You are on page 1of 13

Focus Article

Carbon black and titanium dioxide


nanoparticles induce distinct
molecular mechanisms of toxicity
Sonja Boland,1∗ Salik Hussain2 and Armelle Baeza-Squiban1

Increasing evidence link nanomaterials with adverse biological outcomes and due
to the variety of applications and potential human exposures to nanoparticles, it
is thus important to evaluate their toxicity for the risk assessment of workers and
consumers. It is crucial to understand the underlying mechanisms of their toxi-
city as observation of similar effects after different nanomaterial exposures does
not reflect similar intracellular processing and organelle interactions. A thorough
understanding of mechanisms is needed not only for accurate prediction of poten-
tial toxicological impacts but also for the development of safer nanoapplications by
modulating the physicochemical characteristics. Furthermore biomedical applica-
tions may also take advantage of an in depth knowledge about the mode of action of
nanotoxicity to design new nanoparticle-derived drugs. In the present manuscript
we discuss the similarities and differences in molecular pathways of toxicity after
carbon black (CB) and titanium dioxide (TiO2 ) nanoparticle exposures and identify
the main toxicity mechanisms induced by these two nanoparticles which may also
be indicative for the mode of action of other insoluble nanomaterials. We address
the translocation, cell death induction, genotoxicity, and inflammation induced by
TiO2 and CB nanoparticles which depend on their internalization, reactive oxygen
species (ROS) production capacities and/or protein interactions. We summarize
their distinct cellular mechanisms of toxicity and the crucial steps which may be tar-
geted to avoid adverse effects or to induce them for nanomedical purposes. Several
physicochemical characteristics could influence these general toxicity pathways
depicted here and the identification of common toxicity pathways could support
the grouping of nanomaterials in terms of toxicity. © 2014 Wiley Periodicals, Inc.

How to cite this article:


WIREs Nanomed Nanobiotechnol 2014, 6:641–652. doi: 10.1002/wnan.1302

INTRODUCTION link nanomaterials with adverse biological outcomes


and a thorough understanding of underlying mecha-
N anomaterial induced biological processes are
among the most studied topics in the recent lit-
erature. However, molecular mechanisms leading to
nisms is needed not only for accurate prediction of
potential toxicological impacts but also for the devel-
opment of safer nanotechnology applications or to
these processes are not fully understood and are hot
conceive new biomedical applications.
topics for further explorations. Increasing evidence
Titanium dioxide (TiO2 ) nanoparticles are
∗ Correspondence
increasingly used for various industrial and con-
to: boland@univ-paris-diderot.fr
1 Univ
sumer product applications including paints, waste
Paris Diderot, (Sorbonne Paris Cité), UMR 8251 CNRS, Unit
of Functional and Adaptive Biology (BFA), Laboratory of Molecular
water treatment, sterilization, cosmetics, food addi-
and Cellular Responses to Xenobiotics (RMCX), Paris, France tive, biomedical ceramic, and implant biomaterials.
2 Clinical Research Program, NIEHS, Research Triangle Park, NC, Current estimates indicate that 3800–7800 tons of
USA nano-TiO2 are produced annually in the United
Conflict of interest: The authors have declared no conflicts of States.1 Woodrow Wilson Center database indicates
interest for this article. that approximately 9% of the consumer products

Volume 6, November/December 2014 © 2014 Wiley Periodicals, Inc. 641


Focus Article wires.wiley.com/nanomed

currently listed to contain nanomaterials contain TiO2 allow the identification of possible early stage effects
nanoparticles.2 These products include but are not or responses to low doses. Thus, it is imperative
limited to personal care products such as sunscreens, to meticulously elaborate molecular pathways after
cosmetics, and food products. Nano TiO2 is also used nanomaterial exposure to make well informed deci-
in biomedical applications like scaffolds, coatings, sions about nano-safety and for the development of
and dental implants. Both nano and micro TiO2 are safer nanomedical applications.
useful tools in tissue engineering to repair, replace, or Nanomaterial induced toxicity cannot be
enhance tissue function.3 Owing to the wide variety of explained by any single factor and a combination of
applications, TiO2 nanoparticles present substantial physical (physicochemical characteristics of particles)
potential for human exposures. Human exposures can and biological (cell type and nano-bio interactions)
occur during manufacturing and utilization of these factors determines the final outcome. These factors
nanoparticles. Limited data are available on human also dictate what type of intracellular signaling cas-
exposure to TiO2 nanoparticles through inhalation cades will be activated after nanomaterial exposure.
(most relevant route for toxicological outcomes). CB and TiO2 are both low-solubility nanoparticles
On the basis of rodent chronic inhalation studies, but differ in many physicochemical characteristics
NIOSH has recommended airborne exposure limits including crystal structure, photo activation, reactive
of 0.3 mg/m3 (10 h/day, 40 h/week) for workplace oxygen species (ROS) generation abilities and surface
exposure and 1.2 mg/m3 by Japanese NEDO (8 h/day, reactivity. These may not only influence the final
40 h/week).4 Other major routes for TiO2 expo- outcome of exposure but also explain differences in
sures are dermal (through application of cosmetics cellular mechanisms. Unfortunately very few studies
and sunscreens) and gastrointestinal [through food directly compared the effect of these two nanoma-
(E171 food additive, etc.)]. However, minimal toxic terials and thus this review aims to elucidate their
responses are observed after exposure through both respective toxicity mechanisms. We illustrate that
of these routes. CB and TiO2 nanoparticles induce similar effects
Carbon black (CB) nanoparticles are produced (apoptosis, genotoxicity, inflammation, etc.) but
in huge quantities and current worldwide production through very distinct mechanisms. In the present
is about 8.1 million metric tons (http://www.carbon- manuscript we discuss the similarities and differ-
black.org/files/carbonblackuserguide.pdf). Major ap- ences in molecular pathways of toxicity after CB and
plications of CB nanoparticles are in rubber (90% TiO2 nanoparticle exposures and identify the main
of the production), and pigment industry (9%). It is toxicity mechanisms induced by these two nanopar-
important to note that engineered CB particles differ ticles which may also be indicative for the mode of
significantly from soot particles which are unwanted action of other low-solubility nanomaterials. Several
carbonaceous byproducts of incomplete combustion physicochemical characteristics could influence these
of hydrocarbon-containing materials. These differ- general toxicity pathways depicted in this review and
ences include purity (higher in CB which is nearly the identification of common toxicity pathways could
97% carbon) surface uniformity of nodules (higher in support the grouping of nanomaterials in terms of
CB), agglomeration (higher in CB), and polycyclic aro- toxicity.
matic hydrocarbon content (higher in soot). In work-
ing environments, daily doses of CB can reach up to
0.12 mg/kg [assuming a threshold limiting value (TLV) BIODISTRIBUTION OF TiO2 AND CB
for respirable CB of 2.5 mg/m3 , 60% deposition, an
NANOPARTICLES
average weight of a person of 70 kg, an 8 h work day,
and a respiratory volume of 0.7 m3 /h).5 The deposition of nanoparticles after inhalation is
Owing to the variety of applications and poten- more dependent on the aerodynamic or thermody-
tial human exposures to CB and TiO2 nanoparticles namic diameter of the nanoparticle aggregates than on
it is thus important to evaluate their toxicity for the their chemical nature.6 However, the alveolar clear-
risk assessment of workers and consumers. It is cru- ance through nanoparticle uptake by macrophages
cial to understand the underlying mechanisms of their could be influenced by the composition and inter-
toxicity as observation of similar effects after dif- action of nanoparticles with the lung lining flu-
ferent nanomaterial exposures does not reflect simi- ids will also depend on surface characteristics such
lar intracellular processing and organelle interactions. as charge and hydrophobicity which differ between
Modern approaches of toxicology focus on the under- TiO2 and carbonaceous nanoparticles. Transloca-
standing of molecular and cellular pathways rather tion of nanoparticles to the systemic circulation has
than just investigating the final outcome as this may been observed after inhalation but the fraction of

