Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Environmental Immunology: Lessons

Learned from Exposure to a Select Panel of


Immunotoxicants
This information is current as Joanna M. Kreitinger, Celine A. Beamer and David M.
of October 21, 2019. Shepherd
J Immunol 2016; 196:3217-3225; ;
doi: 10.4049/jimmunol.1502149
http://www.jimmunol.org/content/196/8/3217

Downloaded from http://www.jimmunol.org/ by guest on October 21, 2019


References This article cites 115 articles, 23 of which you can access for free at:
http://www.jimmunol.org/content/196/8/3217.full#ref-list-1

Why The JI? Submit online.


• Rapid Reviews! 30 days* from submission to initial decision
• No Triage! Every submission reviewed by practicing scientists
• Fast Publication! 4 weeks from acceptance to publication
*average

Subscription Information about subscribing to The Journal of Immunology is online at:


http://jimmunol.org/subscription
Permissions Submit copyright permission requests at:
http://www.aai.org/About/Publications/JI/copyright.html
Email Alerts Receive free email-alerts when new articles cite this article. Sign up at:
http://jimmunol.org/alerts

The Journal of Immunology is published twice each month by


The American Association of Immunologists, Inc.,
1451 Rockville Pike, Suite 650, Rockville, MD 20852
Copyright © 2016 by The American Association of
Immunologists, Inc. All rights reserved.
Print ISSN: 0022-1767 Online ISSN: 1550-6606.
The Journal of
Brief Reviews Immunology
Environmental Immunology: Lessons Learned from
Exposure to a Select Panel of Immunotoxicants
Joanna M. Kreitinger,* Celine A. Beamer,† and David M. Shepherd†
Exposure to environmental contaminants can produce contaminants impact immune responsiveness not only helps
profound effects on the immune system. Many classes in the effective regulation of pollutants and improving public
of xenobiotics can significantly suppress or enhance im- health, it provides novel insights into basic functions of the
mune responsiveness depending on the levels (i.e., dose) immune system. Therefore, the intent of this article is not to
and context (i.e., timing, route) of exposure. Although present an overview of the immunotoxic effects of environ-
defining the effects that toxicants can have on the im- mental toxicants as it pertains to human health (because this
mune system is a valuable component to improving information is much too extensive for a Brief Review) but
public health, environmental immunology has greatly instead to emphasize recent contributions that environmental

Downloaded from http://www.jimmunol.org/ by guest on October 21, 2019


enhanced our understanding of how the immune system immunology has made to advance the field of immunology.
functions and has provided innovative avenues to ex- Consequently, this Brief Review focuses exclusively on three
plore new immunotherapies. This Brief Review focuses cutting-edge areas of research that are providing critical
on three examples of how immunotoxicology has insights into basic immune function, the effects of environ-
mental compounds on immune function, and the develop-
benefitted the field of immunology, presenting informa-
ment of potentially novel immunotherapies.
tion on the aryl hydrocarbon receptor signaling path-
way, the immunomodulatory effects of nanomaterials, Discovering the aryl hydrocarbon receptor. Although multiple sci-
entific advances coalesced into the discovery of the aryl hydro-
and the impact of xenobiotic exposure on the develop-
carbon receptor (AhR), a common theme existed among them:
ing immune system. Collectively, contributions from
the necessity to understand mechanisms of xenobiotic toxicity.
immunotoxicology have significantly enhanced public
Specifically, the observation in the 1970s that polycyclic aro-
health and spurred seminal advances in both basic matic hydrocarbons appeared to induce their own metabolism
and applied immunology. The Journal of Immunol- drove the ultimate discovery of a receptor that could bind these
ogy, 2016, 196: 3217–3225. compounds (1–3). A prodigious contribution to AhR research
came with the discovery that 2,3,7,8 tetrachlorodibenzo-p-dioxin
(TCDD), a halogenated aromatic hydrocarbon and ubiquitous

I
t is widely accepted that human health is a product of
both genetics and the environment, a premise that also environmental contaminant, could bind AhR and, in fact, could
holds true for the immune system. Although our genetic do so in mammalian hepatocytes with 30,000 times the affinity
make-up is essentially set at birth, the environment that we of other environmental compounds, including polycyclic aromatic
experience is constantly changing, presenting novel challenges hydrocarbons (4). With this discovery, AhR research and its
in the development, regulation, and function of immunity. impacts across multiple physiological systems, including
Assessing the consequences of exposure to environmental immunology, were just beginning.
compounds within the immune system is not a straightforward In the years following identification of AhR, characterizing
process. Toxicants can enter the human body through four its expression in nonhepatic tissues became a key focus. In
primary routes: inhalation (respiratory tract), ingestion (gas- 1984, thymic expression of AhR in Sprague-Dawley rats was
trointestinal tract), dermal contact (skin), and parenteral determined to occur at high levels for twice as long (42 d versus
(circulation/muscles) as a result of intentional or accidental 21 d) after parturition compared with nonimmune tissues,
exposure (Fig. 1). Therefore, the study of how xenobiotics such as liver and lungs (5). These observations led to further
influence the immune system, also known as immunotox- investigation of AhR activity in immune organs and the
icology or environmental immunology, is a critical compo- pivotal discovery that AhR activation had adverse effects on
nent of immunology. Understanding how environmental thymic function. In 1985, PCBs acting through AhR were

*Cellular, Molecular, and Microbial Biology Graduate Program, Division of Biological Address correspondence and reprint requests to Dr. David M. Shepherd, Department of
Sciences, University of Montana, Missoula, MT 59812; and †Department of Biomedical Biomedical and Pharmaceutical Sciences, University of Montana, SB 284, 32 Campus
and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812 Drive, Missoula, MT 59812. E-mail address: david.shepherd@umontana.edu
ORCIDs: 0000-0003-0341-7429 (J.M.K.); 0000-0003-4995-6145 (C.A.B.). Abbreviations used in this article: AhR, aryl hydrocarbon receptor; AhR/NP, AhR
agonist–loaded NP; AuNP, gold NP; BPA, bisphenol A; DC, dendritic cell; EAE,
Received for publication October 2, 2015. Accepted for publication February 16, 2016.
experimental autoimmune encephalomyelitis; HSC, hematopoietic stem cell; ILC, in-
This work was supported by National Institute of Environmental Health Sciences/ nate lymphoid cell; ITE, 2-(19H-indole-39-carbonyl)-thiazole-4-carboxylic acid methyl
National Institute of General Medical Sciences Grant ES013784 and National Institute ester; MWCNT, multi-walled carbon nanotube; NM, nanomaterial; NP, nanoparticle;
of Environmental Health Sciences Grant ES025386 (both to D.M.S.) and by National PEG, polyethylene glycol; TCDD, 2,3,7,8 tetrachlorodibenzo-p-dioxin.
Institute of General Medical Sciences Grant GM103546 (to C.A.B.).
Copyright Ó 2016 by The American Association of Immunologists, Inc. 0022-1767/16/$30.00

www.jimmunol.org/cgi/doi/10.4049/jimmunol.1502149
3218 BRIEF REVIEWS: ENVIRONMENTAL TOXICANTS AND IMMUNITY

FIGURE 1. Exposure to and effects of environmental


toxicants. Toxicants may access the human body through
four points of entry (skin, blood, respiratory, and digestive
tracts), as shown in blue, and affect numerous organs,
including the immune system. Toxicants interact with the
immune system as a result of deliberate or unintentional
exposure, causing either immune activation or immune
suppression. Depending on the disease context, this may
be either beneficial or detrimental because undesirable
immune activation or immune suppression may result in
adverse health effects.

