
ison with the recent GWAS literature results, thus Andrade, C. 2015. Understanding relative risk, odds ratio,
limiting the strength of our observations that need to and related terms: As simple as it can get. J. Clin. Psychiat.
be considered cautiously. 76:e857–861. doi:10.4088/JCP.15f10150.
The characterization of our samples in terms of Andujar, P., A. Lacourt, P. Brochard, J.-C. Pairon, M.-C. Jaurand,
environmental pressure is the major strength of our and D. Jean. 2016. Five years update on relationships between
experimental sample set to aim at increasing the malignant pleural mesothelioma and exposure to asbestos and
other elongated mineral particles. J. Toxicol Environ. Health B
number of samples by selecting better conserved (in 19:151–72. doi:10.1080/10937404.2016.1193361.
terms of DNA preservation) paraffin blocks, control- Anonymous. 2015. RStudio: Integrated development for
ling the DNA quality and finally proceed with a more r. rstudio, inc. http://www.rstudio.com/.
JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH, PART A 13

Ather, J. L., R. A. Martin, K. Ckless, and M. E. Poynter. 2014. Cadby, G., S. Mukherjee, A. W. B. Musk, A. Reid, M. Garlepp,
Inflammasome activity in non-microbial lung I. Dick, C. Robinson, J. Hui, G. Fiorito, S. Guarrera, et al. 2013.
inflammation. J. Environ. Immunol. Toxicol. 1:108–17. A genome-wide association study for malignant mesothelioma
Aust, A. E., P. M. Cook, and R. F. Dodson. 2011. risk. Lung Cancer 82:1–8. doi:10.1016/j.lungcan.2013.04.018.
Morphological and chemical mechanisms of elongated Carbone, M., P. S. Adusumilli, H. R. Alexander, P. Baas,
mineral particle toxicities. J. Toxicol Environ. Health B F. Bardelli, A. Bononi, R. Bueno, E. Felley-Bosco,
14:40–75. doi:10.1080/10937404.2011.556046. F. Galateau-Salle, D. Jablons, et al. 2019. Mesothelioma:
Aust, E. A., L. G. Lund, C. C. Chao, S. H. Park, and R. Fang. 2000. Scientific clues for prevention, diagnosis, and therapy. CA
Role of iron in the cellular effects of asbestos. Inhal Toxicol 12 Cancer J. Clin. 69:402–29. doi:10.3322/caac.21572.
(Suppl 3):75–80. doi:10.1080/08958378.2000.11463232. Carbone, M., Y. I. Baris, P. Bertino, B. Brass, S. Comertpay,
Baumann, F., J. P. Ambrosi, and M. Carbone. 2013. Asbestos A. U. Dogan, G. Gaudino, S. Jube, S. Kanodia, C. R. Partridge,
is not just asbestos: An unrecognised health hazard. Lancet et al. 2011. Erionite exposure in North Dakota and Turkish
Oncol. 14:576–78. doi:10.1016/S1470-2045(13)70257-2. villages with mesothelioma. Proc. Natl. Acad. Sci.
Betti, M., A. Aspesi, D. Ferrante, M. Sculco, L. Righi, 108:13618–23. doi:10.1073/pnas.1105887108.
D. Mirabelli, F. Napoli, M. Rondón-Lagos, E. Casalone, Carbone, M., L. K. Ferris, F. Baumann, A. Napolitano, C. A. Lum,
F. Vignolo Lutati, et al. 2018. Sensitivity to asbestos is E. G. Flores, G. Gaudino, A. Powers, P. Bryant-Greenwood,
increased in patients with mesothelioma and pathogenic T. Krausz, et al. 2012. BAP1 cancer syndrome: Malignant
germline variants in bap1 or other DNA repair genes. Genes mesothelioma, uveal and cutaneous melanoma, and MBAITs.
Chromosomes Cancer 57:573–83. doi:10.1002/gcc.22670. J. Transl. Med. 10:179. doi:10.1186/1479-5876-10-179.