642 © 2014 Wiley Periodicals, Inc. Volume 6, November/December 2014


WIREs Nanomedicine and Nanobiotechnology Carbon black and TiO2 induce different mechanisms of toxicity

nanomaterials reaching the blood circulation is gen- an inflammatory cell death called pyroptosis or cell
erally very low.4,6,7 This translocation toward blood death could be prevented by autophagy, a process
and secondary organs is influenced by the chemical which allows to eliminate damaged cell structures or
composition of the nanomaterial. Indeed, the biodis- organelles (for review see Ref 10). These different
tribution of iridium nanoparticles is much higher types of cell deaths (necrosis, apotosis, and pyrop-
(10%) than those of CB and TiO2 nanoparticles tosis) could be activated by nanomaterials through
which demonstrated similar translocation (3 and 2%, specific molecular and cellular signaling pathways
respectively7 ). (for review see Ref 11). Physiologically, apoptosis of
Translocation of nanomaterials through biolog- for instance airway epithelial cells functions as 1)
ical barriers depends on the physicochemical char- mechanism to reduce the hyperplastic cell numbers
acteristics such as size, shape, and surface charge, due to allergen or chemical induced hyperplasia, 2)
especially if the nanomaterial crosses the epithelial eliminate damaged cells, and 3) control inflamma-
or endothelial barrier by transcytosis involving endo- tion and thereby support barrier function.12 How-
cytic mechanisms. It has for instance been shown that ever, an exuberant apoptotic response is noted in
neutral or positively charged TiO2 nanoparticles are various airway pathologies, e.g., chronic obstructive
taken up by airway epithelial cells in contrast to neg- pulmonary disease (COPD), asthma, cystic fibrosis,
atively charged TiO2 .8 However, the paracellular pas- and interstitial lung disease. Viral/bacterial infections
sage of the endothelium may be a less specific process could lead to apoptosis to protect the organism
as nicely illustrated by a mechanistic study showing against these invading pathogens and environmental
that nanoparticles could enter the intercellular space particle exposures have also been shown to induce
and disrupt adherens junctions (Figure 1). This dis- apoptosis.
ruption has been observed with several nanomateri- Various in vitro and in vivo studies have demon-
als including TiO2 nanoparticles9 and was attributed strated the ability of CB or TiO2 nanoparticles to
to the small size (less than 22.5 nm) of the parti- induce toxicity toward the respiratory system but
cles allowing their penetration into the intercellular most of them did not address the molecular path-
space where the nanoparticles could get in con- ways of toxicity. A detailed comparative analysis
tact with the junctional protein vascular endothe- of CB and TiO2 nanoparticle induced toxic mech-
lial (VE)-cadherin disrupting their interactions and anisms in human bronchial epithelial cells (trans-
inducing the phosphorylation of VE-cadherin and sub- formed and primary cells) was performed13 and an
sequent cellular signal transduction pathways lead- overview of the resulting biological mechanisms is
ing to cell retraction through actin remodeling. This presented in Figure 3. CB and TiO2 nanoparticle
direct physical interaction with VE-cadherin led thus exposure for at least 4 h was sufficient to induce signif-
to endothelial leakiness9 which has been confirmed icant toxicity in bronchial epithelial cells. Both types
in vivo after subcutaneous and intravenous injections of nanoparticles induced cell death through apop-
of TiO2 . Epithelial and blood-brain barriers present tosis induction at same concentrations (20 μg cm−2 )
however also tight junctions to prevent passage of but through distinct pathways. CB nanoparticle expo-
molecules and which may be less easily disrupted by sure induced mitochondrial membrane potential loss
nanomaterials. (↓ΔΨm), activation of the pro-apoptotic protein BAX
and release of cytochrome c from the damaged mito-
CELL DEATH MECHANISMS AND chondria. In contrast, TiO2 nanoparticle exposure led
to lipid peroxidation, lysosomal damage, and release
GENOTOXICITY INDUCED BY TiO2
of cathepsin B.
AND CB NANOMATERIALS Differences in signaling cascades for apopto-
While induction of cell death may be beneficial in sis induction by CB and TiO2 nanoparticles can be
some cases (e.g., killing of tumor cells during ther- explained partly by their inherent capacity to gener-
apy), induction of exuberant cytotoxicity in healthy ate ROS under acellular conditions.14 CB nanoparti-
tissue usually ensue deleterious consequences. TiO2 cles demonstrated particle size/surface area dependent
and CB nanoparticle exposures result in induction oxidation of DTT (dithiothreitol) while TiO2 nanopar-
of a variety of cell death modalities (Figure 2), ticles were minimally reactive. These cellular assays
and thus lead to distinct pathophysiological conse- may, however, give false negative findings due to
quences. Uncontrolled cell death by necrosis will lead similar redox potentials between the test substance
to inflammatory outcomes whereas controlled cell (nanomaterials) and detection agents (DTT, ascor-
death, such as apoptosis induction, will avoid inflam- bate) but inside the cells multiple redox partners are
mation. On the other hand cells could also die through present with varying redox potentials which could