Downloaded from http://www.jimmunol.org/ by guest on October 21, 2019


found to cause severe thymic atrophy and humoral immune play a significant role in AhR-mediated immune suppression.
suppression (6). The succession of rapid discoveries through- AhR activation in DCs increases the expression of regulatory
out the 1980s broadened the expanse of AhR-mediated sup- genes, including IDO-1 and IDO-2 (Ido1 and Ido2), reti-
pression within the immune system to include thymic atrophy, naldehyde dehydrogenase 1 (aldh1a1), and TGF-b3. Conse-
reduced cell-mediated immunity, diminished humoral Ab re- quently, AhR-activated regulatory DCs can be potent inducers
sponses, and an overall reduction in the ability to respond to of Foxp3+ regulatory T cells (16–20). In contrast, AhR func-
and resist infectious diseases (5–9). tions as a regulator of innate/inflammatory responses in mac-
With a potential link made between AhR and observed sup- rophages, as illustrated by the negative regulation of LPS-
pression of the immune system, immunologists entered into induced inflammation and subsequent enhanced sensitivity
the field, which to this point had predominantly been pio- to lethal shock (21). Also, AhR activation in monocytes/
neered by toxicologists and biochemists. The interweaving of macrophages results in dysregulated cytokine and chemokine
discoveries from these disciplines expanded our understanding production, a consistent effect observed in murine and human
of the AhR signaling pathway and its role in innate and cells (22, 23). Lastly, the differentiation and/or maturation of
adaptive immune responses, as recently reviewed by Stockinger monocytes/macrophages and DCs is affected by AhR, an effect
et al. (10). Because several recent reviews focused on AhR acti- that contributes to immune dysregulation (24–26).
vation in T cells, our focus is on what we consider three key Human NK cells, which express AhR in stage 3 immature
areas of ongoing investigation: the role of AhR in the innate NK cells and “NK-22” cells, play an important role in innate
immune system, the crusade to identify endogenous ligands, immune responses to mucosal infections that may be altered
and the development of novel AhR-mediated immunotherapies.
Innate immune cells vary widely in their expression of AhR
(Fig. 2), ranging from high, constitutive expression to low,
conservatively inducible expression. It should be noted that
most reports describing AhR expression in immune cells rely
solely on mRNA levels and not protein expression, representing
a potential important gap in information as recently highlighted
by Esser and Rannug (11). The outcome of AhR activation on
the promotion or regulation of immune responses appears to
vary by cell type and maturational state. Interestingly, both
inflammatory and regulatory genes have an abundance of AhR-
responsive elements (dioxin response elements or xenobiotic
response elements) within their promoter/enhancer regions (12,
13), and it was shown that cross-talk between AhR and NF-kB
can significantly impact multiple proinflammatory signals (14).
Neutrophils, as well as most granulocytes, do not express AhR at
significant levels and appear to be predominantly influenced by
AhR activation through indirect mechanisms (15).
Professional APCs (e.g., macrophages and dendritic cells FIGURE 2. AhR expression in immune cells. AhR expression (x-axis) and
[DCs]) constitutively express AhR at relatively high levels and outcomes of AhR activation (y-axis) by immune cell type.
The Journal of Immunology 3219

by environmental AhR ligands (27, 28). Indeed, AhR regulates there are many factors that must be fully considered when
cytolytic activity in murine NK cells, as represented by reduced intentionally targeting AhR to achieve specific immune regu-
bacterial clearance, and tumor cell recognition capacity in lation. It is well documented that the effects of AhR activa-
AhR-null mice. These studies also suggest that NK cell func- tion during an immune response vary based on the target cell
tion is potentiated following AhR activation (29). Conversely, (10, 11, 32, 37). AhR activation can increase inflammatory
other reports demonstrate that AhR activation can suppress responses or, alternatively, promote immune regulation or tol-
NK cell function, suggesting that additional environmental erance (38). Recently, nanoparticles (NPs) containing ITE,
or maturational components may also contribute to AhR- an AhR agonist, were used successfully to treat experimental
mediated effects in NK cells (14). Likewise, innate lymphoid autoimmune encephalomyelitis (EAE), a mouse model of
cells (ILCs), which play an important role in mucosal im- multiple sclerosis. In these studies, gold NPs (AuNPs) were
munity, were shown to have compromised bacterial clearance used for parenteral delivery of ITE and an antigenic self-
and reduced IL-22 expression when they lack AhR (30). peptide, myelin oligodendrocyte glycoprotein 35–55 (39).
Although investigations into the role of AhR in ILCs are Within 6 h of administration, splenic DCs, which inter-
relatively recent, existing studies indicate AhR activation in nalized ITE-loaded NPs, significantly upregulated AhR-
ILC subsets effectively regulates transcriptional activation and, responsive genes, including aldh1a1 and cytochrome P450
consequently, the clearance of mucosal infections, immune 1a1, the prototypical AhR target gene. Ultimately, a marked
tissue development, and regulation of chronic inflammatory reduction in EAE disease scores was observed following ad-
diseases, such as inflammatory bowel disease (31). Collectively, ministration of ITE-loaded and ITE/myelin oligodendrocyte

Downloaded from http://www.jimmunol.org/ by guest on October 21, 2019


AhR plays a prominent role in the development and function glycoprotein 35–55-loaded NPs, an outcome that was at-
of the innate immune system, and future studies are warranted tributed to a significant induction of disease-specific regula-
to further elucidate the health implications of this relationship. tory T cells (39).
Because the most well-studied AhR ligands are ubiquitous Although parenteral administration of AhR agonist–loaded
environmental pollutants, studies aimed at defining the im- NPs (AhR/NPs) showed significant amelioration of EAE in
pacts of AhR activation within the immune system often have mice, AuNP uptake was not isolated to DCs in these animals;
had dual roles: to identify the immunomodulatory effects of it was observed in several immune and nonimmune cells (39).
AhR signaling in immune cells and to assess the breadth of Because unguided NPs can often yield off-target adverse ef-
immunotoxicity following exposure to large classes of xeno- fects, we hypothesize that using delivery systems that directly
biotics. Early discoveries by toxicologists and biochemists target specific immune cells, such as DCs, will increase the
provided several decades of highly insightful AhR research efficacy of AhR/NP therapy, lower the concentration of AhR/
within the immune system. However, now that AhR research NPs necessary to reach therapeutic levels, and reduce or
has gained considerable traction among immunologists, future eliminate potential adverse responses that may occur from
studies on this ligand-activated transcription factor are expected nonspecific uptake of AhR/NPs by bystander cells. Ulti-
to continue to bolster our understanding of immune function mately, intentional modulation of AhR holds great promise
and regulation. To this end, the recent discovery of putative for the development of novel immunotherapeutics to treat
endogenous AhR ligands yielded several compounds that can patients suffering from multiple sclerosis and potentially other
bind and activate AhR with relatively high affinity (32). For devastating immune-mediated diseases.
example, 2-(19H-indole-39-carbonyl)-thiazole-4-carboxylic acid Nanomaterials and the immune system. Nanotechnology repre-
methyl ester (ITE) from porcine lung tissue (33) and the sents the merging of science, engineering, and technology
tryptophan derivatives 6-formylindolo[3,2-b]carbazole and at the next great industrial revolution: control of matter at
L-kynurenine are “natural” AhR-activating compounds that can the nanoscale. At this level, matter exhibits unique physical,
significantly impact the generation of immunity (18, 34–37). chemical, and biological properties, which differ from the
However, although these chemicals activate AhR and can induce canonical properties of bulk materials and single atoms or
similar transcriptional responses to TCDD, such as the induc- molecules. These unique properties, although enabling novel
tion of signature cytochrome P450s (CYP1A1, CYP1A2, applications and technological advances, also give rise to safety
CYP1B1), they are highly susceptible to metabolism by these concerns that cannot be ignored. Several of those same char-
xenobiotic-metabolizing enzymes, thereby limiting their effec- acteristics that make nanomaterials (NMs) so promising from
tive half-life and associated toxicities (37). At present, at least a technological standpoint, also make their interactions with
one of these compounds (ITE) is being evaluated for potential biological systems difficult to anticipate and critically impor-
therapeutic use in humans (discussed below). Notably, we have tant to investigate methodically.
a limited understanding of the specific biomolecular interactions Over the last decade, researchers from industry, academia,
that occur between ligands and AhR as the result of the lack of a and federal agencies have been collaborating to establish spe-
defined crystal structure for this receptor. Therefore, it is diffi- cific assays and NMs criteria that will assess the impact of
cult to accurately predict what the clinical outcomes will be if/ NMs on immune function. Currently, no comprehensive
when AhR-binding compounds are eventually tested in hu- guidelines exist. Because NMs represent such physically and
mans, who are generally considered to express a 10-fold lower- chemically diverse materials, classical immunotoxicological
affinity AhR compared with (most) rodents. This relationship methods cannot always be applied, and novel approaches may
remains to be confirmed in significantly larger human cohorts, be required to overcome challenges inherent to NMs research.
thereby representing a prominent gap in our understanding of For example, many NMs absorb in the UV–visible range and
AhR biology. may even catalyze enzyme reactions or quench fluorescent
With AhR now recognized as a potent immune regulator, it dyes commonly used in end-point and kinetic assays. Simi-
has become an attractive target for immunotherapy. However, larly, although exposure to a particular NM may result in no
3220 BRIEF REVIEWS: ENVIRONMENTAL TOXICANTS AND IMMUNITY