Betti, M., A. Aspesi, M. Sculco, G. Matullo, C. Magnani, and Carbone, M., and H. Yang. 2012. Molecular pathways:
I. Dianzani. 2019. Genetic predisposition for malignant Targeting mechanisms of asbestos and erionite carcino-
mesothelioma: A concise review. Mutat. Res/Rev Mutat. genesis in mesothelioma. Clin. Cancer Res. 18:598–604.
Res. 781:1–10. doi:10.1016/j.mrrev.2019.03.001. doi:10.1158/1078-0432.CCR-11-2259.
Betti, M., E. Casalone, D. Ferrante, A. Aspesi, G. Morleo, A. Biasi, Carbone, M., H. Yang, H. I. Pass, T. Krausz, J. R. Testa, and
M. Sculco, G. Mancuso, S. Guarrera, L. Righi, et al. 2017. G. Gaudino. 2013. BAP1 and cancer. Nat. Rev. Cancer
Germline mutations in DNA repair genes predispose 13:153–59. doi:10.1038/nrc3459.
asbestos-exposed patients to malignant pleural mesothelioma. Casali, M., M. Carugno, A. Cattaneo, D. Consonni, C. Mensi,
Cancer Lett. 405:38–45. doi:10.1016/j.canlet.2017.06.028. U. Genovese, D. M. Cavallo, A. Somigliana, and
Bianchi, C., A. Brollo, L. Ramani, and R. Berté. 1991. Exposure A. C. Pesatori. 2015. Asbestos lung burden in necroscopic
to asbestos in Monfalcone, Italy. A necropsy-based study. samples from the general population of Milan, Italy. Ann.
IARC Sci. Publ. 112:127–40. Occup. Hyg. 59:909–21. doi:10.1093/annhyg/mev028.
Bianchi, C., A. Brollo, L. Ramani, and C. Zuch. 1993. Chew, S. H., and S. Toyokuni. 2015. Malignant mesothelioma as
Asbestos-related mesothelioma in Monfalcone, Italy. Am. an oxidative stress-induced cancer: An update. Free Radic. Biol.
J. Ind. Med. 24:149–60. doi:10.1002/ajim.4700240203. Med. 86:166–78. doi:10.1016/j.freeradbiomed.2015.05.002.
Bianchi, C., and T. Bianchi. 2007. Malignant mesothelioma: Christiani, D. C. 2000. Smoking and the molecular epide-
Global incidence and relationship with asbestos. Ind miology of lung cancer. Clin. Chest Med. 21:87–93.
Health 45:379–87. doi:10.2486/indhealth.45.379. doi:10.1016/S0272-5231(05)70009-5.
Bisel, H. F., F. Wroblewski, and J. S. Ladue. 1953. Incidence and Clayton, D. G. 2009. Sex chromosomes and genetic associa-
clinical manifestations of cardiac metastases. J. Am. Med. tion studies. Genome Med 1:110. doi:10.1186/gm110.
Assoc. 153:712–15. doi:10.1001/jama.1953.02940250018005. Crovella, S., R. R. Moura, S. Cappellani, F. Celsi, E. Trevisan,
Borelli, V., C. Brochetta, M. Melato, C. Rizzardi, M. Polentarutti, M. Schneider, A. Brollo, E. M. Nicastro, F. Vita, L. Finotto,
C. Busatto, F. Vita, R. Abbate, R. Gotter, and G. Zabucchi. et al. 2018. A genetic variant of nlrp1 gene is associated
2007. A procedure for the isolation of asbestos bodies from with asbestos body burden in patients with malignant
lung tissue by exploiting their magnetic properties: A new pleural mesothelioma. J. Toxicol. Environ. Health A
approach to asbestos body study. J. Toxicol. Environ. Health 81:98–105. doi:10.1080/15287394.2017.1416911.
A 70:1232–40. doi:10.1080/15287390701380906. Crovella, S., A. M. Bianco, J. Vuch, L. Zupin, R. R. Moura,
Borelli, V., R. R. Moura, E. Trevisan, and S. Crovella. 2015. E. Trevisan, M. Schneider, A. Brollo, E. M. Nicastro,
NLRP1 and NLRP3 polymorphisms in mesothelioma patients A. Cosenzi, et al. 2016. Iron signature in asbestos-induced
and asbestos exposed individuals a population-based autopsy malignant pleural mesothelioma: A population-based
study from north east Italy. Infect. Agents Cancer 10:26. autopsy study. J. Toxicol. Environ. Health A 79:129–41.
doi:10.1186/s13027-015-0022-0. doi:10.1080/15287394.2015.1123452.
