You are on page 1of 11

Journal of Immunotoxicology, 2010; 7(4): 268–278

REVIEW ARTICLE

Dysregulation of the immune system caused by silica and


asbestos
Megumi Maeda1, Yasumitsu Nishimura1, Naoko Kumagai1, Hiroaki Hayashi1, Tamayo Hatayama1,
Minako Katoh1, Naomi Miyahara1, Shoko Yamamoto1, Junichi Hirastuka2, and Takemi Otsuki1
Department of Hygiene, Kawasaki Medical School, Kurashiki, Japan, and 2Department of Radiation Oncology, Kawasaki
1

Medical School, Kurashiki, Japan


Journal of Immunotoxicology Downloaded from informahealthcare.com by Gazi Univ. on 01/09/15

Abstract
Silica and asbestos cause pneumoconioses known as silicosis and asbestosis, respectively, that are each character-
ized by progressive pulmonary fibrosis. While local effects of inhaled silica particles alter the function of alveolar
macrophages and sequential cellular and molecular biological events, general systemic immunological effects
may also evolve. One well-known health outcome associated with silica exposure/silicosis is an increase in the
incidence of autoimmune disorders. In addition, while exposure to silica—in the crystalline form—has also been
seen to be associated with the development of lung cancers, it remains unclear as to whether or not silicosis is
a necessary condition for the elevation of silica-associated lung cancer risks. Since asbestos is a mineral silicate,
it would be expected to also possess generalized immunotoxicological effects similar to those associated with
For personal use only.

silica particles. However, asbestos-exposed patients are far better known than silicotic patients for development
of malignant diseases such as lung cancer and mesothelioma, and less so for the development of autoimmune
disorders. With both asbestos and crystalline silica, one important dysregulatory outcome that needs to be consid-
ered is an alteration in tumor immunity that allows for silica- or asbestos- (or asbestos-associated agent)-induced
tumors to survive and thrive in situ. In this review, the immunotoxicological effects of both silica and asbestos are
presented and contrasted in terms of their abilities to induce immune system dysregulation that then are manifest
by the onset of autoimmunity or by alterations in host-tumor immunity.
Keywords:  Silica; asbestos; autoimmunity; tumor immunity

Introduction collagenous structure, scavenger receptor (SR)-AI and


Silica and asbestos cause pneumoconioses known as silicosis SR-AII.
and asbestosis, respectively, that are characterized by progres- 2. Capture of silica/asbestos by macrophages and entrap-
sive pulmonary fibrosis (Singh and Davis, 2002; Cohen et al., ment within lysosomes.
2008). Inhaled silica particles and asbestos fibers are cap- 3. Activation of nucleotide-binding domain and leucine-
tured by alveolar macrophages, and subsequent cellular and rich repeat containing proteins 3 inflammasome to
molecular cascaded events are known to give rise to increases cleave procaspase 1 to an active form.
in the numbers of collagenic fibroblasts and the appearance 4. Cleavage of prointerleukin-1β (proIL1β) to an active
of laminated fibrous changes at areas surrounding the silica form for release to form fibrotic nodules.
particles and silica-bearing macrophages (Nishimura et al., 5. Production of reactive oxygen species (ROS) and reactive
2007; Hamilton et al., 2008; Thakur et al., 2008; Wells et al., nitrogen species (RNS) in the macrophages.
2009). These sequential cellular events have been analyzed 6. Induction of cellular and tissue damages due to the pro-
by numerous investigators and the following mechanisms/ duction of ROS and RNS.
pathways for these outcomes have been proposed: 7. Apoptosis of the alveolar macrophages.
8. Production of various cytokines/chemokines such as
1. Initial recognition of silica/asbestos by cell mem- IL-1β, tumor necrosis factor-α (TNF-α), macrophage
brane receptors such as the macrophage receptor with inflammatory protein-1/2, monocyte-chemoattractant

Address for Correspondence:  Dr. Takemi Otsuki, Department of Hygiene, Kawasaki Medical School, 577 Matsushima, Kurashiki 701-0192, Japan. E-mail: takemi@
med.kawasaki-m.ac.jp

(Received 20 June 2010; revised 11 July 2010; accepted 29 July 2010)

ISSN 1547-691X print/ISSN 1547-6901 online © 2010 Informa Healthcare USA, Inc.
DOI: 10.3109/1547691X.2010.512579 http://www.informahealthcare.com/imt
Silica/asbestos and immune dysregulation   269

protein-1, and IL-8 to cause chronic inflammation and of data pertaining to select indices of asbestos-induced
proliferation of collagenic fibers. immunomodulation (i.e. induction of autoimmunity), it
9. Release of silica particles and asbestos fibers from alveo- would not be farfetched to suppose that asbestos—if acting
lar macrophages and the repeating of similar cellular akin to silica—might now also facilitate the progression (i.e.
reactions described above by newly-recognizing nearby growth, expansion, metastases) of any newly-transformed
macrophages. cells by disrupting a host’s normal abilities to recognize/
10. Transfer to silica particles and (partially cleaved) asbes- remove these cells, i.e. by altering host-tumor immunity.
tos fibers to regional lymph nodes. It remains to be determined if silica (as crystalline form)
also is able to alter host-tumor immunity. Indeed, there is
As these cellular and molecular reactions are continuously ample evidence that exposure to crystalline silica is associ-
repeated, pulmonary fibrosis will gradually and progressively ated with an increase in incidence of lung cancers among
appear (Nishimura et al., 2007; Hamilton et al., 2008; Thakur workers in several occupations (see International Agency
et al., 2008; Wells et al., 2009). for Research on Cancer IARC 1977; Brown, 2009; Erren et al.,
Apart from suffering from pulmonary fibrosis, it is well 2009; Lacasse et al., 2009). There is also a pool of literature
known that silicosis patients (SPs) also often evince com- that indicates that exposure to silica [by inhalation/other
Journal of Immunotoxicology Downloaded from informahealthcare.com by Gazi Univ. on 01/09/15

plications from autoimmune diseases such as rheumatoid routes (injections)] can affect host resistance to injected/
arthritis (known as Caplan syndrome), systemic lupus ery- implanted syngeneic and non-syngeneic tumors (Keller,
thematosus, systemic scleroderma, and/or anti-neutrophil 1976; Fuji and Murakami, 1983; Sandstrom and Chow, 1987;
cytoplasmic autoantibody-related vasculitis/nephritis Gresser et  al., 1990); unfortunately, changes in resistance
(Uber and McReynolds, 1982; Steenland and Goldsmith, against de novo in situ neoplasias have not been evaluated/
1995; Shanklin and Smalley, 1998). The effect of silica on the reported in the literature to the same degree. In many of these
human immune system has been considered to be a result, in cited studies, it appeared that the timing of exposure to silica
part, of the potential adjuvant activity of silica. Additionally, in relation to introduction of the tumor cells was critical, i.e.
the increasing extracellular presence of various autoanti- silica treatment had to occur concurrently (or near-so) with
gens such as DNA, RNA, and other organelles released from the implantation for any significant effect to manifest. This
For personal use only.