change in immune function when delivered via the oral and accumulation, which may disrupt autophagy (57, 58). In
dermal route, it may induce sensitization after intradermal turn, disruption of autophagy may enhance NLRP3 inflam-
injection. Lastly, risk to human health is not entirely linked to masome accumulation, resulting in exaggerated IL-1b pro-
NM production volume and probability of exposure, but duction and lung fibrosis (59). Interestingly, variation in
rather to reactivity and potency of impact on specific organs. the dimensions and surface properties of NMs affects the
Consequently, NMs produced in lower quantities that have autophagic responses of cells (60, 61). Similarly, variation
potent and selective effects on the immune system (e.g., in MWCNT surface structure activates alternate autophagy
nickel, gold, and cobalt NM) may exhibit greater immuno- signaling pathways (mTOR dependent to mTOR indepen-
toxicity. Thus, we must assemble a comprehensive under- dent) (62). Agents that can selectively induce autophagic cell
standing of the relationship between diverse NMs and specific death in tumor cells are promising tools to treat or supple-
components of the immune response resulting from different ment current cancer therapies. Thus, modulation of auto-
testing scenarios. phagy by NMs may be a potential therapeutic strategy. Taken
As a result of NM size and unique properties, the immune together, these studies provide novel insights on how NMs,
system efficiently recognizes engineered NMs as foreign bod- such as MWCNTs, contribute to inflammasome activation,
ies, resulting in multilevel responses that can range from acute autophagic cell death, and induction of the stress response, as
to chronic, and from immunostimulatory to immunosup- well as the role of NM surface chemistry in the modulation of
pressive (40). Although NMs-induced immune activation these signaling processes for therapeutic benefit. These find-
may increase the incidence of allergic reactions, inflammatory ings are particularly relevant given that dysfunction or dys-

Downloaded from http://www.jimmunol.org/ by guest on October 21, 2019


responses, or autoimmunity, NMs-induced suppression may regulation of inflammasome activation and autophagy is
reduce maturation and proliferation of immune cells, result- associated with several immune-mediated diseases.
ing in increased susceptibility to infectious diseases or tumor Given the rapid growth of nanotechnology and the myriad
growth. Although a desirable interaction between NMs and potential physicochemical properties, NMs will influence and
the immune system may lead to beneficial outcomes, such as advance emerging medical technologies in both therapeutics
vaccines or therapeutics for inflammatory and autoimmune and diagnostic applications. Scientists are engineering NM-
disorders, an undesirable interaction may result in adverse based approaches to specifically target and/or circumvent
outcomes, such as hypersensitivity reactions and inflamma- the immune system. The resulting treatments may be more
tion, or lowered response to infection and cancerous cells. precise, may diagnose or treat disease earlier, and may exhibit
Moreover, inadvertent recognition of NMs as foreign by im- fewer adverse side effects. Nanotechnology platforms are being
mune cells may result in host toxicity and/or reduced thera- investigated as vaccine carriers, adjuvants, and drug-delivery
peutic efficacy of conventional pharmaceuticals. systems to target inflammation and inflammatory-associated
Carbon nanotubes are a family of NMs made up entirely of disorders. Some formulations are already in clinical trials,
carbon. Within this family, multi-walled carbon nanotubes whereas many others are in various phases of preclinical de-
(MWCNTs) are of special interest to the industry because of velopment. NMs-based delivery systems offer the following
their broad applications in electronics, cosmetics, cleaning potential advantages: site-specific delivery of drugs, peptides,
materials, coatings, food packaging, and medicines. Concerns and genes; improved in vitro and in vivo stability; and reduced
over MWCNT-induced toxicity have emerged, in part due side effects. Although in recent years our understanding of
to their high production volume, as well as their structural NMs interaction with components of the immune system has
similarity to asbestos. improved, many questions remain.
Numerous studies have since scrutinized the effects of It is now well accepted that NM size, surface charge,
MWCNT length and diameter, purity, aspect ratio or rigidity, hydrophobicity/hydrophilicity, and the steric effects of par-
and functionalization in relation to in vitro and in vivo toxicity, ticle coating can dictate NM compatibility with the immune
inflammation, and fibrosis, particularly in the lungs (41–43). system. For example, NMs can be designed by attaching to
Numerous studies clearly demonstrate that MWCNTs induce polyethylene glycol (PEG) of other types of polymers to create
lung injury through acute and chronic inflammation, granu- a hydrophobic environment, thus shielding them from im-
loma formation, and substantial interstitial lung fibrosis, as mune recognition (63). Although these polymers may shield
well as exacerbate asthma-like conditions in mice (44–46). NMs from most immune surveillance, some data suggest
This section surveys recent efforts to define the effects of the formation of PEG-specific Abs (64, 65) and accelerated
MWCNTs on the innate immune system. Once NMs deposit clearance of PEG liposomes from the blood, thus changing
in the lungs, alveolar macrophages are responsible for their the pharmacokinetic profiles (66–68). Therefore, generation
uptake and processing through phagocytosis, macro- of NM-specific Abs may affect the efficacy and safety of NM-
pinocytosis, and clathrin/caveolin-dependent and -indepen- based therapeutics. In contrast, NMs can be designed to elicit
dent endocytosis (47–49). A number of studies similarly an immune response by either direct activation of APCs or
linked MWCNT-induced oxidant stress, phagolysosomal delivering Ag to specific cellular compartments (69). One of
permeabilization, cathepsin B release, NLRP3 inflammasome the most heavily researched areas in nanomedicine is NP-
assembly, and caspase-1 activation with secretion of important based drug delivery (70, 71). Although this research is pro-
regulatory cytokines (e.g., IL-1b and IL-18) (50–55). Acti- ducing effective reagents and making poorly soluble drugs
vation of the NLRP3 inflammasome by MWCNTs is con- useful through encapsulation, controlled release, and target-
sistent with reports showing that other inhaled particles ing, the NPs themselves are usually not therapeutic.
(e.g., silica and asbestos) cause lung injury through In this final section, we briefly review the effects of AuNPs
NLRP3 inflammasome activation (56). Upon internalization, on immune function. As a result of their fairly simple prep-
MWCNTs induce lysosomal dysfunction and autophagosome aration and functionalization with drugs or other ligands,
The Journal of Immunology 3221

unique optical properties, and tuneability with regard to a improved stability, favorable biodistribution profiles, slower
desired size or shape (72–74), AuNP-based immunotherapies drug-release kinetics, lower immunotoxicity, and targeting to
have been a promising carrier for immune therapies in can- specific immune cell populations. However, it is essential to
cer Ag and immune adjuvant delivery (75–77), as well as address the potential adverse health risks that NMs pose in
in photothermal ablation and light-triggered drug delivery occupational, medicinal, consumer, or environmental expo-
(78–80). However, because AuNPs can accumulate in the sure settings because of their unpredictable chemical and bi-
immune system and exhibit many toxic side effects, they ological nature. Immune reactions are a key concern for
should be used cautiously as a delivery system. potential adverse effects of NMs. Although certain predictions
As with other NMs, the size, shape, and electrical charge can be made regarding the immunotoxicity of NMs, the
alter the recognition and uptake of AuNPs in both phagocytic unique nature of these structures make it difficult, if not
and nonphagocytic cells. Interestingly, in nonphagocytic cells, impossible, to predict how some NMs will interact with in-
positively charged AuNPs appear to be taken up to a much tracellular structures, such as DNA, cell membranes, and
greater extent than negatively charged ones, whereas in cytoskeletal proteins. NMs exposure has not yet been linked to
phagocytic cells, particle charge has little effect on uptake. human disease because of the relatively recent emergence of
Furthermore, in nonphagocytic cells, AuNPs were taken up via nanotechnology and the lack of epidemiologic data. Thus,
clathrin-mediated endocytosis and localized to secondary ly- future research into NMs is highly relevant from a public
sosomes, whereas in phagocytic cells, AuNPs were taken up health perspective, as well as for the novel insights that it will
through phagocytosis and located in phagosomes (81). Later yield on the basic functions of the immune system.