Bueno, R., E. W. Stawiski, L. D. Goldstein, S. Durinck, A. De Dianzani, I., L. Gibello, A. Biava, M. Giordano, M. Bertolotti,
Rienzo, Z. Modrusan, F. Gnad, T. T. Nguyen, B. S. Jaiswal, M. Betti, D. Ferrante, S. Guarrera, G. P. Betta, D. Mirabelli,
L. R. Chirieac, et al. 2016. Comprehensive genomic analy- et al. 2006. Polymorphisms in DNA repair genes as risk
sis of malignant pleural mesothelioma identifies recurrent factors for asbestos-related malignant mesothelioma in
mutations, gene fusions and splicing alterations. Nat. a general population study. Mutat. Res. 599:124–34.
Genet. 48:407–16. doi:10.1038/ng.3520. doi:10.1016/j.mrfmmm.2006.02.005.
14 F. CELSI ET AL.

Dostert, C., V. Pétrilli, R. Van Bruggen, C. Steele, C, arsenic, metals, fibres, and dusts: This publication represents
B. T. Mossman, and J. Tschopp. 2008. Innate immune activa- the views and expert opinions of an IARC working group on the
tion through Nalp3 inflammasome sensing of asbestos and evaluation of carcinogenic risks to humans, which met in Lyon,
silica. Science 320:674–77. doi:10.1126/science.1156995. 17–24 March 2009. Lyon: IARC.ISBN-13: 978-9283213208.
Faul, F., E. Erdfelder, A. Buchner, and A.-G. Lang. 2009. Jiang, L., S. Akatsuka, H. Nagai, S.-H. Chew, H. Ohara,
Statistical power analyses using g*power 3.1: Tests for Y. Okazaki, Y. Yamashita, Y. Yoshikawa, H. Yasui, K. Ikuta,
correlation and regression analyses. Behav. Res. Meth. et al. 2012. Iron overload signature in chrysotile-induced
41:1149–60. doi:10.3758/BRM.41.4.1149. malignant mesothelioma. J. Pathol. 228:366–77. doi:10.1002/
Gemignani, F., M. Neri, F. Bottari, R. Barale, P. A. Canessa, path.4075.
F. Canzian, M. Ceppi, I. Spitaleri, M. Cipollini, G. P. Ivaldi, Kamp, D. W. 2009. Asbestos-induced lung diseases: An update.
et al. 2009. Risk of malignant pleural mesothelioma and Transl Res 153:143–52. doi:10.1016/j.trsl.2009.01.004.
polymorphisms in genes involved in the genome stability Kamp, D. W., and S. A. Weitzman. 1999. The molecular basis
and xenobiotics metabolism. Mutat. Res. 671:76–83. of asbestos induced lung injury. Thorax 54:638–52.
doi:10.1016/j.mrfmmm.2009.09.003. doi:10.1146/annurev-pathol-020712-163942.
Ghio, A. J., E. D. Hilborn, J. G. Stonehuerner, L. A. Dailey, Kettunen, E., H. Hernandez-Vargas, M.-P. Cros, G. Durand,
J. D. Carter, J. H. Richards, K. M. Crissman, R. F. Foronjy, F. Le Calvez-Kelm, K. Stuopelyte, S. Jarmalaite,
D. L. Uyeminami, and K. E. Pinkerton. 2008. Particulate K. Salmenkivi, S. Anttila, H. Wolff, et al. 2017. Asbestos-
matter in cigarette smoke alters iron homeostasis to pro- associated genome-wide DNA methylation changes in lung
duce a biological effect. Am. J. Respir. Crit. Care Med. cancer. Int. J. Cancer 141 (10):2014–29. doi:10.1002/
178:1130–38. doi:10.1164/rccm.200802-334OC. ijc.30897.