apoptotic alveolar macrophages that had phagocytized silica suggests that for silica-induced tumors to flourish in a host via
particles accordingly increase the opportunities for reactive any asbestos-like ‘altered tumor immunity’ route, the silica
T-lymphocytes to encounter these autoantigens (Nishimura exposures would have to ongoing/continuous in nature (as
et al., 2007; Hamilton et al., 2008; Thakur et al., 2008; Wells opposed to the effect evolving from silica that had accumu-
et al., 2009). lated in the lungs in the past).
As asbestos (i.e. as actinolite, amosite, anthophyllite, This review does not seek to educate the reader on the
chrysotile, crocidolite, or tremolite) is a mineral silicate con- differing mechanisms underlying how/why various forms of
taining iron, magnesium, and calcium, it is not reasonable to silica differ in cell transforming potentials nor how/why crys-
question whether exposure to asbestos also causes immuno- talline silica differs from asbestos in these aspects. Instead,
logical alterations like (immunomodulatory) silica (Uber and this review provides a summary of our (and others’) investiga-
McReynolds, 1982; Steenland and Goldsmith, 1995; Shanklin tions regarding the immunotoxicological effects of silica and
and Smalley, 1998). To date, only a handful of reports have asbestos as they pertain to autoimmunity and to disruptions
been published detailing autoimmune disorders in asbestos- of host-tumor immunity. With regard to the latter, it is hoped
exposed patients (Tellesson, 1961; Pfau et al., 2005; Noonan the information here will allow readers to glean a clearer
et al., 2006). The most typical health complications arising understanding of any potential differences in mechanisms
from asbestos exposure involve the development of malig- that enable the post-transformation growth/progression of the
nant tumors associated with mesothelioma and lung cancers cancers that each agent can induce in situ.
(Nicholson, 1984, 2001; Antman, 1986; Gruber, 1990; Niklinski
et al., 2004). In addition, the incidence of solitary tumors of
Silica particles and dysregulation of immune
the larynx, bladder, and gastrointestinal tracts are reported
response leading to autoimmunity
to occur at a higher level among asbestos-exposed individu-
als than in the non-exposed population (Rolston and Oury, As mentioned above, SPs often manifest a disturbance of the
2004). immune system as autoimmunity. Silica is considered one of
With respect to these various types of cancers, it is well- the most important environmental substances, in the fash-
established that the asbestos itself can induce malignant ion of vinyl chloride and other chemicals (including trichlo-
transformation of cells by various mechanisms. These can roethene and epoxy resins), that give rise to autoimmune
include an enhanced formation (non-enzymatic) of ROS and diseases in exposed hosts (Trice and Pinals, 1985; Goldman,
RNS that is facilitated by the iron present in asbestos, direct 1996; D’Cruz, 2000; Hess, 2002; Cooper et al., 2009).
physical disruption of spindle bodies during cell division, To ascertain how silica particles might be inducing
and/or adhesion of various mutagens to the asbestos fibers autoimmune outcomes in exposed hosts, our research first
(Lee, 1985; Rom et al., 1991, Brandt-Rauf et al., 1994, Partanen focused its attention on the Fas/CD95 death receptor and
et  al., 1994; Toyokuni, 1996, 2009). Despite the paucity related molecules. This is because Fas/CD95 is known as
270   M. Maeda et al.

one of the most important molecules for the induction of and these cells may have developed out of an excessive
apoptosis of lymphocytes. Furthermore, animal models pos- transcription of the alternatively-spliced Fas gene and other
sessing a mutated Fas gene or Fas ligand gene (i.e. mlr or gld variant messages. Therefore, these cells may be resistant to
mice respectively), display various clinical manifestations the functional anti-Fas autoantibody, secrete higher levels
of autoimmunity (Watanabe-Fukunaga et al., 1992; Watson of soluble Fas, DCR3, and spliced variants, and are resistant
et al., 1992; Sobel et al., 1993; Wu et al., 1993; Takahashi et al., to Fas-mediated apoptosis (Otsuki et al., 2003, 2006, 2007;
1994; Nagata and Suda, 1995). As such, using clinical samples Murakami et  al., 2009). As reported previously, patients
derived from SPs, we examined alterations of Fas protein and with a weaker MFI of membrane Fas often have a higher
related molecules in their blood and immune cells. All of titer of anti-nuclear antibodies, and self-recognizing clones
the patients were Japanese brickyard workers in Bizen City in silicosis may be included in the fraction because these
(Okayama prefecture, Japan), and were monitored at either clones may survive longer and show resistance to apopto-
Kusaka Hospital or the Hinase-Uragami Clinic. The silica in sis. The other population represents stronger membrane Fas
the materials handled by these workers (e.g. dirt, sand, mud, expressers that: may be sensitive to Fas-mediated apoptosis
concrete), and thus present as a potential risk for being including cell death caused by anti-Fas autoantibody; show
inhaled by these individuals in their work environment, was a reduced expression of intracellular inhibitor genes of Fas-
Journal of Immunotoxicology Downloaded from informahealthcare.com by Gazi Univ. on 01/09/15

estimated to reach levels as high as 40–60% (by mass). The mediated apoptosis; and, undergo apoptosis. These cells
subjects were diagnosed with pneumoconiosis according to may be recruited from bone marrow after reaching the final
the International Labor Organization (ILO) 2000 Guideline stage of cell death. This recruited fraction would not have
(ILO, 2002). These patients displayed neither clinical symp- encountered silica and would be sensitive to silica/silicate-
toms related to autoimmune diseases (e.g. sclerotic skin, induced apoptosis. As a result, cells in this fraction would
Raynaud’s phenomenon, facial erythema, or arthralgia) nor be continuously undergoing renewal and then apoptosis
any cancers. The following findings were obtained from these (Otsuki et al., 2003, 2006, 2007; Murakami et al., 2009).
patients (see Figure 1): According to recent developments concerning regula-
tion of T-lymphocyte reactions against intrinsic or foreign
1. Detection of autoantibody to Fas and caspase-8, as well antigens, a modification of the population (i.e. numbers)/
For personal use only.

as topoisomerase I and desmoglein (Ueki A et al., 2001, functions of CD4+CD25+ forkhead box P3 (FoxP)+ regulatory
2002; Ueki H et al., 2001; Takata-Tomokuni et al., 2005). T-lymphocytes (Treg) may be a very important factor for the
2. Anti-Fas autoantibody detected in SPs was functionally manifestation of several antigen-antibody-based events,
active and caused Fas-mediated apoptosis (Takata- including autoimmunity, allergy, tumor immunity, resistance
Tomokuni et al., 2005). to infection, graft-versus-host diseases, and control of preg-
3. The level of serum soluble Fas was higher in SPs nancy (Fehérvari and Sakaguchi, 2004; Hori and Sakaguchi,
than in healthy donors (HDs), although the level of 2004; Sakaguchi, 2005; Dejaco et al., 2006; Sakaguchi et al.,
serum soluble Fas ligand did not differ between SPs and 2006; Oh et al., 2010). In particular, the maintenance of hyper-
HDs (Tomokuni et al., 1997, 1999). reactivity to autoantigens may be caused, in part, by a reduc-
4. The mean fluorescent intensity (MFI) of membrane tion in the functions/numbers of Treg in a host. As a result of
Fas was lower with lymphocytes from SPs than from HDs, their impact upon both autoimmunity and tumor immunity,
although total numbers of Fas-positive lymphocytes we determined it was critical to examine Treg status (i.e. func-
(membrane Fas expression) did not differ between the tions/numbers) in hosts suffering from silicosis.
two populations (Otsuki et al., 2006). Since FoxP3+ cells cannot be readily collected for use in
5. The weaker membrane Fas expressers (among assays of biological function (as FoxP3 is an intranuclear
lymphocytes) were identified to be weaker Fas message molecule and the cell membrane has to be destroyed [i.e.
expressers (Otsuki et al., 2006). permea-bilized] to detect it), we analyzed CD4+CD25+ cells
6. The gene expression levels of extracellular inhibitor present within PBMC derived from SPs and HDs. Overtly,
competing membrane Fas–Fas ligand binding such as it seemed that the percentage of PMBC associated with
soluble Fas, decoy receptor 3 (DCR3), and other alter- CD4+CD25+ fraction did not differ between the two groups;
natively-spliced variants of the Fas gene were higher in however, when the samples were analyzed in the context
peripheral blood mononuclear cells (PBMC) from SPs of/corrected for donor age, the values were in fact lower in
than HDs (Otsuki et al., 2000a, b). the SP group than among the HD samples (Wu et al., 2006).
7. The intracellular apoptosis-inhibitory genes including In addition, the results of the functional assays using these
I-FLICE, sentrin, survivine, and ICAD showed a lower same isolates (i.e. non-permeabilized) showed that the
expression in PBMC from SPs than HDs (Otsuki et al., CD4+CD25+ cells from SP hosts possessed a reduced sup-
2000c; Guo et al., 2001). pressive effect against alloantigen-stimulated responder
T-lymphocyte (CD4+CD25− cells) proliferation. Based on
Although significant mutations of Fas and Fas ligand genes these endpoints, it was concluded that Treg function and
were not detected, these results indicated that two popula- total numbers in the peripheral CD4+CD25+ fraction was
tions of lymphocytes may exist in the peripheral blood of likely to have been reduced as a result of a worker’s chronic
SPs. One population is a weaker membrane Fas expresser exposure to silica.
Silica/asbestos and immune dysregulation   271

Silica

Functional Anti-Fas autoantibody Fas Ligand

Membrane Fas
Other
variant
FADD messa
ges of
Fas tra
DcR3 nscript
Caspase 8 I-Flice

Sentrin Soluble
Fas
Caspase 3
Survivin

CAD/ ICAD

CAD Reduced apoptotic signals


Journal of Immunotoxicology Downloaded from informahealthcare.com by Gazi Univ. on 01/09/15

DNA fragmentation
Apoptosis Long-term survival
(including self-recognizing clones)
Fraction repeating apoptosis caused by silica and recruitment from bone marrow
CD4+CD25−responder T-lymphocytes
CD4+CD25+FoxP3+ Treg

Chronic Activation
CD4+CD25+PD-1+chronically-
Treg Fas/CD95+++ activated responder T-lymphocytes
Peripheral
CD4+CD25+
For personal use only.