Volume 6, November/December 2014 © 2014 Wiley Periodicals, Inc. 643


Focus Article wires.wiley.com/nanomed

(a) Intracellular Extracellular Intracellular Before

p120 β-cat
α-cat

Actin

Y731
Y658
VE-cadherin

~22.5nm

Classical soluble factors


(b) (VEGF, histamine)
After

Receptor

Src
?
Nanoparticle

P P
Y731
Y658

1 2
Endocytic Lysosome
vesicle

4 3
Cell
retraction
Phosphorylation Internalization Lysosomal
>>22.5nm degradation

FIGURE 1 | Proposed mechanism of nano-endothelial leakiness. (a) The anatomy of an adherens junction. Intact monolayer of connected
endothelial cells is maintained by stable vascular endothelial (VE)–cadherin homophilic interactions with neighboring cells. VE–cadherin forms a
trans-homophillic interaction at the EC domains with another cis-paired VE–cadherin complex. 𝛽-catenin, p120 and VE–cadherin form a complex.
Formation of this ternary complex stabilizes the adherens junction. Distance of adherens junction is at least 22.5 nm. (b) Titanium dioxide (TiO2 )–NM
are small enough to migrate into the adherens junction; they bind and disrupt VE–cadherin homophilic interaction (1). This disruption induces the
phosphorylation of Y658 of VE-cadherin via a currently unknown kinase pathway, while the Y731 residue is phosphorylated by Src kinase. The
phosphorylation at the two residues induces the loss of interaction between VE–cadherin and 𝛽-catenin and with p120 (2). The loss of interaction of
the VE–cadherin–𝛽-catenin–p120 complex destabilizes actin and lead to actin remodeling (3). As a result the cell retracts and leakiness occurs (4).
After the binding of TiO2 -NM to VE–cadherin, VE–cadherin might be internalized and further degraded by lysosomes. Fate of VE–cadherin:
phosphorylation of VE–cadherin due to NanoEL may result in internalization and lysosomal degradation. This minimizes the overall amounts of
VE–cadherin near the vicinity of the cell membrane. TiO2 –NM might be internalized alongside VE–cadherin as it remained bound to VE–cadherin but
the final fate of the TiO2 –NM is uncertain. (Reprinted with permission from Ref 9. Copyright 2013 Nature Publishing Group)

interact with the nanomaterials. Apoptotic effects of (partly produced extracellular by surface reactions)
CB nanoparticles were completely inhibited by a spe- while TiO2 induced different types of ROS as the cel-
cific hydrogen peroxide (H2 O2 ) scavenger (PEGy- lular effects were not inhibited by this H2 O2 scav-
lated catalase) while there was no significant effect enger. ROS production by CB nanoparticles has also
on TiO2 nanoparticle induced toxicity. This further been shown to be responsible for the induction of
confirmed that CB induced H2 O2 mediated apoptosis apoptosis through activation of epidermal growth

644 © 2014 Wiley Periodicals, Inc. Volume 6, November/December 2014


WIREs Nanomedicine and Nanobiotechnology Carbon black and TiO2 induce different mechanisms of toxicity

It is interesting to note that these distinct signal-


Apoptosis
ing cascades of TiO2 and CB nanoparticles depicted
in Figure 3 were not due to differential localization
or uptake kinetics of these nanomaterials.16 However,
TiO2 nanoparticles formed larger sized intracellular
TiO2NPs CB NPs
aggregates with sharp edges which potentially explain
the selective lysosomal damage induced by these nano-
Autophagy
materials leading to the release of lysosomal pro-
teins as well as TiO2 NPs into the cytoplasm which
are thus no longer membrane bound. This is in line
with TEM images showing that CB nanoparticles are
Necrosis
mostly present in membrane bound vesicles in con-
trast to TiO2 nanoparticle aggregates which could be
FIGURE 2 | Types of cell death induced by carbon black (CB) and
observed in membrane bound vesicles as well as free
titanium dioxide nanoparticles (TiO2 NPs). Various modalities of cell
death could be induced by CB and TiO2 NPs in human cells.
in the cytoplasm.14
Several studies have confirmed the induction
of lysosomal perturbations by TiO2 in different cell
Carbon Titanium types4 and membrane damages, either physically
Black (CB) Dioxide or through ROS production by TiO2 nanopar-
(TiO2)
ticles, resulted in dose dependent hemolysis of
PEG ↑ ↑ ↑ ROS
↑ ↑ ↑ ROS
Lipid erythrocytes.17 Attachment of nanoparticles induced
catalase Perox erythrocyte surface changes leading to hemaggluti-
nation and abnormal sedimentation.17 Both nano
↓MMP Caspase-8 LMP and micro sized particles of TiO2 were also shown
Ba ax
to induce hemolysis in human erythrocytes.18 How-
Bax Bax
Lysosome ever, it is unlikely that TiO2 exposures could cause
Cathepsin B intravascular hemolysis as plasma was shown to
Cyt C
effectively abolish this response.
Interestingly, some reports describe that TiO2
Caspase-3, -7
nanoparticles induce mitochondria mediated apopto-
sis in other cell types and mitochondrial dysfunction
in vivo.4,19,20 The mitochondrial damage by nanopar-
ticles could however be induced indirectly after lysoso-
APOPTOSIS mal rupture through intracellular ROS production or
lysosomal hydrolase release.10 Discrepancies between
FIGURE 3 | Schema of the hypothetic pathways of cell death
studies may also be due to cell type, concentra-
induction by carbon black (CB) and titanium dioxide nanoparticles (TiO2
NPs) in bronchial epithelial cells. CB NP induce apoptosis by a reactive tions, endocytic mechanisms, and nanomaterial used.
oxygen species (ROS) dependent mitochondrial pathway involving loss Indeed, crystal structures (anatase or rutile) or size
of the mitochondrial membrane potential (MMP), activation of bax, and are shown to determine OHo radical generation by
release of cytochrome c resulting in activation of caspases and TiO2 nanoparticles21 and these parameters influence
subsequent DNA fragmentation. TiO2 NPs induce cell death through accordingly the apoptosis induction as anatase TiO2
lipid peroxidation and lysosomal membrane destabilization leading to nanoparticles had greater effects in neuronal cells than
cathepsin B release and subsequent activation of caspases and final rutile nanoparticles and nanometer sized TiO2 was
apoptotic events. Modulation of oxidative stress by PEG catalase
more cytotoxic than micrometer TiO2 .20
prevents cell death by blocking downstream events only in case of CB
NPs. (Image drawn in part using Servier medical art). (Reprinted with
TiO2 and CB nanoparticles have also been
permission from Ref 13. Copyright 2010 BioMed Central Ltd) shown to induce genotoxicity in vitro and in vivo
demonstrating their clastogenicity, mutagenicity and
carcinogenicity.4,22 Even though some investigations
factor receptor (EGFR) and downstream signaling.15 have shown negative results, several studies have evi-
Another alternative cell death induction mechanism denced the induction of micronuclei, gene mutations,
induced by nanomaterials is cell death receptor activa- DNA strand breaks, chromosomal alterations, and
tion and TiO2 nanoparticle exposure has been shown cell transformations mostly through ROS dependent
to activate FAS receptor.4 but also some ROS independent mechanisms. For