Ghio, A. J., E. N. Pavlisko, and V. L. Roggli. 2015. Iron and Kong, H., Y. Wang, X. Zeng, Z. Wang, H. Wang, and W. Xie.
iron-related proteins in asbestosis. J. Environ. Pathol. Toxicol. 2015. Differential expression of inflammasomes in lung
Oncol. 34:277–85. doi:10.1615/JEnvironPatholToxicolOncol. cancer cell lines and tissues. Tumour Biol. 36:7501–13.
v34.i4. doi:10.1007/s13277-015-3473-4.
Gilham, C., C. Rake, G. Burdett, A. G. Nicholson, L. Davison, Landi, S., F. Gemignani, M. Neri, R. Barale, S. Bonassi,
A. Franchini, J. Carpenter, J. Hodgson, A. Darnton, and J. Peto. F. Bottari, P. A. Canessa, F. Canzian, M. Ceppi, R. Filiberti,
2016. Pleural mesothelioma and lung cancer risks in relation to et al. 2007. Polymorphisms of glutathione-s-transferase M1
occupational history and asbestos lung burden. Occup. and manganese superoxide dismutase are associated with the
Environ. Med. 73:290–99. doi:10.1136/oemed-2015-103074. risk of malignant pleural mesothelioma. Int. J. Cancer
Girardelli, M., I. Maestri, R. R. Rinaldi, M. Tognon, 120:2739–43. doi:10.1002/ijc.22590.
R. Boldorini, M. Bovenzi, S. Crovella, and M. Comar. Le Calvez, F., A. Mukeria, J. D. Hunt, O. Kelm, R. J. Hung,
2012. NLRP1 polymorphisms in patients with P. Tanière, P. Brennan, P. Boffetta, D. G. Zaridze, and
asbestos-associated mesothelioma. Infect. Agents Cancer P. Hainaut. 2005. TP53 and KRas mutation load and types
7:25. doi:10.1186/1750-9378-7-25. in lung cancers in relation to tobacco smoke: Distinct pat-
Graffelman, J. 2015. Exploring diallelic genetic markers: The terns in never, former, and current smokers. Cancer Res.
HardyWeinberg package. J. Stat. Software 64:1–23. 65:5076–83. doi:10.1158/0008-5472.CAN-05-0551.
doi:10.18637/jss.v064.i03. Lemen, R. A. 2016. Mesothelioma from asbestos exposures:
Hayward, J. A., A. Mathur, C. Ngo, and S. M. Man. 2018. Epidemiologic patterns and impact in the United States.
Cytosolic recognition of microbes and pathogens: J. Toxicol Environ. Health B 19:250–65. doi:10.1080/
Inflammasomes in action. Microbiol. Mol. Biol. Rev. 82. 10937404.2016.1195323.
doi:10.1128/MMBR.00015-18. Lewis, C. M., and J. Knight. 2012. Introduction to genetic
Hillegass, J. M., J. M. Miller, M. B. MacPherson, C. M. Westbom, association studies. Cold Spring Harb. Protoc.
M. Sayan, J. K. Thompson, S. L. Macura, T. N. Perkins, 2012:297–306. doi:10.1101/pdb.top068163.
S. L. Beuschel, V. Alexeeva, et al. 2013. Asbestos and erionite Liu, C.-Y., I. Stücker, C. Chen, G. Goodman, M. K. McHugh,
prime and activate the NLRP3 inflammasome that stimulates A. M. D’Amelio, C. J. Etzel, S. Li, X. Lin, and
autocrine cytokine release in human mesothelial cells. Part D. C. Christiani. 2015. Genome-wide gene-asbestos expo-
Fibre Toxicol 10:39. doi:10.1186/1743-8977-10-39. sure interaction association study identifies a common sus-
Hirvonen, A., S. T. Saarikoski, K. Linnainmaa, K. Koskinen, ceptibility variant on 22q13.31 associated with lung cancer
K. Husgafvel-Pursiainen, K. Mattson, and H. Vainio. 1996. risk. Cancer Epidemiol. Biomarkers Prev. 24:1564–73.
Glutathione s-transferase and n-acetyltransferase genotypes doi:10.1158/1055-9965.EPI-15-0021.
and asbestos-associated pulmonary disorders. J. Natl. London, S. J., A. K. Daly, J. Cooper, W. C. Navidi, C. L. Carpenter,
Cancer Inst. 88 (24):1853–56. doi:10.1093/jnci/88.24.1853. and J. R. Idle. 1995. Polymorphism of glutathione s-transferase
Inai, K. 2008. Pathology of mesothelioma. Environ. Health m1 and lung cancer risk among African-Americans and
Prev. Med. 13:60–64. doi:10.1007/s12199-007-0017-6. Caucasians in Los Angeles County, California. J. Natl. Cancer
International Agency for Research on Cancer, and Inst. 87:1246–53. doi:10.1093/jnci/87.16.1246.