Excess loss fraction Entry/substitution

Reduced function Activation of Autoimmunity

Figure 1.  Schematic models of alteration in Fas and Fas-related molecules found in silicosis and speculated change of the peripheral CD4+CD25+ fraction
in which the regulatory T-lymphocytes (Treg) should exist. CAD, caspase-activated deoxyribonuclease; DCR3, decoy receptor 3; ICAD, inhibitor of cas-
pase-activated deoxyribonuclease; I-FLICE, inhibitor of FADD-like interleukin-1β-converting enzyme; FADD, Fas-associated death domain protein.

However, as we reported previously, silica can stimulate 2. Responder T-lymphocytes chronically-activated by silica
peripheral T-lymphocytes in vitro—as monitored by CD69, an become CD4+CD25+ (PD-1+) expressers.
early activation marker of T-lymphocyte expression (Hyodoh 3. Treg activated by silica express higher Fas/CD95 and are
et  al., 2005). If responder T-lymphocytes were activated sensitive to Fas-mediated apoptosis.
chronically, their surface marker expression will become 4. After an ongoing progression of these events, the com-
CD4+CD25+ and would be similar to the natural Treg. As a position of the peripheral CD4+CD25+ fraction in SPs
result, the peripheral CD4+CD25+ fraction may include true changes to reflect a loss of Treg (by Fas-mediated apop-
Treg and chronically-activated responder T-lymphocytes. tosis) and a gain of activated responder T-lymphocytes;
To validate the possibility that the CD4+CD25+ fraction of this reduction of Treg function makes SPs more sensitive
SPs includes Treg and activated responder T-lymphocytes, to a disruption of self-tolerance.
we analyzed programmed cell death 1 (PD-1) expression,
as an activated T-lymphocyte marker, in CD4+CD25− and
CD4+CD25+ fractions from SPs and HDs. Results showed Disruption of tumor immunity due to silica
that PD-1 was highly expressed in both the CD4+CD25+ and
CD4+CD25− fractions isolated from SPs, while both frac- Based on what is known about silica and its impact on
tions recovered from the HDs hardly expressed this marker. both responder T-lymphocytes and Treg cells, it would not
Furthermore, Treg CD4+CD25+FoxP3+ cells from SPs revealed be unreasonable to expect that silica-exposed individu-
higher membrane Fas expression than did the cells from als should have an increased response against tumor cells
HDs. Taken together with our previous Fas and Fas-related that form in situ (i.e. with fewer Treg present, anti-tumor
analyses, the following speculations arose (and which we are responses should not be down-regulated). Yet, exposure to
currently investigating): crystalline silica is correlated with increases in lung cancer
formation. Thus, it might be that the impact of silica (in the
1. Silica activates both responder T-lymphocytes and Treg context of altered tumor immunity) might be more ‘critical’
(as shown in Figure 1). at the level of the macrophage.
272   M. Maeda et al.

The potential for silica to induce altered macrophage the timeframe in which exposure correlates with reductions
function and so affect neoplastic cell removal is clear. For in tumor resistance seems to be confined to a period where
example, Rakhmilevich et al. (Buhtoiarov et al., 2006; Lum cells levels are increased.
et al., 2006) showed that while macrophages could be acti- It may be that the Fas/FasL could affect the lung macro-
vated by CD40 ligation to be cytotoxic against tumor cells phages so that they might not be able to activate themselves
in vitro and in vivo—and that anti-CD40 monoclonal anti- (autocrine) or other cells critical to recognizing/remov-
body (mAb) therapy could lead to inhibited tumor growth ing any newly-transformed cells. Unfortunately, effects of
even in the absence of T-lymphocytes, natural killer (NK) increased FasL expression on macrophage capacity to release
cells, and neutrophils [polymorphonuclear leukocytes proinflammatory appear to be variable; in some cases, the
(PMN)]—this effect of anti-CD40 mAb was inhibited by spontaneous release of TNF-α is increased by silica (and the
silica (injections). A ready explanation for those findings subsequent FasL presence; see Borges et al., 2001) while in
may be that silica simply caused reductions in macrophage other studies, the absence of FasL (i.e. demonstrated using
levels. However, as noted by Keller (1976), exposure to silica lpr/lpr vs. wild-type mice) results in greater spontaneous
leads to increases in macrophage numbers (albeit in tran- release of this and other cytokines (Brown et al., 2004) key
sitory manner). Furthermore, a hallmark of silicosis is that to inflammation and anti-tumor activities. However, as the
Journal of Immunotoxicology Downloaded from informahealthcare.com by Gazi Univ. on 01/09/15

silica induces macrophage (and PMN) infiltration into the overwhelming evidence in the literature supports the former
lungs and subsequent over-production of proinflammatory scenario (see reviews by Castranova, 2004 and Rimal et al.,
cytokines, chemokines, and ROS that eventually give rise to 2005), and an increased release in cytokines/chemokines
the associated pathologies. would help move the lungs towards a silicotic state, we are
If it is not an effect on macrophage numbers per se that left to wonder why these newly-recruited immune cells
may be key to any reduction in host-tumor immunity after (macrophages and PMN) would be ineffective against any
silica exposure, then an impact on one/more critical cell now-present transformed cells.
function(s) is likely. In one study, mice administered silica (by To this end, we believe there are at least two means
intraperitoneal injection) displayed variable cell type-specific by which the expected effects of an increased presence
effects, i.e. accumulation of splenic macrophages and PMN, of Fas/FasL might be negated in a silica-exposed host’s
For personal use only.

reductions in B-lymphocyte levels, and modest changes in lungs (thereby mitigating any impact on macrophage
T-lymphocyte abundance (Rao and Frey, 1998). Interestingly, numbers/activity against tumor cells). First, the presence
while silica did not affect the spontaneous release of TNF-α of a DCR3 (or analogue) has been shown to abrograte
or IL-1β by local (i.e. peritoneal) macrophages, IL-12 release the ability of FasL to induce inflammatory responses in
was stimulated ≈ 5-fold; further, the ability of these cells to the lungs of mice (Wortinger et  al., 2003). As we noted
present antigen to T-lymphocytes (in vitro) or to prime anti- earlier, the gene expression levels of extracellular inhibi-
gen-specific T- and B-lymphocytes was greatly inhibited. This tors of FasL binding to receptors (like soluble Fas and
data suggests that silica could impact negatively on one set DCR3) were higher in PBMC from SPs than in cells from
of macrophage functions critical to anti-tumor responses, i.e. HDs (Otsuki et al., 2000a, b, 2006). Second, an increased
antigen processing and presentation—even during a concur- presence of transforming growth factor-β (TGF-β) could
rent increase in release of an important anticancer cytokine, result in a shift of FasL from an inflammatory to a sup-
IL-12 (see Su et al., 2001, Weiss et al., 2007). However, given pressive function. Indeed, a critical study by Chen et al.
the documented shift toward induction of autoimmune (1998) showed that an increased presence of TGF-β not
responses in silica-exposed hosts, reductions in macrophage only inhibited neutrophil (and likely macrophage) activa-
antigen processing/presentation seem incongruous. Absent tion by FasL, but also led to a reduction in tumor (cell)
a reduction in local macrophage number/capacity to act as rejection in the treated host. There is ample evidence that
antigen-presenting cells, it seems that modulation of other there is an increase in TGF-β expression in the lungs of
key tumor-killing functions—either directly or indirectly— silica-exposed hosts (Williams et al., 1993; Jagirdar et al.,
would therefore underlie any change(s) in tumor immunity 1996; Ji et al., 2003a, b; Barbarin et al., 2004). If the TGF-β
in silica-exposed hosts. can mitigate effects of FasL so as to: (1) leave macrophage
Because of their role in affecting lymphocyte function, levels intact by diminishing induction of apoptosis; (2)
the question arises as to if silica-induced increases in Fas/ lessen the release of select cytokines and/or chemokines
FasL expression might impact on lung macrophages and so that are essential to mounting an anti-tumor response (as
reduce their role in tumor immunity. It is well known that well as being proinflammatory); and, (3) cause a shift in
inhaled silica causes increased FasL expression in the lungs FasL-associated activities from an inflammatory to a sup-
(Borges et al., 2001, 2002; Corsini et al., 2003; Zhang et al., pressive function with other immune cell types needed
2006). Consequently, an eventual increase in apoptotic death to recognize and/or remove (silica-induced) transformed
among macrophages in an exposed host’s lungs is expected cells, this would provide a reasonable explanation for the
that, with respect to tumor immunity, should then allow ‘time-sensitive’ (re: exposure-to-tumor cell appearance)
any silica-transformed cells that evolve in situ to remain findings of Keller (1976) and those of other investigators
unharmed and able to proliferate. As noted above, while silica noted above, as well as for how silica might disrupt tumor
exposure leads to eventual diminution of macrophage levels, immunity in silicotic patients.
Silica/asbestos and immune dysregulation   273