Downloaded from http://www.jimmunol.org/ by guest on October 21, 2019


studies showed that phagocytosis of 30- versus 150-nm- Effects of xenobiotics on the developing immune system. To develop
diameter AuNPs occurred via different mechanisms (e.g., novel and safe therapies that provide efficient protection
clathrin-mediated pinocytosis and scavenger receptor–medi- against infectious diseases or immune-mediated diseases in
ated phagocytosis, respectively) (82). In vitro AuNPs induce children or those that develop during childhood, it is
proinflammatory cytokine expression in macrophages in a necessary to understand immune system ontogeny. Because
size-dependent manner (83). In contrast, other laboratories immunological development is dependent on both genetic
showed that AuNPs can inhibit IL-1b–mediated inflamma- and environmental influences, investigations into both
tory responses and TLR9 responses, also in a size-dependent fields advance our understanding of immune function and
manner (84, 85). Although these studies also suggested that provide the foundation for the development of novel
particles , 5 nm had the greatest impact on immune re- immunotherapeutics. Notably, the generation of research
sponse, others determined that more hydrophobic ∼2-nm tools, such as genetically altered animals and targeted gene
AuNPs exhibited the greatest expression of inflammatory modulation, has greatly enhanced our understanding of the
cytokines by mouse splenocytes (86). Oligonucleotide- impact that genetic abnormalities have on the developing
conjugated 13-nm AuNPs increased the expression of in- immune system. However, over the last two decades, the
flammatory and defense response genes as categorized by gene effects of pollutants have been systematically evaluated,
ontology analysis in human PBMCs, yet similar responses further adding to our understanding of how the environment
were not observed in the 293 T cell line (87), suggesting that affects the developing immune system. These studies revolve
assessing the immunotoxicity of AuNPs on immortalized cell around two central tenets: the developing immune system
lines, as opposed to primary cells, may yield differing results. may be more sensitive to environmental chemical exposures
Therefore, the immune response to AuNPs may be tissue and than the adult immune system, and the environment
context dependent, as well as particle size and shape depen- significantly impacts the developmental origins of health
dent. Finally, because AuNPs inevitably accumulate in high and disease. Disruption of the developing immune system
concentrations in the liver and spleen for extended periods of by early-life environmental insults may not only adversely
time (88, 89), it is important to better understand how affect the health of the exposed offspring later in life,
AuNPs interact with the immune system before develop- it potentially extends to additional generations through
ing them as a therapeutic delivery platform. It appears that epigenetic changes. Therefore, uncovering the effects of
therapeutic AuNPs may be similar to chemotherapeutics’ environmental insults on the developing immune system is
double-edged sword because they can be therapeutic, yet they expected to benefit public health and provide novel insights
can also be toxic. Additionally, the differing results regard- into immune system ontogeny. The increased risk for injury
ing the inflammatory or anti-inflammatory effects of AuNPs to the developing immune system has been seen across
merit further consideration. Although the distribution and several categories of drugs and chemicals, as well as heavy
immune-modulatory activity of AuNPs in vivo may cause metals and mold toxins. Moreover, prenatal and early
inflammatory or anti-inflammatory side effects, these prop- postnatal exposures are more likely to produce a broader
erties might also be manipulated for therapeutic applications, array of immune parameters affected, and an increased risk
such as vaccine and immune adjuvant delivery. These studies for adverse immune outcomes persisting into later life (90).
highlight the need to design NMs that do not adversely affect These differential immune effects can take many forms, which
the biological systems where they will be used and demon- are briefly described below for a select few toxicants.
strate that it is crucial to simultaneously ascertain the thera- Perhaps one of the more comprehensive datasets for de-
peutic and toxic properties of any metal-containing NP. velopmental immunotoxicity exists for TCDD and TCDD-
Nanotechnology has generated, and will continue to gen- like chemicals. To this end, developmental exposure to
erate, a wide spectrum of novel NMs that will revolutionize AhR-activating chemicals, such as TCDD, perturbs immune
many fields. Existing studies demonstrated that nanotech- cell development, resulting in attenuated hematopoietic stem
nology offers advantages over traditional medicines, such as cell (HSC) differentiation and long-term self-renewal (91).
3222 BRIEF REVIEWS: ENVIRONMENTAL TOXICANTS AND IMMUNITY

These effects can contribute to increased incidence of auto- innate immunity in adult offspring mice but did not impair
immunity and decreased responsiveness to infectious patho- the adaptive immune response to influenza A virus infection
gens, such as influenza A (92, 93). Furthermore, activation of (112). Perinatal exposure to low doses of BPA rendered the
AhR by TCDD induces epigenetic changes in lymphocytes, neonatal immune system more susceptible to food intolerance
especially T cells, causing DNA hypermethylation and (113), and prenatal, but not postnatal, BPA exposure can
ultimately reducing CD8+ T cell antiviral immunity (94). enhance the postnatal development of experimental allergic
Conversely, early life exposure to TCDD was recently dem- asthma (114). Because BPA was shown to induce epigenetic
onstrated to enhance the CD4+ T cell response to viral in- regulation (i.e., DNA methylation, microRNA expression,
fection in the lung; however, this response ultimately resulted and histone acetylation) in reproductive tissues, it will be
in greater bronchopulmonary inflammation and, con- interesting to determine whether similar effects exist in the
sequently, reduced antiviral immunity (95, 96). Additionally, developing immune system that may consequently affect the
the AhR signaling pathway plays a requisite role in the de- generation of inflammatory or immune-mediated disease
velopment and function of HSCs and progenitor cells; con- (115). At the least, it is expected that further research on the
sequently, AhR-null mice possess abnormalities in these potential immunotoxic effects of BPA and other endocrine
cellular populations (97–100). Moreover, recent reports de- disruptors will help us to better understand how the immune
scribed a critical usefulness for AhR antagonists (along with system develops and is shaped by environmental insults.
Notch ligand agonists) in the regulation and generation of Disruption of the ordered process of immunological
HSCs from cord blood, providing significant advances for development by xenobiotics can generate several potential

Downloaded from http://www.jimmunol.org/ by guest on October 21, 2019


allograft transplantation (97, 101). Taken together, AhR is outcomes, including immune deficiency, deviation, or dys-
now recognized as a critical regulator of key cells in the de- regulation. Although it is hypothesized that these effects
veloping immune system; consequently, a greater under- manifest themselves during the course of development or
standing of its effects will benefit the development of novel maturation of an organism, it is also possible that immuno-
immunotherapies and result in improved human health. toxicity can be transgenerational, although additional studies
Exposure to other chemicals in the environment, such as are necessary to firmly establish this potential outcome. If
pesticides, heavy metals, and endocrine disruptors, also impact confirmed, these long-term effects could have profound im-
the developing immune system. Several pesticides, including plications on the immune system and its ability to respond
organochlorines (i.e., chlordane, DDT/DDE), organophos- effectively to infectious pathogens and tumor cells or to reg-
phates (i.e., diazinon), and carbamates (i.e., carbofuran), were ulate appropriately self-directed responses that underlie au-
reported to induce developmental immunotoxicity in rodents toimmunity. However, it is expected that, with an increased
(102). Additionally, prenatal exposure of humans to DDE is understanding of immunological ontogeny, opportunities will
associated with reduced levels of IgG and recurrent respiratory develop for the generation of novel therapies to mitigate the
infections in infants (103, 104). These results are intriguing effects of environmental stressors.
because of reports that DDE can also enhance allergic re-
sponses in infants via induction of Th2 cytokines (105) but Conclusions
not via modulation of IgE, IL-33, or TSLP (106). Exposure of Environmental immunology/immunotoxicology is a relatively
the developing immune system to heavy metals, such as lead, new and vital area of investigation in the field of immunology.
a known developmental toxicant, can adversely affect the Understanding how anthropogenic toxicants and natural
generation of innate immune cells, such as macrophages and toxins affect the development, maintenance, and function of
DCs (107). Specifically, dysregulated macrophage and DC the immune system is critical for achieving optimal human
effector functions, including reduced phagocytosis, lysosomal health. Surprisingly perhaps, the U.S. Environmental Pro-
activation, and excessive production of reactive oxygen species tection Agency is the only chemical regulatory agency to have
and other inflammatory mediators (i.e., TNF-a, PGE2), specific immunotoxicity testing requirements, and these are
contribute to Th2-biased immune responses in lead-exposed primarily for assessing pesticides (116). In contrast, the phar-
animals (108–110), ultimately contributing to imbalanced maceutical industry uses a weight-of-evidence approach that
and inappropriate immune responses that can be persistent is only exercised for immunological evaluation if standard
and increase the risk for inflammatory disorders and disease toxicological assays show cause for concern. Regardless, nu-
later in life (107). Thus, these studies have advanced our merous examples exist demonstrating the potential adverse
understanding of ontogeny of the innate immune system and effects that exposure to environmental chemicals can and do
support the hypothesis that early life exposure to these envi- have on the immune system. Although xenobiotic exposure
ronmental stressors can program the immune system for can come from environmental, occupational, or recreational
subsequent dysfunction. Lastly, endocrine disruptors, such as sources, this list of potential threats has expanded recently to
bisphenol A (BPA; found in plastics, food sources, and many include symbiotic microbial exposure. Thus, it is extremely
other consumer products), can alter various physiological valuable for public health to fully understand the impact that
systems by interfering with the activity or production of environmental chemicals have on the immune system, the
hormones. As expected, exposure to these types of chemicals, mechanisms underlying these effects, and how they might be
which occurs at the highest levels in fetuses, infants, and mitigated.
children, is thought to adversely affect human development. It Overall, environmental immunology has provided highly
is not known whether these environmental chemicals ad- tangible benefits in the study of the immune system. To better
versely affect the developing immune system, although, very understand how AhR regulates immune development and
recently, this has been an area of intense investigation (111). function, the use of chemicals, such as TCDD, has been in-
For example, developmental exposure to BPA modulated voked as environmental stressors, as well as biological probes.
The Journal of Immunology 3223