Weltgesundheitsorganisation, eds. 2012. IARC monographs Markowitz, S. B., S. M. Levin, A. Miller, and A. Morabia. 2013.
on the evaluation of carcinogenic risks to humans, volume 100 Asbestos, asbestosis, smoking, and lung cancer. New findings
JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH, PART A 15

from the North American insulator cohort. Am. J. Respir. Crit. asbestos exposure, cigarette smoking, myeloperoxidase
Care Med. 188:90–96. doi:10.1164/rccm.201302-0257OC. (MPO) genotypes, and lung cancer risk. Am. J. Ind. Med.
Martinon, F., K. Burns, and J. Tschopp. 2002. The inflamma- 42:29–37. doi:10.1002/ajim.10084.
some: A molecular platform triggering activation of Schneider, J., and U. Bernges. 2009. CYP1A1 and cyp1b1 poly-
inflammatory caspases and processing of proIL-beta. Mol. morphisms as modifying factors in patients with pneumoco-
Cell. 10:417–26. doi:10.1016/s1097-2765(02)00599-3. niosis and occupationally related tumours: A pilot study.
Matullo, G., S. Guarrera, M. Betti, G. Fiorito, D. Ferrante, Mol. Med. Rep. 2:1023–28. doi:10.3892/mmr_00000209.
F. Voglino, G. Cadby, C. Di Gaetano, F. Rosa, A. Russo, et al. Shannahan, J. H., A. J. Ghio, M. C. Schladweiler, J. K. McGee,
2013. Genetic variants associated with increased risk of malig- J. H. Richards, S. H. Gavett, and U. P. Kodavanti. 2011.
nant pleural mesothelioma: A genome-wide association study. The role of iron in Libby amphibole-induced acute lung
PLoS ONE 8:e61253. doi:10.1371/journal.pone.0061253. injury and inflammation. Inhal Toxicol 23:313–23.
Mossman, B. T., A. Shukla, N. H. Heintz, C. F. Verschraegen, doi:10.3109/08958378.2011.569587.
A. Thomas, and R. Hassan. 2013. New insights into under- Smith, M. J., and B. Naylor. 1972. A method for extracting
standing the mechanisms, pathogenesis, and management ferruginous bodies from sputum and pulmonary tissue.
of malignant mesotheliomas. Am. J. Pathol. 182:1065–77. Am. J. Clin. Pathol. 58:250–54. doi:10.1093/ajcp/58.3.250.
doi:10.1016/j.ajpath.2012.12.028. Thompson, J. K., M. B. MacPherson, S. L. Beuschel, and
Mossman, B. T. 1990. In vitro studies on the biologic effects A. Shukla. 2017. Asbestos-induced mesothelial to fibroblastic
of fibers: Correlation with in vivo bioassays. Environ. transition is modulated by the inflammasome. Am. J. Pathol.
Health Perspect. 88:319–22. doi:10.1289/ehp.9088319. 187:665–78. doi:10.1016/j.ajpath.2016.11.008.
Mossman, B. T., and A. Churg. 1998. Mechanisms in the Travis, W. D., Brambilla, E., Nicholson, A. G., Yatabe, Y.,
pathogenesis of asbestosis and silicosis. Am. J. Respir. Crit. Austin, J. H. M., Beasley, M. B., Chirieac, L. R., Dacic, S.,
Care Med. 157:1666–80. doi:10.1164/ajrccm.157.5.9707141. Duhig, E., Flieder, D. B., et. 2015. The 2015 world health
Murakami, A., Y. Fujimori, Y. Yoshikawa, S. Yamada, K. Tamura, organization classification of lung tumors. J. Thoracic.