Disruption of tumor immunity due to asbestos receptors were also decreased. The decreased expression of
fibers these activating receptors also led to reduced phosphoryla-
tion of extracellular signal-regulated kinases in the NK cells.
The immunocompetent cells that directly kill tumor cells dur- Although the asbestos-exposed cell line displayed decreases
ing a normal immune response are the NK cells and CD8+ in NKG2D and 2B4 expression, freshly-isolated NK cells
cytotoxic T-lymphocytes (CTL; Herberman, 1985; Kimber, derived from mesothelioma patients and ex vivo expanded
1985; Chambers et al., 1998; van Baarle et al., 2002; Snyder NK cells (from HDs) cultured with asbestos showed reduced
et  al., 2003; Raulet and Guerra, 2009). There may be an expression of another activating receptor, NKp46. Regardless
asbestos-induced reduced function of these cells, and these of the test system employed, our studies clearly revealed that
modifications of tumor immunity may make patients who asbestos exposure decreased the killing activity of NK cells, in
have been exposed to asbestos more sensitive to tumor devel- part, by causing a reduced expression of cell surface activat-
opment after a latent period of 30–40 years (Nicholson, 1984, ing receptors (Nishimura et al., 2009a, b, c; see Figure 2).
2001; Antman, 1986; Gruber, 1990; Niklinski et al., 2004). In addition to the above-noted endpoints, our laboratories
To analyze the effects of chrysotile asbestos (classified in have also been investigating the effects of asbestos exposure
serpentine group of phyllosili-cates and the most commonly on CTL function, differentiation and proliferation regard-
Journal of Immunotoxicology Downloaded from informahealthcare.com by Gazi Univ. on 01/09/15

encountered form of asbestos) on human NK cells, we first ing the role of various cytokines such as IL-10, TNF-α, and
utilized the human NK cell line, YT-A1 (kindly provided by interferon-γ (IFN-γ). Although the results are preliminary, it
Prof. Yodoi, Kyoto University, Japan). In our studies, YT-A1 seems that asbestos has an effect by reducing CTL function.
cells were continuously exposed to chrysotile asbestos and The details of these investigations will be presented soon (i.e.
the cytotoxic activity of and expressions of killing-related awaiting peer-review and publication).
molecules by the cells (now designated as YT-CB5) were In addition to the above-mentioned studies and endpoints,
examined. YT-CB5 cells revealed significant reductions in our laboratories have also been investigating the effects of
surface expression levels of two NK-cell specific activating asbestos on T-lymphocyte lineages. To establish a chronic and
receptors, e.g. NKG2D and 2B4, and of the intracellular ser- low-dose exposure model similar to that of asbestos-exposed
ine protease granzyme A that is normally secreted by the NK patients, we employed a human T-lymphocyte lymphotropic
For personal use only.

cells to kill tumor cells. In addition, the numbers/intensity of virus type 1-immortalized human polyclonal T-lymphocyte
degranulation events normally stimulated via these activating line (i.e. MT-2). This line was selected, in part, because it is

Effects of exposure Immunoediting by tumor cells


asbestos Malignant tumors
exposure
em
st

T-lymphocyte
sy
e

Macrophage Dendritic
un
m

Tumor cells
Im

cell

NK cells

NKG2D
2B4 NKp46 Tumor killing
granzyme
DAP10 CD3z CD107a
perforin

degranulation
Src-family PI3K
kinase NK cells from
mesothelioma

Direct effects PP2 wortmannin


Primary NK cells
exposed to asbestos
Cell-killing
MEK granules

NKcells ERK YT-CB5 NKcell line

p-ERK • Microtubule-organizing center (MTOC)


• Granule polarization

Figure 2.  Effects of asbestos on human natural killer (NK) cells. The figure illustrates the down-regulation of NK cell-activating surface receptors such
as 2B4, NKG2D, and NKp46, and a decrease in the levels (released levels) of the tumor-killing secreted molecule granzyme A, as found in various experi-
ments using a cell line, as well as with freshly-isolated NK cells from healthy donors or patients with malignant mesothelioma.
274   M. Maeda et al.

not tumor-cell derived and it is reported to possess a normal the MT-2 cells to chrysotile, this new continuously-exposed
karyotype. Additionally, screening for sensitivity to chrysotile MT-2 subline was found to have acquired a resistance to
asbestos revealed that the MT-2 cells and a few Epstein–Barr asbestos-induced apoptosis. These cells also were found to
virus-immortalized B-lymphoblastoid cell lines were rela- display an: increase in activation of Src-family kinases; excess
tively more sensitive to cell death in comparison to other expression and secretion of IL-10; increased activation/phos-
T- and B-lymphocyte tumor-derived lines. As it has been sug- phorylation of signal transducer and activator of transcrip-
gested that effects on T-lymphocytes may be more important tion 3 by utilization of over-produced IL-10 (via autocrine
than those on B-lymphocytes (regarding alteration of tumor mechanisms); and, over-expression of the anti-apoptotic
immunity), this was another key reason the MT-2 cells were molecule, Bcl-2. In addition, the continuously-exposed MT-2
chosen for use in our in vitro experiments (Miura et al., 2006, subline were found to evince an excess production of TGF-β
2008; Nishimura et al., 2006; Maeda et al., 2008; Otsuki et al., and reduced production of IFN-γ, TNF-α, and IL-6. Moreover,
2009). this subline displayed an over-expression of multiple forms of
In our experiments, short-term (i.e. 1–4 days) and high- the T-lymphocyte Vβ receptors, including Vβ 5.2, 9, 13.2, 14,
dose exposure of the MT-2 cells to chrysotile asbestos caused and 21.3, suggesting that asbestos imparted a super-antigenic
progressive time- and dose-dependent apoptosis. This out- activity upon human T-lymphocytes. Parts of these findings
Journal of Immunotoxicology Downloaded from informahealthcare.com by Gazi Univ. on 01/09/15

come was seen to occur in conjunction with activation of the were confirmed using clinical samples derived from patients
mitochondrial apoptotic pathway, production of ROS, activa- with pleural plaque, for whom there was evidence of past
tion of proapoptotic signaling via p38 and c-Jun N-terminal asbestos exposure or malignant mesothelioma. For instance,
kinase (among the mitogen-activated protein kinase path- plasma levels of IL-10 and TGF-β and gene expression of bcl-2
ways), as well as activation of caspases-9 and -3. These results in peripheral CD4+ T-lymphocytes were seen to be elevated
were similar to the cellular changes that occurred in alveolar in samples from SPs compared to samples from HDs, results
epithelial and pleural mesothelial cells exposed in vitro to that were in accordance with those suggested by the results
asbestos as shown in many previous reports (BéruBé et al., seen with the chronically-exposed MT-2 cells (Miura et al.,
1996; Broaddus et al., 1996; Fung et al., 1997; Ollikainen et al., 2006, 2008; Nishimura et al., 2006; Maeda et al., 2008; Otsuki
2000; Aljandali et al., 2001; Buder-Hoffmann et al., 2001). et al., 2009; see Figure 3).
For personal use only.