These studies have resulted in a greater understanding of the 15. Wheeler, T. A., K. S. Lawrence, D. O. Porter, W. Keeling, and B. G. Mullinix, Jr.
2013. The relationship between environmental variables and response of cotton to
critical role of the AhR in HSCs, progenitor cells, and innate and nematicides. J. Nematol. 45: 8–16.
adaptive immune cells. In the study of NMs, immunotoxicol- 16. Stockinger, B., K. Hirota, J. Duarte, and M. Veldhoen. 2011. External influences
on the immune system via activation of the aryl hydrocarbon receptor. Semin.
ogists have been on the cutting-edge of discovery, identifying Immunol. 23: 99–105.
powerful biomedical uses and establishing necessary safety pa- 17. Benson, J. M., and D. M. Shepherd. 2011. Dietary ligands of the aryl hydrocarbon
rameters for these xenobiotics. Experiments using NMs also receptor induce anti-inflammatory and immunoregulatory effects on murine
dendritic cells. Toxicol. Sci. 124: 327–338.
yielded novel, fundamental insights into inflammatory processes, 18. Bankoti, J., B. Rase, T. Simones, and D. M. Shepherd. 2010. Functional and
such as the generation and regulation of the inflammasome, as phenotypic effects of AhR activation in inflammatory dendritic cells. Toxicol. Appl.
Pharmacol. 246: 18–28.
well as intracellular organelles (i.e., lysosomes, phagosomes) and 19. Marshall, N. B., W. R. Vorachek, L. B. Steppan, D. V. Mourich, and
their function (i.e., autophagy) in immune cells. Moreover, recent N. I. Kerkvliet. 2008. Functional characterization and gene expression analysis of
CD4+ CD25+ regulatory T cells generated in mice treated with 2,3,7,8-tetra-
studies involving nanomedicines have ushered in a new era for chlorodibenzo-p-dioxin. J. Immunol. 181: 2382–2391.
discovery of biomedical probes and drug-delivery systems that 20. Quintana, F. J., G. Murugaiyan, M. F. Farez, M. Mitsdoerffer, A. M. Tukpah,
have the potential to revolutionize medicine in the coming years. E. J. Burns, and H. L. Weiner. 2010. An endogenous aryl hydrocarbon receptor
ligand acts on dendritic cells and T cells to suppress experimental autoimmune
Lastly, experiments aimed at defining the effects of xenobiotics encephalomyelitis. Proc. Natl. Acad. Sci. USA 107: 20768–20773.
on the developing immune system are beginning to expand our 21. Kimura, A., T. Naka, T. Nakahama, I. Chinen, K. Masuda, K. Nohara, Y. Fujii-
Kuriyama, and T. Kishimoto. 2009. Aryl hydrocarbon receptor in combination
understanding of immunological ontogeny. Previously unknown with Stat1 regulates LPS-induced inflammatory responses. J. Exp. Med. 206:
or unappreciated molecular regulators of immune cell/tissue 2027–2035.
22. N’Diaye, M., E. Le Ferrec, D. Lagadic-Gossmann, S. Corre, D. Gilot,
development, such as AhR signaling modulators (both agonists V. Lecureur, P. Monteiro, C. Rauch, M. D. Galibert, and O. Fardel. 2006. Aryl

Downloaded from http://www.jimmunol.org/ by guest on October 21, 2019


and antagonists), environmental pollutants (i.e., TCDD, PCBs) hydrocarbon receptor- and calcium-dependent induction of the chemokine CCL1
that induce epigenetic modifications, and endocrine disruption by the environmental contaminant benzo[a]pyrene. J. Biol. Chem. 281: 19906–
19915.
by xenoestrogens (i.e., BPA), are expected to enhance public 23. Lahoti, T. S., J. A. Boyer, A. Kusnadi, G. E. Muku, I. A. Murray, and
health, as well as spur development of novel immunotherapeu- G. H. Perdew. 2015. Aryl Hydrocarbon Receptor Activation Synergistically In-
duces Lipopolysaccharide-Mediated Expression of Proinflammatory Chemokine
tics designed to improve childhood health. In this manner, en- (c-c motif) Ligand 20. Toxicol. Sci. 148: 229–240.
vironmental immunology provides innovation at the intersection 24. Jux, B., S. Kadow, and C. Esser. 2009. Langerhans cell maturation and contact
of immunology and the environment in which we live. hypersensitivity are impaired in aryl hydrocarbon receptor-null mice. J. Immunol.
182: 6709–6717.
25. Platzer, B., S. Richter, D. Kneidinger, D. Waltenberger, M. Woisetschläger, and
H. Strobl. 2009. Aryl hydrocarbon receptor activation inhibits in vitro differen-
Disclosures tiation of human monocytes and Langerhans dendritic cells. J. Immunol. 183: 66–
The authors have no financial conflicts of interest. 74.
26. van Grevenynghe, J., S. Rion, E. Le Ferrec, M. Le Vee, L. Amiot, R. Fauchet, and
O. Fardel. 2003. Polycyclic aromatic hydrocarbons inhibit differentiation of hu-
References man monocytes into macrophages. J. Immunol. 170: 2374–2381.
27. Cella, M., A. Fuchs, W. Vermi, F. Facchetti, K. Otero, J. K. Lennerz,
1. Nebert, D. W., and L. L. Bausserman. 1970. Genetic differences in the extent of
J. M. Doherty, J. C. Mills, and M. Colonna. 2009. A human natural killer cell
aryl hydrocarbon hydroxylase induction in mouse fetal cell cultures. J. Biol. Chem.
subset provides an innate source of IL-22 for mucosal immunity. Nature 457: 722–
245: 6373–6382.
725.
2. Okey, A. B. 2007. An aryl hydrocarbon receptor odyssey to the shores of toxi-
28. Hughes, T., B. Becknell, A. G. Freud, S. McClory, E. Briercheck, J. Yu, C. Mao,
cology: the Deichmann Lecture, International Congress of Toxicology-XI. Toxicol.
C. Giovenzana, G. Nuovo, L. Wei, et al. 2010. Interleukin-1beta selectively ex-
Sci. 98: 5–38.
pands and sustains interleukin-22+ immature human natural killer cells in sec-
3. Poland, A., E. Glover, and A. S. Kende. 1976. Stereospecific, high affinity binding
ondary lymphoid tissue. Immunity 32: 803–814.
of 2,3,7,8-tetrachlorodibenzo-p-dioxin by hepatic cytosol. Evidence that the
29. Shin, J. H., L. Zhang, O. Murillo-Sauca, J. Kim, H. E. Kohrt, J. D. Bui, and
binding species is receptor for induction of aryl hydrocarbon hydroxylase. J. Biol.
J. B. Sunwoo. 2013. Modulation of natural killer cell antitumor activity by the aryl
Chem. 251: 4936–4946. hydrocarbon receptor. Proc. Natl. Acad. Sci. USA 110: 12391–12396.
4. Kouri, R. R., H. Ratrie, III, S. A. Atlas, A. Niwa, and D. W. Nebert. 1974. Aryl 30. Qiu, J., J. J. Heller, X. Guo, Z. M. Chen, K. Fish, Y. X. Fu, and L. Zhou. 2012.
hydrocarbon hydroxylase induction in human lymphocyte cultures by 2,3,7,8- The aryl hydrocarbon receptor regulates gut immunity through modulation of
tetrachlorodibenzo-p-dioxin. Life Sci. 15: 1585–1595. innate lymphoid cells. Immunity 36: 92–104.
5. Gasiewicz, T. A., W. C. Ness, and G. Rucci. 1984. Ontogeny of the cytosolic 31. Serafini, N., C. A. Vosshenrich, and J. P. Di Santo. 2015. Transcriptional regu-
receptor for 2,3,7,8-tetrachlorodibenzo-p-dioxin in rat liver, lung, and thymus. lation of innate lymphoid cell fate. Nat. Rev. Immunol. 15: 415–428.
Biochem. Biophys. Res. Commun. 118: 183–190. 32. Cella, M., and M. Colonna. 2015. Aryl hydrocarbon receptor: Linking environ-
6. Silkworth, J. B., and L. Antrim. 1985. Relationship between Ah receptor-mediated ment to immunity. Semin. Immunol. 27: 310–314.
polychlorinated biphenyl (PCB)-induced humoral immunosuppression and thy- 33. Song, J., M. Clagett-Dame, R. E. Peterson, M. E. Hahn, W. M. Westler,
mic atrophy. J. Pharmacol. Exp. Ther. 235: 606–611. R. R. Sicinski, and H. F. DeLuca. 2002. A ligand for the aryl hydrocarbon receptor
7. Silkworth, J. B., L. Antrim, and L. S. Kaminsky. 1984. Correlations between isolated from lung. Proc. Natl. Acad. Sci. USA 99: 14694–14699.
polychlorinated biphenyl immunotoxicity, the aromatic hydrocarbon locus, and 34. Mezrich, J. D., J. H. Fechner, X. Zhang, B. P. Johnson, W. J. Burlingham, and
liver microsomal enzyme induction in C57BL/6 and DBA/2 mice. Toxicol. Appl. C. A. Bradfield. 2010. An interaction between kynurenine and the aryl hydro-
Pharmacol. 75: 156–165. carbon receptor can generate regulatory T cells. J. Immunol. 185: 3190–3198.
8. Silkworth, J. B., L. A. Antrim, and G. Sack. 1986. Ah receptor mediated sup- 35. Rannug, U., A. Rannug, U. Sjöberg, H. Li, R. Westerholm, and J. Bergman. 1995.
pression of the antibody response in mice is primarily dependent on the Ah Structure elucidation of two tryptophan-derived, high affinity Ah receptor ligands.
phenotype of lymphoid tissue. Toxicol. Appl. Pharmacol. 86: 380–390. Chem. Biol. 2: 841–845.
9. Nikolaidis, E., B. Brunström, and L. Dencker. 1988. Effects of the TCDD con- 36. Duarte, J. H., P. Di Meglio, K. Hirota, H. Ahlfors, and B. Stockinger. 2013.
geners 3,39,4,49-tetrachlorobiphenyl and 3,39,4,49-tetrachloroazoxybenzene on Differential influences of the aryl hydrocarbon receptor on Th17 mediated re-
lymphoid development in the bursa of Fabricius of the chick embryo. Toxicol. sponses in vitro and in vivo. PLoS One 8: e79819.
Appl. Pharmacol. 92: 315–323. 37. Wheeler, J. L., K. C. Martin, E. Resseguie, and B. P. Lawrence. 2014. Differential
10. Stockinger, B., P. Di Meglio, M. Gialitakis, and J. H. Duarte. 2014. The aryl consequences of two distinct AhR ligands on innate and adaptive immune re-
hydrocarbon receptor: multitasking in the immune system. Annu. Rev. Immunol. sponses to influenza A virus. Toxicol. Sci. 137: 324–334.
32: 403–432. 38. Lee, Y. H., C. H. Lin, P. C. Hsu, Y. Y. Sun, Y. J. Huang, J. H. Zhuo, C. Y. Wang,
11. Esser, C., and A. Rannug. 2015. The aryl hydrocarbon receptor in barrier organ Y. L. Gan, C. C. Hung, C. Y. Kuan, and F. S. Shie. 2015. Aryl hydrocarbon
physiology, immunology, and toxicology. Pharmacol. Rev. 67: 259–279. receptor mediates both proinflammatory and anti-inflammatory effects in
12. Frericks, M., M. Meissner, and C. Esser. 2007. Microarray analysis of the AHR lipopolysaccharide-activated microglia. Glia 63: 1138–1154.
system: tissue-specific flexibility in signal and target genes. Toxicol. Appl. Phar- 39. Yeste, A., M. Nadeau, E. J. Burns, H. L. Weiner, and F. J. Quintana. 2012.
macol. 220: 320–332. Nanoparticle-mediated codelivery of myelin antigen and a tolerogenic small
13. Kerkvliet, N. I. 2009. AHR-mediated immunomodulation: the role of altered gene molecule suppresses experimental autoimmune encephalomyelitis. Proc. Natl.
transcription. Biochem. Pharmacol. 77: 746–760. Acad. Sci. USA 109: 11270–11275.
14. Hanieh, H. 2014. Toward understanding the role of aryl hydrocarbon receptor in 40. Hayes, M. D., V. Ovcinnikovs, A. G. Smith, I. Kimber, and R. J. Dearman. 2014.
the immune system: current progress and future trends. BioMed Res. Int. 2014: The aryl hydrocarbon receptor: differential contribution to T helper 17 and T
520763. cytotoxic 17 cell development. PLoS One 9: e106955.
3224 BRIEF REVIEWS: ENVIRONMENTAL TOXICANTS AND IMMUNITY