Volume 6, November/December 2014 © 2014 Wiley Periodicals, Inc. 645


Focus Article wires.wiley.com/nanomed

instance, TiO2 nanoparticles induced ROS indepen- used. Furthermore, at equal mass the CB nanoma-
dent phosphorylation of H2 A histone family member terial used in this study had a much greater surface
X (H2 AX), a sensitive marker of DNA strand breaks, area compared to the TiO2 nanoparticles which may
in an internalization dependent manner.23 Interest- also explain the greater effect of the CB samples.
ingly, surface coating of nanoparticles with albu- Interestingly, they demonstrated that CB nanoparti-
min attenuates phosphorylation of histone H2 AX. cles induced an inflammasome dependent pyroptosis,
The reduction of TiO2 nanoparticle toxicity by coat- a specific form of inflammatory cell death that requires
ing was also observed in other studies using PEGy- caspase-1 activity. This activation could however be
lation of TiO2 or silica coating (for review see due to ROS production which is also able to activate
Ref 4). Genotoxic effects of nanoparticles are also caspase-129 and further studies are necessary to con-
observed in vivo but are often reported to be due firm these novel results and to understand the under-
to the induction of inflammation leading to oxida- lying mechanisms.
tive stress. The role of inflammation in TiO2 nanopar- Inflammation after in vivo subchronic inhalation
ticle genotoxicity has been evidenced not only in of TiO2 and CB nanoparticles has been illustrated30
the lung but also in spleen, kidney, and liver (for but was often attributed to lung overload. TiO2
review see Ref 4). However, DNA damage in the and CB nanoparticles induced similar inflammatory
liver has also been shown to be due to direct inter- potency after 24 h at same surface area doses31 but
action of TiO2 nanoparticles with oxygen or nitro- interestingly comparative time course studies have
gen atoms of the nucleotides forming P-O(N)-TiO however shown a more persistent inflammation for
bonds.24 TiO2 nanoparticles compared to CB nanoparticles.
Short term inhalation confirmed the induction of
inflammation by TiO2 but not CB nanoparticles.30
TOXICITY MECHANISMS LEADING TO Nanoparticles can also have immunomodula-
INFLAMMATION tory effects32 which have been demonstrated for TiO2
Many in vitro studies have shown pro-inflammatory as well as carbonaceous nanomaterials. Low doses of
responses after treatment with these two nanoparticles TiO2 nanoparticles, which did not induce inflamma-
involving mitogen-activated protein kinases (MAPK), tion in healthy animals, induced epithelial damage,
nuclear factor-𝜅B, or receptor activation to induce and aggravated inflammatory and asthmatic responses
cytokine production (for review see Refs 4, 25). Some in a mouse model of occupational asthma33 and a
comparative studies report, however, different inflam- similar effect was observed for CB nanoparticles in
mogenic potencies of TiO2 and CB nanoparticles. ovalbumin sensitized mice.34 In another study it was
For instance, exposure of lung fibroblasts to TiO2 demonstrated that nano TiO2 significantly increase
nanoparticles induced IL-1𝛽 secretion and subsequent the dermal sensitization potency of a known skin
MMP1 induction in contrast to CB nanoparticles sensitizer by skewing the immune response toward
which had no effect.26 This could be due to the acti- a Th2 phenotype.35 It is worth mentioning that the
vation of the inflammasome by TiO2 nanoparticles micro-scale form of TiO2 failed to elicit similar effects.
which allows the maturation of pro-IL1𝛽 by caspase-1 These findings demonstrate the ability of TiO2 as well
activation. Cathepsin B could activate the inflamma- as CB nanoparticles to induce toxicity in compromised
some and it has indeed been shown that Il-1𝛽 release subjects with pre-existing conditions and modulate
in response to rutile and anatase TiO2 nanoparticles the immune responses. The underlying molecular and
is dependent on active cathepsin B and caspase-1.27 cellular mechanisms are however still unclear. This
The release of cathepsin B from lysosomes has been immunomodulatory effect could be due to activation
demonstrated after TiO2 nanoparticle treatment due of dendritic cells as several in vitro studies have shown
to lysosomal rupture as reported above.13 Thus, apop- maturation of dendritic cells by TiO2 and CB.36,37
tosis induction and pro-inflammatory responses in However, for the moment the findings are still con-
response to TiO2 nanoparticle exposure could be tradictory as some studies report maturation toward
induced by the same mechanism of inflammasome Th1-biased responses via activation of the inflamma-
activation through lysosomal destabilization. Contra- some complex36 and a protective effect of TiO2 in
dictory results were, however, obtained by Reisetter asthmatic animals38 while others demonstrate induc-
et al.28 who compared the toxicity mechanisms of tion of Th2 type33–35,37 responses (or Th1 as well as
CB and TiO2 nanoparticles in alveolar macrophages Th2 responses39 ). More immunotoxicological studies
showing that TiO2 nanoparticles did not elicit tox- are thus needed to clearly establish the immunomodu-
icity at the tested concentrations which may how- latory effect of these NPs and to understand the differ-
ever be due to different types of TiO2 nanoparticles ences in these studies which could be due to different