We also examined the effects from a prolonged exposed to Although the effects of asbestos on Treg cell function
the asbestos agent. After ≈ 1 year of continuous exposure of and numbers still need to be analyzed directly, most of

Short-term high-dose exposure Asbestos Long-term low-dose exposure


fibers
IL-10
secretion Autocrine
stimulation
Vα Vβ
Cβ Cβ IL-10
receptor

JNK/p38 Activation Multiple (not


Src family
ROS clonal) over-
expression, of kinase
production Cytoplasm
TcR-Vβ STAT3 Activation
Activation

cytochrome-c Release
Bc l -2 Bax Bc l -2 Bax

Caspase-9
Mitochondria Activation
Caspase-3

Nucleus
IL-10 up-regulation

Apoptosis Resistance to asbestos-induced apoptosis


Long-term survival
Activation of multiple TcR-Vβ repertoires resembling
superantigen exposure

Figure 3.  Experimental findings of the immunological effects of asbestos-induced by temporary high-dose exposure and continuous low-dose exposures
of the human T-lymphocyte line, MT-2.
Silica/asbestos and immune dysregulation   275

our findings indicated that chronic and low-dose exposure form of particle is known to be associated with increases in
to asbestos by immunocompetent cells would likely cause the incidence of lung cancers. To that end, in this review, we
reductions in a host’s-tumor immunity. Such findings would have sought to present the reader with the most up-to-date
help to explain why many patients who had been exposed to information concerning the immunotoxicological effects of
asbestos appeared to also be sensitive to the growth, expan- both silica and asbestos—with regard to how silica can facili-
sion, and/or eventual metastatic spread of malignantly- tate development of an immune dysregulation that leads to
transformed cells in their bodies. autoimmunity and how both agents might target different
immune cell types and cause changes in their respective
functions that ultimately could lead to a reduction in immu-
Conclusion nity against tumors induced by these agents themselves.
While the IARC has long-recognized asbestos (i.e. as A summary of the findings described in this review is
actinolite, amosite, anthophyllite, chrysotile, crocidolite, provided in Figure 4. Recent advances in immunomolecu-
or tremolite) and silica (crystalline, not amorphous, form) lar studies have led to detailed analyses of the immunoto-
as carcinogens, and the literature is replete with studies of xicological effects of asbestos and silica. Both agents affect
the immunomodulatory effects of silica (especially autoim- immunocompetent cells and these effects may be associated
Journal of Immunotoxicology Downloaded from informahealthcare.com by Gazi Univ. on 01/09/15

mune disorders), it still remains unclear whether asbestos is with the pathophysiological development of complications in
also a potential immunomodulant. Furthermore, if asbestos silica- and asbestos-exposed patients, such as the occurrence
does impact upon host immune function, it remains to be of autoimmune disorders and malignant tumors. Ongoing
determined if one key set of functions is the host immune immunological analyses in our laboratories and those of
response against neoplasia (i.e. tumor immunity). Indeed, other investigators may eventually lead to the discovery of
because of its strong capacity to induce cell transformations new clinical tools such as cell-mediated therapies, treatments
in situ, one needs to consider if the asbestos also plays any with various cytokines/chemokines, and molecule-targeting
role in altering tumor immunity in these now ‘expected to be therapies that could be used to insure a better regulation
tumor-prone’ individuals. This outcome is also important to of autoimmunity or a mitigation of the dysregulation that
determine with respect to crystalline silica as exposure to this leads to altered tumor immunity that seem to be major
For personal use only.

Experimental high-dose and short-term Experimental low-dose and long-term


exposure exposure

Asbestos
•Crocidolite
Hydrates
•Amosite Silicates Silica
Lymphocytes
•Chrysotile
T-lymphocytes

Alveolar epithelial cells


Pleural mesothelial cells Chronic and recurrent long-term exposure

Production of ROS & RNS Alveolar Lymphocytes NK cells


Peripheral blood T-
Activation of mitochondrial macrophages T-lymphocytes
lymphocytes from
apoptotic pathway silicosis patients
Reduction of activation Peripheral blood
signal T-lymphocytes
TGFβ, IL-1β, TNFα •Reduced expression of from healthy donors
DNA damage secretion activating receptors
Apoptosis (NKp46, NKG2D, 2B4) CD4+CD25+Regulatory
•Decreased granzyme A T-lymphocytes

CTL Induction of early


Accumulation of DNA damage activation marker, CD69,
expression
Pulmonary Acquisition of
Escape from apoptosis Dysregulation of Fas
fibrosis apoptosis resistance
(including genetic alteration) molecule
•IL-10 over-production
•Bcl-2 over-expression
β
•Multiple activation of
TcR-Vβ repertoire-like Dysfunction?
super-antigen exposure Substituted by chronically-activated
•Others T-lymphocytes
Lung cancers

Reduction in tumor
Mesothelioma immunity
Dysregulated autoimmunity
Various autoantibodies
Other cancers
Complicated autoimmune diseases

Figure 4.  Summary of immunotoxicological alterations induced by silica and asbestos. CTL, cytotoxic T-lymphocytes; NK cells, natural killer cells; ROS,
reactive oxygen species; RNS, reactive nitrogen species; TGF-β, transforming growth factor-β;TNF-α, tumor necrosis factor-α.
276   M. Maeda et al.

pathophysiological outcomes resulting from exposures to Cohen, R. A., Patel, A. and Green, F. H. 2008. Lung disease caused by exposure
to coal mine and silica dust. Semin. Respir. Crit. Care. Med. 29:651–661.
these two types of airborne particulate pollutants. Cooper, G. S., Makris, S. L., Nietert, P. J. and Jinot, J. 2009. Evidence of autoim-
mune-related effects of trichloroethylene exposure from studies in mice
and humans. Environ. Health. Perspect. 117:696–702.
Declaration of interest Corsini, E., Giani, A., Lucchi, L., Peano, S., Viviani, B., Galli, C. L. and
Marinovich, M. 2003. Resistance to acute silicosis in senescent rats: role
The experimental results performed by us and presented of alveolar macrophages. Chem. Res. Toxicol. 16:1520–1527.
partly in this review were supported by Special Funds for D’Cruz, D. 2000. Autoimmune diseases associated with drugs, chemicals and
Promoting Science and Technology (H18-1-3-3-1), JSPS environmental factors. Toxicol. Lett. 112-113:421–432.
Dejaco, C., Duftner, C., Grubeck-Loebenstein, B. and Schirmer, M. 2006.
KAKENHI (22790550, 22700933, 20390178, 20890270, Imbalance of regulatory T cells in human autoimmune diseases.
19689153, 19790431, 19790411, 18390186, 16390175 and Immunology 117:289–300.
09670500), the Takeda Science Foundation (Tokutei Kenkyu Erren, T. C., Glende, C. B., Morfeld, P. and Piekarski, C. 2009. Is exposure to
silica associated with lung cancer in the absence of silicosis? A meta-an-
Josei I, 2008), and a Research Grant from the Project for Young alytical approach to an important public health question. Int. Arch. Occup.
Investigator 2007 from the Japanese Society of Hygiene, Environ. Health. 82:997–1004.
Kawasaki Medical School Project Grants (16-212S, 16-401N, Fehérvari, Z. and Sakaguchi, S. 2004. Development and function of CD25+CD4+
regulatory T-cells. Curr. Opin. Immunol. 16:203–208.
17-210S, 17-404M, 17-611O, 18-209T, 18-403, 18-601, 19-205Y,
Journal of Immunotoxicology Downloaded from informahealthcare.com by Gazi Univ. on 01/09/15

Fuji, H. and Murakami, M. 1983. Differential tumor immunogenicity of DBA/2


19-506, 19-407M, 19-603T, 20-412I, 20-210O, 20-109N, mouse lymphoma L1210 and its sublines. III. Control of host resistance to
20-402O, 20-410I, 20-410I, 21-401 and 21-107). drug-resistant L1210 sublines by H-2-linked and non-H-2-linked genes. J.
Natl. Cancer Inst. 70:119–125.
Fung, H., Kow, Y. W., Van Houten, B. and Mossman, B. T. 1997. Patterns of
References 8-hydroxydeoxyguanosine formation in DNA and indications of oxida-
tive stress in rat and human pleural mesothelial cells after exposure to
Aljandali, A., Pollack, H., Yeldandi, A., Li, Y., Weitzman, S. A. and Kamp, D. W. crocidolite asbestos. Carcinogenesis 18:825–832.
2001. Asbestos causes apoptosis in alveolar epithelial cells: role of iron- Goldman, J. A. 1996. Connective tissue disease in people exposed to organic
induced free radicals. J. Lab. Clin. Med. 137:330–339. chemical solvents: systemic sclerosis (scleroderma) in dry cleaning plant
Antman, K. H. 1986. Asbestos-related malignancy. Crit. Rev. Oncol. Hematol. and aircraft industry workers. J. Clin. Rheumatol. 2:185–190.
6:287–309. Gresser, I., Maury, C., Carnaud, C., De Maeyer, E., Maunoury, M. T. and
Barbarin, V., Arras, M., Misson, P., Delos, M., McGarry, B., Phan, S. H., Lison, D. Belardelli, F. 1990. Anti-tumor effects of interferon in mice injected with
and Huaux, F. 2004. Characterization of the effect of interleukin-10 on sili- interferon-sensitive and interferon-resistant Friend erythroleukemia cells.
For personal use only.