41. Borm, P. J., D. Robbins, S. Haubold, T. Kuhlbusch, H. Fissan, K. Donaldson, 67. Ishida, T., M. Ichihara, X. Wang, K. Yamamoto, J. Kimura, E. Majima, and
R. Schins, V. Stone, W. Kreyling, J. Lademann, et al. 2006. The potential risks of H. Kiwada. 2006. Injection of PEGylated liposomes in rats elicits PEG-specific
nanomaterials: a review carried out for ECETOC. Part. Fibre Toxicol. 3: 11. IgM, which is responsible for rapid elimination of a second dose of PEGylated
42. Kendall, M., and S. Holgate. 2012. Health impact and toxicological effects of liposomes. J. Control. Release 112: 15–25.
nanomaterials in the lung. Respirology 17: 743–758. 68. Ishida, T., and H. Kiwada. 2008. Accelerated blood clearance (ABC) phenomenon
43. Mercer, R. R., J. F. Scabilloni, A. F. Hubbs, L. A. Battelli, W. McKinney, upon repeated injection of PEGylated liposomes. Int. J. Pharm. 354: 56–62.
S. Friend, M. G. Wolfarth, M. Andrew, V. Castranova, and D. W. Porter. 2013. 69. Kalkanidis, M., G. A. Pietersz, S. D. Xiang, P. L. Mottram, B. Crimeen-Irwin,
Distribution and fibrotic response following inhalation exposure to multi-walled K. Ardipradja, and M. Plebanski. 2006. Methods for nano-particle based vaccine
carbon nanotubes. Part. Fibre Toxicol. 10: 33. formulation and evaluation of their immunogenicity. Methods 40: 20–29.
44. Landsiedel, R., U. G. Sauer, L. Ma-Hock, J. Schnekenburger, and M. Wiemann. 70. Yamada, M., M. Foote, and T. W. Prow. 2015. Therapeutic gold, silver, and
2014. Pulmonary toxicity of nanomaterials: a critical comparison of published in platinum nanoparticles. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 7: 428–
vitro assays and in vivo inhalation or instillation studies. Nanomedicine (Lond.) 9: 445.
2557–2585. 71. Moon, J. J., B. Huang, and D. J. Irvine. 2012. Engineering nano- and micro-
45. Beamer, C. A., T. A. Girtsman, B. P. Seaver, K. J. Finsaas, C. T. Migliaccio, particles to tune immunity. Adv. Mater. 24: 3724–3746.
V. K. Perry, J. B. Rottman, D. E. Smith, and A. Holian. 2013. IL-33 mediates 72. Kodiha, M., Y. M. Wang, E. Hutter, D. Maysinger, and U. Stochaj. 2015. Off to
multi-walled carbon nanotube (MWCNT)-induced airway hyper-reactivity via the the organelles - killing cancer cells with targeted gold nanoparticles. Theranostics 5:
mobilization of innate helper cells in the lung. Nanotoxicology 7: 1070–1081. 357–370.
46. Girtsman, T. A., C. A. Beamer, N. Wu, M. Buford, and A. Holian. 2014. IL-1R 73. Khlebtsov, N., and L. Dykman. 2011. Biodistribution and toxicity of engineered
signalling is critical for regulation of multi-walled carbon nanotubes-induced acute gold nanoparticles: a review of in vitro and in vivo studies. Chem. Soc. Rev. 40:
lung inflammation in C57Bl/6 mice. Nanotoxicology 8: 17–27. 1647–1671.
47. Maruyama, K., H. Haniu, N. Saito, Y. Matsuda, T. Tsukahara, S. Kobayashi, 74. Almeida, J. P., A. L. Chen, A. Foster, and R. Drezek. 2011. In vivo biodistribution
M. Tanaka, K. Aoki, S. Takanashi, M. Okamoto, and H. Kato. 2015. Endocytosis of nanoparticles. Nanomedicine (Lond.) 6: 815–835.
of Multiwalled Carbon Nanotubes in Bronchial Epithelial and Mesothelial Cells. 75. Lee, I. H., H. K. Kwon, S. An, D. Kim, S. Kim, M. K. Yu, J. H. Lee, T. S. Lee,
BioMed Res. Int. 2015: 793186. S. H. Im, and S. Jon. 2012. Imageable antigen-presenting gold nanoparticle vac-
48. Shannahan, J. H., W. Bai, and J. M. Brown. 2015. Implications of scavenger cines for effective cancer immunotherapy in vivo. Angew. Chem. Int. Ed. Engl. 51:
receptors in the safe development of nanotherapeutics. Receptors Clin. Investig. 2: 8800–8805.