646 © 2014 Wiley Periodicals, Inc. Volume 6, November/December 2014


WIREs Nanomedicine and Nanobiotechnology Carbon black and TiO2 induce different mechanisms of toxicity

concentrations, treatment times, or different nanopar- to inhibit enzyme activity and induce conformational
ticles used. changes in arylamine N-acetyletransferase-1,44,45 a
The cardiovascular effects induced by nanoma- xenobiotic metabolizing enzyme involved in activa-
terials may either be due to direct interactions of tion/detoxification of carcinogens. Although in this
nanomaterial reaching the bloodstream or to indi- case both nanomaterials induce similar effects, the
rect consequences of inflammation induced in target magnitude of the response varies hugely. Interest-
organs. For instance, acute and chronic inflamma- ingly, the interaction of nanomaterials with proteins
tion could induce the so called acute phase response could also lead to gain of function42 as shown for
which is strongly linked to increased risk of car- fibrinogen unfolding and subsequent receptor acti-
diovascular disease. Saber et al.40 have shown that vation after binding to gold nanoparticles. A vari-
pulmonary inflammation after nanomaterial expo- ety of other proteins are also known to bind with
sure, including TiO2 and CB nanoparticles, corre- either CB or TiO2 nanomaterials including cytokines.
lated strongly with acute phase protein expression A comparative study has shown that both, CB and
in the lung but interestingly not in the liver. The TiO2 nanoparticles, adsorbed cytokines but the bind-
authors state that this pulmonary acute phase response ing was dependent on the nature of cytokine as well
may constitute a direct link between particle inhala- as on the nanoparticles.46 Furthermore, binding of
tion and risk of cardiovascular disease and may nanoparticles to proteins of the complement system
be a predictive biomarker. Indeed, the acute phase could induce the activation of this innate immune
protein serum amyloid A which was increased by system.32
TiO2 and CB nanoparticles is known to promote Interactions of several nanomaterials (includ-
atherosclerotic plaque progression and impairment ing TiO2 ) with the cytoskeleton have also been
of endothelial dysfunction which have been reported reported, leading to retardation of cell migration
after exposure to nanoparticles including TiO2 and due to destabilization of microtubule network.47
CB.4,41 Impairment of the cytoskeleton could, beside other
cellular perturbations, disturb lysosomal trafficking
leading to autophagy dysfunctions which are com-
CONSEQUENCES OF THE
monly observed after nanomaterial exposures (for
INTERACTION OF NANOMATERIALS review see Ref 10). As discussed above, nanoparticles
WITH PROTEINS able to enter the intercellular space have been shown
Another probable mechanism through which CB and to bind to junctional proteins inducing adherence
TiO2 nanomaterial can induce toxicity include their junction rupture and induction of intracellular signal
propensity to interact with proteins. The adsorp- transduction pathways leading to cell retraction and
tion of proteins on the surface of nanomaterials in endothelial leakiness.9 The interaction of nanoma-
physiological environments leads to the formation of terials with ion channels or cellular receptors, either
the so-called protein corona which has been exten- directly or through the protein corona, could also
sively studied.42 This corona could modulate the induce cell signaling. It has for instance been shown
effect of nanomaterials by influencing the aggrega- that TiO2 nanoparticles could activate membrane
tion state, biodistribution, cellular uptake, and reac- L-type calcium channels and Toll Like Recep-
tivity of the nanomaterials. To predict the protein tors (TLR) and respective downstream signaling4
affinity and selectivity in a corona formation pro- which however needs to be confirmed. Cell death
cess, a biological surface adsorption index (BSAI) receptors could also be induced by nanomaterials
was developed and specific nanodescriptors represent- as discussed above. CB nanoparticles induce EGFR
ing different adsorption forces were defined.43 This signaling leading to disruption of gap junctional
approach allowed to cluster successfully metal oxide intercellular communication,4,48 cell proliferation,
nanomaterial such as TiO2 nanoparticles as weak and apoptosis.15 CB nanoparticles may however
and carbonaceous nanomaterial as high adsorption activate EGFR indirectly through ROS production
materials. and activation of lipid raft signaling.49
In addition, this interaction of nanomaterials The binding/adsorption of TiO2 and CB
with proteins could impact the activity of the pro- nanoparticles to proteins can thus lead to unpre-
tein and thus have important physiological conse- dicted consequences. On the one hand the protein
quences. Interaction of nanomaterials with proteins corona which will form around the nanomaterials
essential for cellular functions could inactivate them in physiological environments will influence the
by either conformational changes and/or denatura- aggregation state, biodistribution, cellular uptake, and
tion. Both CB and TiO2 nanoparticles were shown reactivity of the nanomaterials. On the other hand the

Volume 6, November/December 2014 © 2014 Wiley Periodicals, Inc. 647


Focus Article wires.wiley.com/nanomed

TiO2 or CB NP

Protein

Receptors/
Enzymes Cytoskeleton Cytokines Junctional proteins
channels

Alteration of Disruption of Cell function


Disturbance of Disturbance of by signal
cell migration, cell communication
metabolism/ immune transduction
cell function, /anchoring
catabolism responses alteration
autophagy Endothelial leakiness

FIGURE 4 | Consequences of protein–nanomaterial interactions. Nanomaterials can interact with different types of proteins which could lead to a
variety of changes in normal cell function.

interaction of nanomaterials with different types of which drive the cellular effects is important not only
proteins could lead to activation/inhibition of surface to predict the toxicity of nanomaterials but also to
or cellular proteins which could induce a variety of allow their grouping in terms of hazard character-
changes in normal cell function (depicted in Figure 4). ization. A thorough mechanistic comprehension is
The interaction with enzymes or cytokines could also necessary to produce nanomaterials safer by
lead, respectively, to disturbance of the metabolism design by modulating the physicochemical charac-
and immune responses while the modification of teristics responsible for the induction of signaling
structural proteins such as cytoskeleton or cell junc- pathways leading to adverse effects, especially in
tional proteins could lead to important changes in cell case of utilization of nanomaterials in nanomedicine.
functioning (autophagy, cell migration/anchoring, cell However, biomedical applications may also take
communication, or barrier function). The normal advantage of an in depth knowledge about the mode
cell function could also be disrupted by an interaction of action of nanomaterial toxicity to design new
of nanomaterials with proteins involved in signal nanoparticle-derived drugs.
transduction pathways such as receptors or ion chan- Table 1 and Figure 5 summarize the distinct
nels, leading to the activation or inhibition of cell cellular mechanisms of toxicity induced by these
signaling. Unfortunately, there are not enough current two nanomaterials and the crucial steps which may
research efforts for these adverse scenarios and there be targeted to avoid adverse effects or in contrast
is urgent need to explore these processes as these to induce them for nanomedical purposes. Other
different protein–nanomaterial interactions could nanomaterials could share these mechanisms induced
induce important perturbations of cell function which by TiO2 and/or CB nanoparticle and deciphering their
may ultimately modulate or induce diseases rang- distinct mode of action or similarities may thus allow
ing from immune, neurodegenerative, or metabolic depicting some common nanotoxicological pathways
disorders to cancer. and physicochemical characteristics responsible for
their toxicity.
The internalization of TiO2 nanoparticles is a
CONCLUSIONS crucial step for their toxicity as the accumulation
TiO2 and CB nanoparticles could induce a large in lysosomes leads to their rupture and release of
variety of cellular mechanisms leading to toxic- hydrolases such as cathepsins and intracellular ROS
ity, some of which are similar between these two production. Furthermore, nanomaterials will also be
nanomaterials and represent maybe general pathways released into the cytoplasm allowing their access to
of nanotoxicity, whereas others are specific to each essential biomolecules or oxidative stress induction.
particle due to their different physicochemical char- Predictive toxicology or strategies to prevent TiO2
acteristics. Understanding the underlying mechanisms toxicity may thus focus on the endocytosis of TiO2