ca-induced lung fibrosis in mice. Am. J. Respir. Cell Mol. Biol. 31:78–85. VIII. Role of the immune system in the inhibition of visceral metastases.
BéruBé, K. A., Quinlan, T. R., Fung, H., Magae, J., Vacek, P., Taatjes, D. J. and Int. J. Cancer. 46:468–474.
Mossman, B. T. 1996. Apoptosis is observed in mesothelial cells after expo- Gruber, U. F. 1990. Asbestos-related benign disease and cancer: symptoms and
sure to crocidolite asbestos. Am. J. Respir. Cell Mol. Biol. 15:141–147. treatment. Anticancer. Drugs. 1:187–197.
Borges, V. M., Falcão, H., Leite-Júnior, J. H., Alvim, L., Teixeira, G. P., Guo, Z. Q., Otsuki, T., Shimizu, T., Tachiyama, S., Sakaguchi, H., Isozaki, Y.,
Russo, M., Nóbrega, A. F., Lopes, M. F., Rocco, P. M., Davidson, W. F., Tomokuni, T., Hyodoh, F., Kusaka, M. and Ueki, A. 2001. Reduced expres-
Linden, R., Yagita, H., Zin, W. A. and DosReis, G. A. 2001. Fas ligand trig- sion of survivin gene in PBMC from silicosis patients. Kawasaki. Med. J.
gers pulmonary silicosis. J. Exp. Med. 194:155–164. 27:75–81.
Borges, V. M., Lopes, M. F., Falcão, H., Leite-Júnior, J. H., Rocco, P. R., Hamilton, R. F. Jr, Thakur, S. A. and Holian, A. 2008. Silica binding and toxicity
Davidson, W. F., Linden, R., Zin, W. A. and DosReis, G. A. 2002. Apoptosis in alveolar macrophages. Free Radic. Biol. Med. 44:1246–1258.
underlies immunopathogenic mechanisms in acute silicosis. Am. J. Respir. Herberman, R. B. 1985. Multiple functions of natural killer cells, includ-
Cell Mol. Biol. 27:78–84. ing immunoregulation as well as resistance to tumor growth. Concepts.
Brandt-Rauf, P. W., Luo, J. C., Carney, W. P., Smith, S., De Vivo, I., Milling, C., Immunopathol. 1:96–132.
Hemminki, K., Koskinen, H., Vainio, H. and Neugut, A. I. 1994. Detection Hess, E. V. 2002. Environmental chemicals and autoimmune disease: cause and
of increased amounts of the extracellular domain of the c-erbB-2 oncopro- effect. Toxicology 181-182:65–70.
tein in serum during pulmonary carcinogenesis in humans. Int. J. Cancer. Hori, S. and Sakaguchi, S. 2004. Foxp3: a critical regulator of the development
56:383–386. and function of regulatory T cells. Microbes. Infect. 6:745–751.
Broaddus, V. C., Yang, L., Scavo, L. M., Ernst, J. D. and Boylan, A. M. 1996. Hyodoh, F., Takata-Tomokuni, A., Miura, Y., Sakaguchi, H., Hatayama, T.,
Asbestos induces apoptosis of human and rabbit pleural mesothelial cells Hatada, S., Katsuyama, H., Matsuo, Y. and Otsuki, T. 2005. Inhibitory
via reactive oxygen species. J. Clin. Invest. 98:2050–2059. effects of anti-oxidants on apoptosis of a human polyclonal T-cell line,
Brown, N. J., Hutcheson, J., Bickel, E., Scatizzi, J. C., Albee, L. D., Haines, G. K. MT-2, induced by an asbestos, chrysotile-A. Scand. J. Immunol. 61:
3rd, Eslick, J., Bradley, K., Taricone, E. and Perlman, H. 2004. Fas death 442–448.
receptor signaling represses monocyte numbers and macrophage activa- IARC (International Agency for Research on Cancer). 1977. IARC Monographs
tion in vivo. J. Immunol. 173:7584–7593. on the Evaluation of the Carcinogenic Risk of Chemicals to Man: Asbestos.
Brown, T. 2009. Silica exposure, smoking, silicosis and lung cancer–complex IARC Monographs on the Evaluation of Carcinogenic Risk of Chemicals to
interactions. Occup. Med. (Lond). 59:89–95. Man. 14:1–106.
Buder-Hoffmann, S., Palmer, C., Vacek, P., Taatjes, D. and Mossman, B. 2001. IARC (International Agency for Research on Cancer). 1997. Working Group on
Different accumulation of activated extracellular signal-regulated kinases the Evaluation of Carcinogenic Risks to Humans: Silica, Some Silicates,
(ERK ½) and role in cell-cycle alterations by epidermal growth factor, Coal Dust and Para-Aramid Fibrils. IARC Monographs on the Evaluation
hydrogen peroxide, or asbestos in pulmonary epithelial cells. Am. J. Respir. of Carcinogenic Risks to Humans. 68:1–475.
Cell Mol. Biol. 24:405–413. ILO (International Labor Office). 2002. Guidelines for the Use of the ILO
Buhtoiarov, I. N., Lum, H. D., Berke, G., Sondel, P. M. and Rakhmilevich, A. L. International Classification of Radiographs of Pneumoconioses, Revised
2006. Synergistic activation of macrophages via CD40 and TLR9 results in Edition 2000 (Occupational Safety and Health Series, No. 22). International
T-cell-independent anti-tumor effects. J. Immunol. 176:309–318. Labour Office: Geneva.
Castranova, V. 2004. Signaling pathways controlling the production of inflam- Jagirdar J., Begin R., Dufresne A., Goswami S., Lee T. C. and Rom W. N. 1996.
matory mediators in response to crystalline silica exposure: role of reac- Transforming growth factor-beta (TGF-β) in silicosis. Am. J. Resp. Crit. Care
tive oxygen/nitrogen species. Free Radic. Biol. Med. 37:916–925. Med. 154:1076–1081.
Chambers, B. J., Wilson, J. L., Salcedo, M., Markovic, K., Bejarano, M. T. and Ji, W. J., Yang, L., Ding, J. S., Wang, Z. L., Liu, C. and He, H. Z. 2003a.
Ljunggren, H. G. 1998. Triggering of natural killer cell mediated cyto- Immunohistochemical method for the detecting expression of transform-
toxicity by co-stimulatory molecules. Curr. Top. Microbiol. Immunol. ing growth factor-β1 in lung tissues of silica-treated mice. Chinese J. Ind.
230:53–61. Hyg. Occup. Dis. 21:182–184.
Chen, J. J., Sun, Y. and Nabel, G. J. 1998. Regulation of the proinflammatory Ji, W. J., Yang, L., Wang, Z. L., Ding, J. S., Liu, C. and He, H. Z. 2003b. RT-PCR
effects of Fas ligand (CD95L). Science 282:1714–1717. method for detecting the expression of transforming growth factor-β1
Silica/asbestos and immune dysregulation   277

gene in lung tissues of silica-treated mice. Chinese J. Ind. Hyg. Occup. Dis. Otsuki, T., Maeda, M., Miura, Y., Hayashi, H., Murakami, S., Kumagai, N. and
21:185–187. Nishimura, Y. 2009. Immunological effects of asbestos. In: Asbestos: Risks,
Keller, R. 1976. Promotion of tumor growth in vivo by anti-macrophage agents. Environment and Impact (Soto, A. and Salazar, G., Eds.), New York: Nova
JNCI 57:1355–1361. Science Publishers, Inc., pp. 185–193.
Kimber, I. 1985. Natural killer cells. Med. Lab. Sci. 42:60–77. Otsuki, T., Maeda, M., Murakami, S., Hayashi, H., Miura, Y., Kusaka, M.,
Lacasse, Y., Martin, S., Gagné, D. and Lakhal, L. 2009. Dose-response meta- Nakano, T., Fukuoka, K., Kishimoto, T., Hyodoh, F., Ueki, A. and Nishimura,
analysis of silica and lung cancer. Cancer. Causes. Control. 20:925–933. Y. 2007. Immunological effects of silica and asbestos. Cell. Mol. Immunol.
Lee, K. P. 1985. Lung response to particulates with emphasis on asbestos and 4:261–268.
other fibrous dusts. Crit. Rev. Toxicol. 14:33–86. Otsuki, T., Miura, Y., Nishimura, Y., Hyodoh, F., Takata, A., Kusaka, M.,
Lum, H. D., Buhtoiarov, I. N., Schmidt, B. E., Berke, G., Paulnock, D. M., Katsuyama, H., Tomita, M., Ueki, A. and Kishimoto, T. 2006. Alterations
Sondel, P. M. and Rakhmilevich, A. L. 2006. In vivo CD40 ligation can of Fas and Fas-related molecules in patients with silicosis. Exp. Biol. Med.
induce T-cell-independent anti-tumor effects that involve macrophages. (Maywood). 231:522–533.
J. Leukocyte. Biol. 79:1181–1192. Otsuki, T., Sakaguchi, H., Tomokuni, A., Aikoh, T., Matsuki, T., Isozaki, Y.,
Maeda, M., Miura, Y., Nishimura, Y., Murakami, S., Hayashi, H., Kumagai, Hyodoh, F., Kawakami, Y., Kusaka, M., Kita, S. and Ueki, A. 2000a.
N., Hatayama, T., Katoh, M., Miyahara, N., Yamamoto, S., Fukuoka, K., Detection of alternatively spliced variant messages of Fas gene and
Kishimoto, T., Nakano, T. and Otsuki, T. 2008. Immunological changes in mutational screening of Fas and Fas ligand coding regions in peripheral
mesothelioma patients and their experimental detection. Clin. Med.: Circ, blood mononuclear cells derived from silicosis patients. Immunol. Lett.
Resp. Pulm. Med. 2:11–17. 72:137–143.
Miura, Y., Nishimura, Y., Katsuyama, H., Maeda, M., Hayashi, H., Dong, M., Otsuki, T., Takata, A., Hyodoh, F. and Ueki, A. 2003. Review of regulation for
Journal of Immunotoxicology Downloaded from informahealthcare.com by Gazi Univ. on 01/09/15