Downloaded from http://www.jimmunol.org/ by guest on October 21, 2019


e811. 76. Lin, A. Y., J. P. Almeida, A. Bear, N. Liu, L. Luo, A. E. Foster, and R. A. Drezek.
49. Hirano, S., S. Kanno, and A. Furuyama. 2008. Multi-walled carbon nanotubes 2013. Gold nanoparticle delivery of modified CpG stimulates macrophages and
injure the plasma membrane of macrophages. Toxicol. Appl. Pharmacol. 232: 244– inhibits tumor growth for enhanced immunotherapy. PLoS One 8: e63550.
251. 77. Massich, M. D., D. A. Giljohann, A. L. Schmucker, P. C. Patel, and C. A. Mirkin.
50. Hamilton, R. F., N. Wu, D. Porter, M. Buford, M. Wolfarth, and A. Holian. 2010. Cellular response of polyvalent oligonucleotide-gold nanoparticle conju-
2009. Particle length-dependent titanium dioxide nanomaterials toxicity and gates. ACS Nano 4: 5641–5646.
bioactivity. Part. Fibre Toxicol. 6: 35. 78. Arvizo, R., R. Bhattacharya, and P. Mukherjee. 2010. Gold nanoparticles: op-
51. Nel, A., T. Xia, L. Mädler, and N. Li. 2006. Toxic potential of materials at the portunities and challenges in nanomedicine. Expert Opin. Drug Deliv. 7: 753–763.
nanolevel. Science 311: 622–627. 79. Hirsch, L. R., R. J. Stafford, J. A. Bankson, S. R. Sershen, B. Rivera, R. E. Price,
52. Murphy, F. A., A. Schinwald, C. A. Poland, and K. Donaldson. 2012. The J. D. Hazle, N. J. Halas, and J. L. West. 2003. Nanoshell-mediated near-infrared
mechanism of pleural inflammation by long carbon nanotubes: interaction of long thermal therapy of tumors under magnetic resonance guidance. Proc. Natl. Acad.
fibres with macrophages stimulates them to amplify pro-inflammatory responses in Sci. USA 100: 13549–13554.
mesothelial cells. Part. Fibre Toxicol. 9: 8. 80. Huang, X., P. K. Jain, I. H. El-Sayed, and M. A. El-Sayed. 2007. Gold nano-
53. Porter, D. W., A. F. Hubbs, B. T. Chen, W. McKinney, R. R. Mercer, particles: interesting optical properties and recent applications in cancer diagnostics
M. G. Wolfarth, L. Battelli, N. Wu, K. Sriram, S. Leonard, et al. 2013. Acute and therapy. Nanomedicine (Lond.) 2: 681–693.
pulmonary dose-responses to inhaled multi-walled carbon nanotubes. Nano- 81. Liu, X., N. Huang, H. Li, Q. Jin, and J. Ji. 2013. Surface and size effects on cell
toxicology 7: 1179–1194. interaction of gold nanoparticles with both phagocytic and nonphagocytic cells.
54. Palomäki, J., E. Välimäki, J. Sund, M. Vippola, P. A. Clausen, K. A. Jensen, Langmuir 29: 9138–9148.
K. Savolainen, S. Matikainen, and H. Alenius. 2011. Long, needle-like carbon 82. França, A., P. Aggarwal, E. V. Barsov, S. V. Kozlov, M. A. Dobrovolskaia, and
nanotubes and asbestos activate the NLRP3 inflammasome through a similar Á. González-Fernández. 2011. Macrophage scavenger receptor A mediates the
mechanism. ACS Nano 5: 6861–6870. uptake of gold colloids by macrophages in vitro. Nanomedicine (Lond.) 6: 1175–
55. Martinon, F., A. Mayor, and J. Tschopp. 2009. The inflammasomes: guardians of 1188.
the body. Annu. Rev. Immunol. 27: 229–265. 83. Yen, H. J., S. H. Hsu, and C. L. Tsai. 2009. Cytotoxicity and immunological
56. Dostert, C., V. Pétrilli, R. Van Bruggen, C. Steele, B. T. Mossman, and response of gold and silver nanoparticles of different sizes. Small 5: 1553–1561.
J. Tschopp. 2008. Innate immune activation through Nalp3 inflammasome 84. Sumbayev, V. V., I. M. Yasinska, C. P. Garcia, D. Gilliland, G. S. Lall,
sensing of asbestos and silica. Science 320: 674–677. B. F. Gibbs, D. R. Bonsall, L. Varani, F. Rossi, and L. Calzolai. 2013. Gold
57. Haniu, H., N. Saito, Y. Matsuda, T. Tsukahara, K. Maruyama, Y. Usui, K. Aoki, nanoparticles downregulate interleukin-1b-induced pro-inflammatory responses.
S. Takanashi, S. Kobayashi, H. Nomura, et al. 2013. Culture medium type affects Small 9: 472–477.
endocytosis of multi-walled carbon nanotubes in BEAS-2B cells and subsequent 85. Tsai, C. Y., S. L. Lu, C. W. Hu, C. S. Yeh, G. B. Lee, and H. Y. Lei. 2012. Size-
biological response. Toxicol. In Vitro 27: 1679–1685. dependent attenuation of TLR9 signaling by gold nanoparticles in macrophages. J.
58. Wan, B., Z. X. Wang, Q. Y. Lv, P. X. Dong, L. X. Zhao, Y. Yang, and L. H. Guo. Immunol. 188: 68–76.
2013. Single-walled carbon nanotubes and graphene oxides induce autophagosome 86. Moyano, D. F., M. Goldsmith, D. J. Solfiell, D. Landesman-Milo, O. R. Miranda,
accumulation and lysosome impairment in primarily cultured murine peritoneal D. Peer, and V. M. Rotello. 2012. Nanoparticle hydrophobicity dictates immune
macrophages. Toxicol. Lett. 221: 118–127. response. J. Am. Chem. Soc. 134: 3965–3967.
59. Li, R., Z. Ji, H. Qin, X. Kang, B. Sun, M. Wang, C. H. Chang, X. Wang, 87. Kim, E. Y., R. Schulz, P. Swantek, K. Kunstman, M. H. Malim, and
H. Zhang, H. Zou, et al. 2014. Interference in autophagosome fusion by rare earth S. M. Wolinsky. 2012. Gold nanoparticle-mediated gene delivery induces wide-
nanoparticles disrupts autophagic flux and regulation of an interleukin-1b pro- spread changes in the expression of innate immunity genes. Gene Ther. 19: 347–
ducing inflammasome. ACS Nano 8: 10280–10292. 353.
60. Seleverstov, O., O. Zabirnyk, M. Zscharnack, L. Bulavina, M. Nowicki, 88. Sadauskas, E., G. Danscher, M. Stoltenberg, U. Vogel, A. Larsen, and H. Wallin.
J. M. Heinrich, M. Yezhelyev, F. Emmrich, R. O’Regan, and A. Bader. 2006. 2009. Protracted elimination of gold nanoparticles from mouse liver. Nano-
Quantum dots for human mesenchymal stem cells labeling. A size-dependent medicine (Lond.) 5: 162–169.
autophagy activation. Nano Lett. 6: 2826–2832. 89. Balasubramanian, S. K., J. Jittiwat, J. Manikandan, C. N. Ong, L. E. Yu, and
61. Zhang, Y., F. Zheng, T. Yang, W. Zhou, Y. Liu, N. Man, L. Zhang, N. Jin, W. Y. Ong. 2010. Biodistribution of gold nanoparticles and gene expression
Q. Dou, Y. Zhang, et al. 2012. Tuning the autophagy-inducing activity of changes in the liver and spleen after intravenous administration in rats. Biomaterials
lanthanide-based nanocrystals through specific surface-coating peptides. Nat. 31: 2034–2042.
Mater. 11: 817–826. 90. Dietert, R. R. 2014. Developmental Immunotoxicity, Perinatal Programming, and
62. Wu, L., Y. Zhang, C. Zhang, X. Cui, S. Zhai, Y. Liu, C. Li, H. Zhu, G. Qu, Noncommunicable Diseases: Focus on Human Studies. Adv. Med. 2014: 867805.
G. Jiang, and B. Yan. 2014. Tuning cell autophagy by diversifying carbon 91. Ahrenhoerster, L. S., E. R. Tate, P. A. Lakatos, X. Wang, and M. D. Laiosa. 2014.
nanotube surface chemistry. ACS Nano 8: 2087–2099. Developmental exposure to 2,3,7,8 tetrachlorodibenzo-p-dioxin attenuates ca-
63. Moghimi, S. M. 2002. Chemical camouflage of nanospheres with a poorly reactive pacity of hematopoietic stem cells to undergo lymphocyte differentiation. Toxicol.
surface: towards development of stealth and target-specific nanocarriers. Biochim. Appl. Pharmacol. 277: 172–182.
Biophys. Acta 1590: 131–139. 92. Hogaboam, J. P., A. J. Moore, and B. P. Lawrence. 2008. The aryl hydrocarbon
64. Ishida, T., X. Wang, T. Shimizu, K. Nawata, and H. Kiwada. 2007. PEGylated receptor affects distinct tissue compartments during ontogeny of the immune
liposomes elicit an anti-PEG IgM response in a T cell-independent manner. J. system. Toxicol. Sci. 102: 160–170.
Control. Release 122: 349–355. 93. Laiosa, M. D., and E. R. Tate. 2015. Fetal Hematopoietic Stem Cells Are the
65. Wang, X., T. Ishida, and H. Kiwada. 2007. Anti-PEG IgM elicited by injection of Canaries in the Coal Mine That Portend Later Life Immune Deficiency. Endo-
liposomes is involved in the enhanced blood clearance of a subsequent dose of crinology 156: 3458–3465.
PEGylated liposomes. J. Control. Release 119: 236–244. 94. Winans, B., A. Nagari, M. Chae, C. M. Post, C. I. Ko, A. Puga, W. L. Kraus, and
66. Ishida, T., K. Atobe, X. Wang, and H. Kiwada. 2006. Accelerated blood clearance B. P. Lawrence. 2015. Linking the aryl hydrocarbon receptor with altered DNA
of PEGylated liposomes upon repeated injections: effect of doxorubicin- methylation patterns and developmentally induced aberrant antiviral CD8+ T cell
encapsulation and high-dose first injection. J. Control. Release 115: 251–258. responses. J. Immunol. 194: 4446–4457.
The Journal of Immunology 3225