N. Hirayama, T. Terada, K. Kuribayashi, C. Tabata, Oncol. 10:1243–60. doi:10.1097/JTO.0000000000000630.
K. Fukuoka, et al. 2012. Heme oxygenase-1 promoter poly- Tunesi, S., D. Ferrante, D. Mirabelli, S. Andorno, M. Betti,
morphism is associated with risk of malignant mesothelioma. G. Fiorito, S. Guarrera, E. Casalone, M. Neri, D. Ugolini,
Lung 190:333–37. doi:10.1016/j.ajpath.2012.12.028. et al. 2015. Gene-asbestos interaction in malignant pleural
Neri, M., D. Ugolini, I. Dianzani, F. Gemignani, S. Landi, mesothelioma susceptibility. Carcinogenesis 36:1129–35.
A. Cesario, C. Magnani, L. Mutti, R. Puntoni, and doi:10.1093/carcin/bgv097.
S. Bonassi. 2008. Genetic susceptibility to malignant pleural Uhlen, M., Zhang, C., Lee, S., Sjöstedt, E., Fagerberg, L., Bidkhori,
mesothelioma and other asbestos-associated diseases. Mutat. G., Benfeitas, R., Arif, M., Liu, Z., Edfors, F., et. al.
Res. 659:126–36. doi:10.1016/j.mrrev.2008.02.002. 2017. A pathology atlas of the human
Norton, E. C., B. E. Dowd, and M. L. Maciejewski. 2018. cancer transcriptome. Science357. doi:10.1126/science.aan2507
Odds ratios—Current best practice and use. J. Am. Med. Wang, L. I., D. Neuberg, and D. C. Christiani. 2004. Asbestos
Assoc. 320:84–85. doi:10.1001/jama.2018.6971. exposure, manganese superoxide dismutase (MnSod) gen-
Ohar, J. A., M. Cheung, J. Talarchek, S. E. Howard, T. D. Howard, otype, and lung cancer risk. J. Occup. Environ. Med.
M. Hesdorffer, H. Peng, F. J. Rauscher, and J. R. Testa. 2016. 46:556–64. doi:10.1097/01.jom.0000128155.86648.a4.
Germline Bap1 mutational landscape of asbestos-exposed malig- Wei, S., L.-E. Wang, M. K. McHugh, Y. Han, M. Xiong,
nant mesothelioma patients with family history of cancer. Cancer C. I. Amos, M. R. Spitz, and Q. W. Wei. 2012. Genome-
Res. 76:206–15. doi:10.1158/0008-5472.CAN-15-0295. wide gene-environment interaction analysis for asbestos
Pascolo, L., G. Zabucchi, A. Gianoncelli, G. Kourousias, E. Trevisan, exposure in lung cancer susceptibility. Carcinogenesis
E. Pascotto, C. Casarsa, C. Ryan, M. Lucattelli, G. Lungarella, 33:1531–37. doi:10.1093/carcin/bgs188.
et al. 2016. Synchrotron x-ray microscopy reveals early calcium Zhang, W. Z., J. J. Butler, and S. M. Cloonan. 2019. Smoking-
and iron interaction with crocidolite fibers in the lung of exposed induced iron dysregulation in the lung. Free Radic. Biol. Med.
mice. Toxicol. Lett. 241:111–20. doi:10.1016/j.toxlet.2015.11.016. 133:238–47. doi:10.1016/j.freeradbiomed.2018.07.024:.
Ruda, T. A., and P. K. Dutta. 2005. Fenton chemistry of Zhang, Y., J. Shi, X. Liu, L. Feng, Z. Gong, P. Koppula,
Fe(III)-exchanged zeolitic minerals treated with antioxidants. K. Sirohi, X. Li, Y. Wei, H. Lee, et al. 2018. BAP1 links
Environ. Sci. Technol. 39:6147–52. doi:10.1021/es050336e. metabolic regulation of ferroptosis to tumour
Schabath, M. B., M. R. Spitz, G. L. Delclos, G. B. Gunn, suppression. Nat. Cell Biol. 20:1181–92. doi:10.1038/
L. W. Whitehead, and X. Wu. 2002. Association between s41556-018-0178-0.

You might also like