648 © 2014 Wiley Periodicals, Inc. Volume 6, November/December 2014


WIREs Nanomedicine and Nanobiotechnology Carbon black and TiO2 induce different mechanisms of toxicity

or other nanomaterials able to destabilize lysosomes.


TABLE 1 Summary of the Cellular Effects and Pathways Induced by For instance, negative surface charge or coating of
CB and TiO2 Nanoparticles
TiO2 nanoparticles successfully prevented uptake and
Protein interactions Junctional proteins (TiO2 9 ) toxicity. In addition, accumulation of nanomaterial
within intact lysosomes may also lead to perturbation
Metabolizing enzymes (CB,44,45 TiO2 45 )
of normal cellular functioning such as autophagy
Cytoskeleton (TiO2 47 ) dysfunction. Conversely, CB nanoparticles induce
Membrane proteins (CB,48,15 TiO2 4 ) toxic mechanisms mainly through ROS production
Cytokines (CB,46 TiO2 46 ) which already occurs extracellularly. Inhibiting the
internalization of CB nanoparticles may thus not be
Apoptosis Mitochondrial pathway (CB,13 TiO2 4,20 )
sufficient to avoid toxicity and reduction of surface
Lysosomal pathway (TiO2 4,13 ) reactivity could instead be aimed to reduce toxicity. It
Cell death receptors (TiO2 4 ) is important to decipher the chronology of the entire
cellular mechanism as for instance the destabilization
Pyroptosis (CB28 )
of lysosomes could induce the production of ROS
Autophagy Induction or disturbance (CB,10 TiO2 10 ) which in turn can activate signaling pathways or
Genotoxicity Clastogenic, mutagenic, carcinogenic (CB,22 alter mitochondria leading to apoptosis. CB, TiO2
TiO2 22–24 ) nanoparticles as well as many other nanoparticles
Immune response NF-𝜅B pathway activation (CB,25 TiO2 4 ) could interact directly with membrane proteins (chan-
nels, receptors, or junctional proteins, etc.) responsible
Inflammasome activation (CB,28 TiO2 27 )
for the induction of signal transduction pathways
Immunomodulatory effects (CB,34,37,39 leading to altered gene expressions, etc. Studying and
TiO2 33,35,36,38 ) predicting the adsorption capacities of nanomaterials
Cardiovascular effects Endothelial dysfunction (CB,41 TiO2 4 ) and protein interactions is thus not only important
Acute phase response (CB,40 TiO2 40 ) for determining the protein corona in physiological
environments, but also to understand the possible
The most relevant references discussed in this review are indicated by their activation/inhibition of surface or cellular proteins.
respective reference number.
In conclusion, three main modes of toxic-
ity mechanisms could be identified: internalization
Internalisation ROS production Membrane with subsequent lysosomal destabilization or accu-
protein interaction mulation, ROS production capacities and protein
interactions. Many of the toxic effects observed after
ROS nanomaterial treatment could be explained by these
Channel, receptor,
junctional protein etc three modes of actions described here, yet other
mechanisms could also be induced by nanomateri-
ROS Signal Gene
expression
als, especially if they are soluble. Cross activations
transduction
Proteins/
modulation and retroactions of these pathways exist such as
Nucleotides
Mitochondria activation of membrane proteins by ROS, etc. and
this schematic illustration of nanomaterial toxicity
Hydrolases
(cathepsin etc) Inflammasome mechanisms maybe incomplete and not represent
the whole complexity of cellular mechanisms. The
Autophagy
dysfunction Apoptosis toxicity mechanisms triggered by nanomaterials will
also depend on the cell type used (depending on their
FIGURE 5 | Main toxicity pathways induced by titanium dioxide phagocytic capacities, anti-oxidant defense mecha-
(TiO2 ) and carbon black (CB) nanoparticles. Three main pathways could
nisms, etc.), the dose and coating of the nanomaterial
be identified which are induced by TiO2 and/or CB nanoparticles and
which could be shared by other nanomaterials. The internalization of by biomolecules present in body fluids. Further stud-
nanomaterial could lead to lysosomal destabilization or accumulation. ies are needed to confirm the importance of these
Nanomaterials could produce reactive oxygen species through surface mechanisms and to verify whether these are rep-
reactions. The interaction of nanomaterial with membrane proteins resentative of other low-solubility nanomaterials.
could lead to activation or inhibition of cell signaling pathways. These More comparative mechanistic studies are needed
different pathways could induce autophagy, inflammasome activation, which should not only compare different physico-
apoptosis, or gene expressions and crucial steps may be targeted to chemical characteristics of the same nanomaterial
avoid these cellular effects (red bars). but also evaluate other types of nanomaterials with
similar properties in parallel. This identification of

Volume 6, November/December 2014 © 2014 Wiley Periodicals, Inc. 649


Focus Article wires.wiley.com/nanomed

common toxicity pathways will support the grouping influence these three general toxicity pathways
of nanomaterials in terms of toxicity and could depicted here and represent promising tools to design
help to identify relevant endpoints for qualitative safer or more reactive nanomaterials for biomedical
structure–toxicity relationship modeling approaches. applications.
Several physicochemical characteristics will indeed

ACKNOWLEDGMENTS
This work was supported (in part) by the Intramural Research Program of the NIH, National Institute of
Environmental Health Sciences (NIEHS) and by the French National Research Agency (ANR) and the German
Federal Ministry of Education and Research (BMBF), under the frame of SIINN, the ERA-NET for a Safe
Implementation of Innovative Nanoscience and Nanotechnology.