95. Boule, L. A., C. G. Burke, B. M. Fenton, K. Thevenet-Morrison, T. A. Jusko, and 105. Corsini, E., M. Sokooti, C. L. Galli, A. Moretto, and C. Colosio. 2013. Pesticide
B. P. Lawrence. 2015. Developmental activation of the AHR increases effector induced immunotoxicity in humans: a comprehensive review of the existing evi-
CD4+ T cells and exacerbates symptoms in autoimmune disease-prone Gnaq+/2 dence. Toxicology 307: 123–135.
mice. Toxicol. Sci. 148: 555–566. 106. Ashley-Martin, J., A. R. Levy, T. E. Arbuckle, R. W. Platt, J. S. Marshall, and
96. Boule, L. A., B. Winans, K. Lambert, B. A. Vorderstrasse, D. J. Topham, L. Dodds. 2015. Maternal exposure to metals and persistent pollutants and cord
M. S. Pavelka, Jr., and B. P. Lawrence. 2015. Activation of the aryl hydrocarbon blood immune system biomarkers. Environ. Health 14: 52.
receptor during development enhances the pulmonary CD4+ T-cell response to 107. Leifer, C. A., and R. R. Dietert. 2011. Early life environment and developmental
viral infection. Am. J. Physiol. Lung Cell. Mol. Physiol. 309: L305–L313. immunotoxicity in inflammatory dysfunction and disease. Toxicol. Environ. Chem.
97. Boitano, A. E., J. Wang, R. Romeo, L. C. Bouchez, A. E. Parker, S. E. Sutton, 93: 1463–1485.
J. R. Walker, C. A. Flaveny, G. H. Perdew, M. S. Denison, et al. 2010. Aryl 108. Dietert, R. R., and M. S. Piepenbrink. 2006. Lead and immune function. Crit.
hydrocarbon receptor antagonists promote the expansion of human hematopoietic Rev. Toxicol. 36: 359–385.
stem cells. Science 329: 1345–1348. 109. Gao, D., T. K. Mondal, and D. A. Lawrence. 2007. Lead effects on development
98. Singh, K. P., F. L. Casado, L. A. Opanashuk, and T. A. Gasiewicz. 2009. The aryl and function of bone marrow-derived dendritic cells promote Th2 immune re-
hydrocarbon receptor has a normal function in the regulation of hematopoietic and sponses. Toxicol. Appl. Pharmacol. 222: 69–79.
other stem/progenitor cell populations. Biochem. Pharmacol. 77: 577–587. 110. Heo, Y., W. T. Lee, and D. A. Lawrence. 1998. Differential effects of lead and cAMP
99. Singh, K. P., R. W. Garrett, F. L. Casado, and T. A. Gasiewicz. 2011. Aryl hy- on development and activities of Th1- and Th2-lymphocytes. Toxicol. Sci. 43: 172–185.
drocarbon receptor-null allele mice have hematopoietic stem/progenitor cells with 111. Rogers, J. A., L. Metz, and V. W. Yong. 2013. Review: Endocrine disrupting
abnormal characteristics and functions. Stem Cells Dev. 20: 769–784. chemicals and immune responses: a focus on bisphenol-A and its potential
100. Smith, B. W., S. S. Rozelle, A. Leung, J. Ubellacker, A. Parks, S. K. Nah, mechanisms. Mol. Immunol. 53: 421–430.
D. French, P. Gadue, S. Monti, D. H. Chui, et al. 2013. The aryl hydrocarbon 112. Roy, A., S. M. Bauer, and B. P. Lawrence. 2012. Developmental exposure to
receptor directs hematopoietic progenitor cell expansion and differentiation. Blood bisphenol A modulates innate but not adaptive immune responses to influenza A
122: 376–385. virus infection. PLoS One 7: e38448.
101. Fares, I., J. Chagraoui, Y. Gareau, S. Gingras, R. Ruel, N. Mayotte, E. Csaszar, 113. Menard, S., L. Guzylack-Piriou, M. Leveque, V. Braniste, C. Lencina, M. Naturel,
D. J. Knapp, P. Miller, M. Ngom, et al. 2014. Cord blood expansion. Pyr- L. Moussa, S. Sekkal, C. Harkat, E. Gaultier, et al. 2014. Food intolerance at
imidoindole derivatives are agonists of human hematopoietic stem cell self-renewal. adulthood after perinatal exposure to the endocrine disruptor bisphenol A. FASEB
Science 345: 1509–1512. J. 28: 4893–4900.

Downloaded from http://www.jimmunol.org/ by guest on October 21, 2019


102. Holladay, S. D. 1999. Prenatal immunotoxicant exposure and postnatal autoim- 114. Nakajima, Y., R. M. Goldblum, and T. Midoro-Horiuti. 2012. Fetal exposure to
mune disease. Environ. Health Perspect. 107(Suppl. 5): 687–691. bisphenol A as a risk factor for the development of childhood asthma: an animal
103. Noakes, P. S., P. Taylor, S. Wilkinson, and S. L. Prescott. 2006. The relationship model study. Environ. Health 11: 8.
between persistent organic pollutants in maternal and neonatal tissues and im- 115. Khan, D., and S. A. Ahmed. 2015. Epigenetic Regulation of Non-Lymphoid Cells
mune responses to allergens: A novel exploratory study. Chemosphere 63: 1304– by Bisphenol A, a Model Endocrine Disrupter: Potential Implications for
1311. Immunoregulation. Front. Endocrinol. (Lausanne) 6: 91.
104. Sunyer, J., R. Garcia-Esteban, M. Alvarez, M. Guxens, F. Goñi, M. Basterrechea, 116. Boverhof, D. R., G. Ladics, B. Luebke, J. Botham, E. Corsini, E. Evans,
M. Vrijheid, S. Guerra, and J. M. Antó. 2010. DDE in mothers’ blood during D. Germolec, M. Holsapple, S. E. Loveless, H. Lu, et al. 2014. Approaches and
pregnancy and lower respiratory tract infections in their infants. Epidemiology 21: considerations for the assessment of immunotoxicity for environmental chemicals:
729–735. a workshop summary. Regul. Toxicol. Pharmacol. 68: 96–107.

You might also like