Hyodoh, F., Tomita, M., Matsuo, Y., Uesaka, A., Kuribayashi, K., the Fas-mediated apoptotic pathway in silicosis patients. Kawasaki. Med.
Nakano, T., Kishimoto, T. and Otsuki, T. 2006. Involvement of IL-10 and J. 29:33–43.
Bcl-2 in resistance against an asbestos-induced apoptosis of T cells. Otsuki, T., Tomokuni, A., Sakaguchi, H., Aikoh, T., Matsuki, T., Isozaki, Y.,
Apoptosis 11:1825–1835. Hyodoh, F., Ueki, H., Kusaka, M., Kita, S. and Ueki, A. 2000b. Over-
Miura, Y., Nishimura, Y., Maeda, M., Murakami, S., Hayashi, H., Fukuoka, K., expression of the decoy receptor 3 (DcR3) gene in peripheral blood mono-
Kishimoto, T., Nakano, T. and Otsuki, T. 2008. Immunological alterations nuclear cells (PBMC) derived from silicosis patients. Clin. Exp. Immunol.
found in mesothelioma patients and supporting experimental evidence. 119:323–327.
Environ. Health. Prev. Med. 13:55–59. Otsuki, T., Tomokuni, A., Sakaguchi, H., Hyodoh, F., Kusaka, M. and Ueki, A.
Murakami, S., Nishimura, Y., Maeda, M., Kumagai, N., Hayashi, H., Chen, Y., 2000c. Reduced expression of the inhibitory genes for Fas-mediated apop-
Kusaka, M., Kishimoto, T. and Otsuki, T. 2009. Cytokine alteration and tosis in silicosis patients. J. Occup. Health. 42:163–168.
speculated immunological pathophysiology in silicosis and asbestos- Partanen, R., Hemminki, K., Koskinen, H., Luo, J. C., Carney, W. P. and Brandt-
related diseases. Environ. Health. Prev. Med. 14:216–222. Rauf, P. W. 1994. The detection of increased amounts of the extracellular
Nagata, S. and Suda, T. 1995. Fas and Fas ligand: lpr and gld mutations. domain of the epidermal growth factor receptor in serum during carcino-
Immunol. Today. 16:39–43. genesis in asbestosis patients. J. Occup. Med. 36:1324–1328.
For personal use only.

Nicholson, W. J. 1984. Research issues in occupational and environmental can- Pfau, J. C., Sentissi, J. J., Weller, G. and Putnam, E. A. 2005. Assessment of
cer. Arch. Environ. Health. 39:190–202. autoimmune responses associated with asbestos exposure in Libby,
Nicholson, W. J. 2001. The carcinogenicity of chrysotile asbestos–a review. Ind. Montana, USA. Environ. Health. Perspect. 113:25–30.
Health. 39:57–64. Rao, T. D. and Frey, A. B. 1998. Administration of silica sensitizes lipopolysac-
Niklinski, J., Niklinska, W., Chyczewska, E., Laudanski, J., Naumnik, W., charide responsiveness of murine macrophages but inhibits T- and B-cell
Chyczewski, L. and Pluygers, E. 2004. The epidemiology of asbestos-re- priming by inhibition of antigen presenting function. Immunol. Invest.
lated diseases. Lung. Cancer. 45 Suppl 1:S7–S15. 27:181–199.
Nishimura, Y., Maeda, M., Kumagai, N., Hayashi, H., Miura, Y. and Otsuki, T. Raulet, D. H. and Guerra, N. 2009. Oncogenic stress sensed by the immune sys-
2009a. Decrease in phosphorylation of ERK following decreased expres- tem: role of natural killer cell receptors. Nat. Rev. Immunol. 9:568–580.
sion of NK cell-activating receptors in human NK cell line exposed to Rimal, B., Greenberg, A. K. and Rom, W. N. 2005. Basic pathogenetic mech-
asbestos. Int. J. Immunopathol. Pharmacol. 22:879–888. anisms in silicosis: current understanding. Curr. Opin. Pulm. Med.
Nishimura, Y., Maeda, M., Kumagai, N., Murakami, S., Hayashi, H., 11:169–173.
Kishimoto, T., Fukuoka, K., Nakano, T. and Otsuki, T. 2009c. Suppressive Rolston, R. and Oury, T. D. 2004. Other neoplasia. In: Asbestos-Associated
effect of asbestos-exposure on cytotoxicity of human NK cells, and the Diseases, 2nd Edition (Roggli, V. L., Oury, T. D. and Aporn, T. A., Eds.),
possibility of NKp46 as a marker to monitor immune status in people New York: Springer Science and Business Media Inc., pp. 217–230.
exposed to asbestos. In: Proceedings of the 2nd China-Japan Joint Asbestos Rom, W. N., Travis, W. D. and Brody, A. R. 1991. Cellular and molecular basis of
Symposium, Industrial Hygiene and Occupational Diseases Section (Chinese the asbestos-related diseases. Am. Rev. Respir. Dis. 143:408–422.
Preventive Medical Association and Japan Asbestos Mesothelioma Study Sakaguchi, S. 2005. Naturally-arising Foxp3-expressing CD25+CD4+ regulatory
Group, Eds.), pp. 129–135. T-cells in immunological tolerance to self and non-self. Nat. Immunol.
Nishimura, Y., Miura, Y., Maeda, M., Hayashi, H., Dong, M., Katsuyama, H., 6:345–352.
Tomita, M., Hyodoh, F., Kusaka, M., Uesaka, A., Kuribayashi, K., Fukuoka, K., Sakaguchi, S., Ono, M., Setoguchi, R., Yagi, H., Hori, S., Fehervari, Z.,
Nakano, T., Kashimoto, T. and Osuki, T. 2006. Expression of the T cell Shimizu, J., Takahashi, T. and Nomura, T. 2006. Foxp3+ CD25+ CD4+ natural
receptor Vbeta repertoire in a human T cell resistant to asbestos-induced regulatory T cells in dominant self-tolerance and autoimmune disease.
apoptosis and peripheral blood T cells from patients with silica and asbes- Immunol. Rev. 212:8–27.
tos-related diseases. Int. J. Immunopathol. Pharmacol. 19:795–805. Sandstrom, P. A. and Chow, D. A. 1987. Regulation of tumor development:
Nishimura, Y., Miura, Y., Maeda, M., Kumagai, N., Murakami, S., Hayashi, H., Biphasic effects of silica and of lipopolysaccharide on natural resistance.
Fukuoka, K., Nakano, T. and Otsuki, T. 2009c. Impairment in cytotoxic- Int. J. Cancer 40:122–130.
ity and expression of NK cell- activating receptors on human NK cells Shanklin, D. R. and Smalley, D. L. 1998. The immunopathology of siliconosis.
following exposure to asbestos fibers. Int. J. Immunopathol. Pharmacol. History, clinical presentation, and relation to silicosis and the chemistry
22:579–590. of silicon and silicone. Immunol. Res. 18:125–173.
Nishimura, Y., Nishiike-Wada, T., Wada, Y., Miura, Y., Otsuki, T. and Singh, N. and Davis, G. S. 2002. Review: occupational and environmental lung
Iguchi, H. 2007. Long-lasting production of TGF-β1 by alveolar mac- disease. Curr. Opin. Pulm. Med. 8:117–125.
rophages exposed to low doses of asbestos without apoptosis. Int. J. Snyder, J. T., Alexander-Miller, M. A., Berzofskyl, J. A. and Belyakov, I. M. 2003.
Immunopathol. Pharmacol. 20:661–667. Molecular mechanisms and biological significance of CTL avidity. Curr.
Noonan, C. W., Pfau, J. C., Larson, T. C. and Spence, M. R. 2006. Nested case- HIV Res. 1:287–294.
control study of autoimmune disease in an asbestos-exposed population. Sobel, E. S., Kakkanaiah, V. N., Cohen, P. L. and Eisenberg, R. A. 1993. Correction
Environ. Health. Perspect. 114:1243–1247. of gld autoimmunity by co-infusion of normal bone marrow suggests that
Oh, S., Rankin, A. L. and Caton, A. J. 2010. CD4+CD25+ regulatory T cells in gld is a mutation of the Fas ligand gene. Int. Immunol. 5:1275–1278.
autoimmune arthritis. Immunol. Rev. 233:97–111. Steenland, K. and Goldsmith, D. F. 1995. Silica exposure and autoimmune dis-
Ollikainen, T., Puhakka, A., Kahlos, K., Linnainmaa, K. and Kinnula, V. L. 2000. eases. Am. J. Ind. Med. 28:603–608.
Modulation of cell and DNA damage by poly(ADP)ribose polymerase in Su, W., Ito, T., Oyama, T., Kitagawa, T., Yamori, T., Fujiwara, H. and Matsuda, H.
lung cells exposed to H2O2 or asbestos fibres. Mutat. Res. 470:77–84. 2001. The direct effect of IL-12 on tumor cells: IL-12 acts directly on
278   M. Maeda et al.