REFERENCES
1. Hendren CO, Mesnard X, Droge J, Wiesner MR. Esti- 11. De Stefano D, Carnuccio R, Maiuri MC. Nanomaterials
mating production data for five engineered nanomate- toxicity and cell death modalities. J Drug Deliv 2012,
rials as a basis for exposure assessment. Environ Sci 2012:167896.
Technol 2011, 45:2562–2569. 12. Tesfaigzi Y. Roles of apoptosis in airway epithelia. Am
2. PEN. The project on emerging nanotechnologies. The J Respir Cell Mol Biol 2006, 34:537–547.
Woodrow Wilson International Center for Scholars, 13. Hussain S, Thomassen LC, Ferecatu I, Borot MC,
Washington, DC, USA, 2014. Andreau K, Martens JA, Fleury J, Baeza-Squiban A,
3. Schanen BC, Karakoti AS, Seal S, Drake DR, Warren Marano F, Boland S. Carbon black and titanium diox-
WL, Self WT. Exposure to titanium dioxide nanoma- ide nanoparticles elicit distinct apoptotic pathways in
terials provokes inflammation of an in vitro human bronchial epithelial cells. Part Fibre Toxicol 2010, 7:10.
immune construct. ACS Nano 2009, 3:2523–2532. 14. Hussain S, Boland S, Baeza-Squiban A, Hamel R,
4. Shi H, Magaye R, Castranova V, Zhao J. Titanium Thomassen LC, Martens JA, Billon-Galland MA,
dioxide nanoparticles: a review of current toxicological Fleury-Feith J, Moisan F, Pairon JC, et al. Oxidative
data. Part Fibre Toxicol 2013, 10:15. stress and proinflammatory effects of carbon black and
5. Vesterdal LK, Folkmann JK, Jacobsen NR, Sheykhzade titanium dioxide nanoparticles: role of particle sur-
M, Wallin H, Loft S, Moller P. Pulmonary exposure face area and internalized amount. Toxicology 2009,
to carbon black nanoparticles and vascular effects. Part 260:142–149.
Fibre Toxicol 2010, 7:33. 15. Sydlik U, Bierhals K, Soufi M, Abel J, Schins RP,
6. Geiser M, Kreyling WG. Deposition and biokinetics of Unfried K. Ultrafine carbon particles induce apoptosis
inhaled nanoparticles. Part Fibre Toxicol 2010, 7:2. and proliferation in rat lung epithelial cells via specific
signaling pathways both using EGF-R. Am J Physiol
7. Kreyling WG, Semmler-Behnke M, Takenaka S,
Lung Cell Mol Physiol 2006, 291:L725–L733.
Möller W. Differences in the biokinetics of inhaled
nano-versus micrometer-sized particles. Acc Chem Res 16. Belade E, Armand L, Martinon L, Kheuang
2012, 46(3):714–722. L, Fleury-Feith J, Baeza-Squiban A, Lanone S,
8. Vranic S, Boggetto N, Contremoulins V, Mornet S, Billon-Galland MA, Pairon JC, Boczkowski J. A
Reinhardt N, Marano F, Baeza-Squiban A, Boland S. comparative transmission electron microscopy study
Deciphering the mechanisms of cellular uptake of engi- of titanium dioxide and carbon black nanoparticles
neered nanoparticles by accurate evaluation of internal- uptake in human lung epithelial and fibroblast cell
ization using imaging flow cytometry. Part Fibre Toxicol lines. Toxicol In Vitro 2012, 26:57–66.
2013, 10:2. 17. Li SQ, Zhu RR, Zhu H, Xue M, Sun XY, Yao SD, Wang
9. Setyawati MI, Tay CY, Chia SL, Goh SL, Fang W, SL. Nanotoxicity of TiO(2) nanoparticles to erythrocyte
Neo MJ, Chong HC, Tan SM, Loo SC, Ng KW, et al. in vitro. Food Chem Toxicol 2008, 46:3626–3631.
Titanium dioxide nanomaterials cause endothelial cell 18. Aisaka Y, Kawaguchi R, Watanabe S, Ikeda M, Igisu H.
leakiness by disrupting the homophilic interaction of Hemolysis caused by titanium dioxide particles. Inhal
VE-cadherin. Nat Commun 2013, 4:1673. Toxicol 2008, 20:891–893.
10. Stern ST, Adiseshaiah PP, Crist RM. Autophagy and 19. Hu R, Zheng L, Zhang T, Gao G, Cui Y, Cheng
lysosomal dysfunction as emerging mechanisms of Z, Cheng J, Hong M, Tang M, Hong F. Molecular
nanomaterial toxicity. Part Fibre Toxicol 2012, 9:20. mechanism of hippocampal apoptosis of mice following

650 © 2014 Wiley Periodicals, Inc. Volume 6, November/December 2014


WIREs Nanomedicine and Nanobiotechnology Carbon black and TiO2 induce different mechanisms of toxicity