tumor cells to activate NF-κB and enhance IFNγ-mediated STAT1 phos- Ueki, H., Kohda, M., Nobutoh, T., Yamaguchi, M., Omori, K., Miyashita, Y.,
phorylation. Biochem. Biophys. Res. Commun. 280:503–512. Hashimoto, T., Komai, A., Tomokuni, A. and Ueki, A. 2001. Anti-
Takahashi, T., Tanaka, M., Brannan, C. I., Jenkins, N. A., Copeland, N. G., desmoglein autoantibodies in silicosis patients with no bullous diseases.
Suda, T. and Nagata, S. 1994. Generalized lymphoproliferative disease in Dermatology 202:16–21.
mice, caused by a point mutation in the Fas ligand. Cell 76:969–976. van Baarle, D., Kostense, S., van Oers, M. H., Hamann, D. and Miedema, F. 2002.
Takata-Tomokuni, A., Ueki, A., Shiwa, M., Isozaki, Y., Hatayama, T., Failing immune control as a result of impaired CD8+ T-cell maturation:
Katsuyama, H., Hyodoh, F., Fujimoto, W., Ueki, H., Kusaka, M., Arikuni, H. CD27 might provide a clue. Trends. Immunol. 23:586–591.
and Otsuki, T. 2005. Detection, epitope-mapping and function of anti-Fas Watanabe-Fukunaga, R., Brannan, C. I., Copeland, N. G., Jenkins, N. A.
autoantibody in patients with silicosis. Immunology 116:21–29. and Nagata, S. 1992. Lymphoproliferation disorder in mice explained
Tellesson, W. G. 1961. Rheumatoid pneumoconiosis (Caplan’s syndrome) in an by defects in Fas antigen that mediates apoptosis. Nature 356:
asbestos worker. Thorax 16:372–377. 314–317.
Thakur, S. A., Hamilton, R. F. Jr, and Holian, A. 2008. Role of scavenger receptor Watson, M. L., Rao, J. K., Gilkeson, G. S., Ruiz, P., Eicher, E. M., Pisetsky, D. S.,
a family in lung inflammation from exposure to environmental particles. Matsuzawa, A., Rochelle, J. M. and Seldin, M. F. 1992. Genetic analysis of
J. Immunotoxicol. 5:151–157. MRL-lpr mice: relationship of the Fas apoptosis gene to disease manifesta-
Tomokuni, A., Aikoh, T., Matsuki, T., Isozaki, Y., Otsuki, T., Kita, S., Ueki, H., tions and renal disease-modifying loci. J. Exp. Med. 176:1645–1656.
Kusaka, M., Kishimoto, T. and Ueki, A. 1997. Elevated soluble Fas/APO-1 Weiss, J. M., Subleski, J. J., Wigginton, J. M. and Wiltrout, R. H. 2007.
(CD95) levels in silicosis patients without clinical symptoms of autoim- Immunotherapy of cancer by IL-12-based cytokine combinations. Expert.
mune diseases or malignant tumours. Clin. Exp. Immunol. 110:303–309. Opin. Biol. Ther. 7:1705–1721.
Tomokuni, A., Otsuki, T., Isozaki, Y., Kita, S., Ueki, H., Kusaka, M., Wells, S. M., Noonan, C., Wells, K. M., Holian, A. and Wibbenmeyer, L. A. 2009.
Journal of Immunotoxicology Downloaded from informahealthcare.com by Gazi Univ. on 01/09/15

Kishimoto, T. and Ueki, A. 1999. Serum levels of soluble Fas ligand in Effects of toxic gases: methamphetamine inhalation. J. Burn. Care. Res.
patients with silicosis. Clin. Exp. Immunol. 118:441–444. 30:152–154.
Toyokuni, S. 1996. Iron-induced carcinogenesis: the role of redox regulation. Williams, A. O., Flanders, K. C. and Saffiotti, U. 1993. Immunohistochemical
Free Radic. Biol. Med. 20:553–566. localization of transforming growth factor-β 1 in rats with experimental
Toyokuni, S. 2009. Mechanisms of asbestos-induced carcinogenesis. Nagoya. silicosis, alveolar Type II hyperplasia, and lung cancer. Am. J. Pathol.
J. Med. Sci. 71:1–10. 142:1831–1840.
Trice, J. M. and Pinals, R. S. 1985. Dimethyl sulfoxide: a review of its use in the Wortinger, M. A., Foley, J. W., Larocque, P., Witcher, D. R., Lahn, M.,
rheumatic disorders. Semin. Arthritis. Rheum. 15:45–60. Jakubowski, J. A., Glasebrook, A. and Song, H. Y. 2003. Fas ligand-induced
Uber, C. L. and McReynolds, R. A. 1982. Immunotoxicology of silica. Crit. Rev. murine pulmonary inflammation is reduced by a stable decoy receptor 3
Toxicol. 10:303–319. analogue. Immunology 110:225–233.
Ueki, A., Isozaki, Y., Tomokuni, A., Hatayama, T., Ueki, H., Kusaka, M., Wu, J., Zhou, T., He, J. and Mountz, J. D. 1993. Autoimmune disease in mice
Shiwa, M., Arikuni, H., Takeshita, T. and Morimoto, K. 2002. Intramolecular due to integration of an endogenous retrovirus in an apoptosis gene.
epitope spreading among anti-caspase-8 autoantibodies in patients with J. Exp. Med. 178:461–468.
silicosis, systemic sclerosis and systemic lupus erythematosus, as well as Wu, P., Miura, Y., Hyodoh, F., Nishimura, Y., Hatayama, T., Hatada, S.,
For personal use only.

in healthy individuals. Clin. Exp. Immunol. 129:556–561. Sakaguchi, H., Kusaka, M., Katsuyama, H., Tomita, M. and Otsuki, T. 2006.
Ueki, A., Isozaki, Y., Tomokuni, A., Ueki, H., Kusaka, M., Tanaka, S., Otsuki, T., Reduced function of CD4+25+ regulatory T-cell fraction in silicosis patients.
Sakaguchi, H. and Hyodoh, F. 2001. Different distribution of HLA class II Int. J. Immunopathol. Pharmacol. 19:357–368.
alleles in anti-topoisomerase I autoantibody responders between silico- Zhang, L. X., Zeng, J. B., Du, H. K., Zhang, S. W. and Wang, S. X. 2006. Expression
sis and systemic sclerosis patients, with a common distinct amino acid of FasL and apoptosis in pulmonary tissue of rats exposed to silica at dif-
sequence in the HLA-DQB1 domain. Immunobiology 204:458–465. ferent times. Chinese J. Ind. Hyg. Occup. Dis. 24:641–644.

You might also like