exposure to titanium dioxide nanoparticles. J Hazard 32. Hussain S, Vanoirbeek JA, Hoet PH. Interactions of
Mater 2011, 191:32–40. nanomaterials with the immune system. Wiley Interdis-
20. Wu J, Sun J, Xue Y. Involvement of JNK and P53 cip Rev Nanomed Nanobiotechnol 2012, 4:169–183.
activation in G2/M cell cycle arrest and apoptosis 33. Hussain S, Vanoirbeek JA, Luyts K, De Vooght V,
induced by titanium dioxide nanoparticles in neuron Verbeken E, Thomassen LC, Martens JA, Dinsdale D,
cells. Toxicol Lett 2010, 199:269–276. Boland S, Marano F, et al. Lung exposure to nanoparti-
21. Li M, Yin JJ, Wamer WG, Lo YM. Mechanistic char- cles modulates an asthmatic response in a mouse model.
acterization of titanium dioxide nanoparticle-induced Eur Respir J 2011, 37:299–309.
toxicity using electron spin resonance. J Food Drug Anal 34. Alessandrini F, Beck-Speier I, Krappmann D, Weichen-
2014, 22:76–85. meier I, Takenaka S, Karg E, Kloo B, Schulz H, Jakob
22. Roller M. Carcinogenicity of inhaled nanoparticles. T, Mempel M, et al. Role of oxidative stress in ultrafine
Inhal Toxicol 2009, 21(Suppl 1):144–157. particle-induced exacerbation of allergic lung inflamma-
23. Toyooka T, Amano T, Ibuki Y. Titanium dioxide tion. Am J Respir Crit Care Med 2009, 179:984–991.
particles phosphorylate histone H2AX independent of 35. Hussain S, Smulders S, De Vooght V, Ectors B, Boland
ROS production. Mutat Res 2012, 742:84–91. S, Marano F, Van Landuyt KL, Nemery B, Hoet PH,
24. Li N, Ma L, Wang J, Zheng L, Liu J, Duan Y, Liu H, Vanoirbeek JA. Nano-titanium dioxide modulates the
Zhao X, Wang S, Wang H, et al. Interaction between dermal sensitization potency of DNCB. Part Fibre Tox-
nano-anatase TiO(2) and liver DNA from mice in vivo. icol 2012, 9:15.
Nanoscale Res Lett 2009, 5:108–115. 36. Schanen BC, Das S, Reilly CM, Warren WL, Self WT,
25. Donaldson K, Tran L, Jimenez LA, Duffin R, Newby Seal S, Drake DR. Immunomodulation and T helper
DE, Mills N, MacNee W, Stone V. Combustion-derived TH1 /TH2 response polarization by CeO2 and TiO2
nanoparticles: a review of their toxicology following nanoparticles. PLoS One 2013, 8:e62816.
inhalation exposure. Part Fibre Toxicol 2005, 2:10. 37. Koike E, Takano H, Inoue K, Yanagisawa R, Kobayashi
26. Armand L, Dagouassat M, Belade E, Simon-Deckers T. Carbon black nanoparticles promote the maturation
A, Le Gouvello S, Tharabat C, Duprez C, Andujar and function of mouse bone marrow-derived dendritic
P, Pairon JC, Boczkowski J, et al. Titanium diox- cells. Chemosphere 2008, 73:371–376.
ide nanoparticles induce matrix metalloprotease 38. Scarino A, Noël A, Renzi PM, Cloutier Y, Vincent
1 in human pulmonary fibroblasts partly via an R, Truchon G, Tardif R, Charbonneau M. Impact
interleukin-1𝛽-dependent mechanism. Am J Respir Cell of emerging pollutants on pulmonary inflammation in
Mol Biol 2013, 48:354–363.
asthmatic rats: ethanol vapors and agglomerated TiO2
27. Morishige T, Yoshioka Y, Tanabe A, Yao X, Tsun- nanoparticles. Inhal Toxicol 2012, 24:528–538.
oda S, Tsutsumi Y, Mukai Y, Okada N, Nakagawa
39. van Zijverden M, van der Pijl A, Bol M, van Pinxteren
S. Titanium dioxide induces different levels of IL-1𝛽
FA, de Haar C, Penninks AH, van Loveren H, Pieters
production dependent on its particle characteristics
R. Diesel exhaust, carbon black, and silica particles
through caspase-1 activation mediated by reactive oxy-
display distinct Th1/Th2 modulating activity. Toxicol
gen species and cathepsin B. Biochem Biophys Res Com-
Appl Pharmacol 2000, 168:131–139.
mun 2010, 392:160–165.
28. Reisetter AC, Stebounova LV, Baltrusaitis J, Powers 40. Saber AT, Lamson JS, Jacobsen NR, Ravn-Haren G,
L, Gupta A, Grassian VH, Monick MM. Induction of Hougaard KS, Nyendi AN, Wahlberg P, Madsen AM,
inflammasome-dependent pyroptosis by carbon black Jackson P, Wallin H, et al. Particle-induced pulmonary
nanoparticles. J Biol Chem 2011, 286:21844–21852. acute phase response correlates with neutrophil influx
linking inhaled particles and cardiovascular risk. PLoS
29. Alfonso-Loeches S, Urena-Peralta JR, Morillo-Bargues One 2013, 8:e69020.
MJ, De La Cruz JO, Guerri C. Role of mitochon-
dria ROS generation in ethanol-induced NLRP3 inflam- 41. Møller P, Mikkelsen L, Vesterdal LK, Folkmann JK,
masome activation and cell death in astroglial cells. Forchhammer L, Roursgaard M, Danielsen PH, Loft S.
Front Cell Neurosci 2014, 1;8:216. doi: 10.3389/fncel. Hazard identification of particulate matter on vasomo-
2014.00216. tor dysfunction and progression of atherosclerosis. Crit
Rev Toxicol 2011, 41:339–368.
30. Klein CL, Wiench K, Wiemann M, Ma-Hock L, van
Ravenzwaay B, Landsiedel R. Hazard identification 42. Walkey CD, Chan WC. Understanding and control-
of inhaled nanomaterials: making use of short-term ling the interaction of nanomaterials with proteins in
inhalation studies. Arch Toxicol 2012, 86:1137–1151. a physiological environment. Chem Soc Rev 2012,
31. Sager TM, Castranova V. Surface area of particle 41:2780–2799.
administered versus mass in determining the pulmonary 43. Xia XR, Monteiro-Riviere NA, Mathur S, Song X, Xiao
toxicity of ultrafine and fine carbon black: comparison L, Oldenberg SJ, Fadeel B, Riviere JE. Mapping the
to ultrafine titanium dioxide. Part Fibre Toxicol 2009, surface adsorption forces of nanomaterials in biological
6:15. systems. ACS Nano 2011, 5:9074–9081.

Volume 6, November/December 2014 © 2014 Wiley Periodicals, Inc. 651


Focus Article wires.wiley.com/nanomed

44. Sanfins E, Dairou J, Hussain S, Busi F, Chaffotte AF, 47. Tay CY, Cai P, Setyawati MI, Fang W, Tan LP, Hong
Rodrigues-Lima F, Dupret JM. Carbon black nanopar- CH, Chen X, Leong DT. Nanoparticles strengthen
ticles impair acetylation of aromatic amine carcinogens intracellular tension and retard cellular migration.
through inactivation of arylamine N-acetyltransferase Nano Lett 2014, 14:83–88.
enzymes. ACS Nano 2011, 5:4504–4511. 48. Ale-Agha N, Albrecht C, Klotz LO. Loss of gap junc-
45. Deng ZJ, Butcher NJ, Mortimer GM, Jia Z, Monteiro tional intercellular communication in rat lung epithelial
MJ, Martin DJ, Minchin RF. Interaction of human ary- cells exposed to carbon or silica-based nanoparticles.
lamine N-acetyltransferase 1 with different nanomate- Biol Chem 2010, 391:1333–1339.
rials. Drug Metab Dispos 2014, 42:377–383. 49. Peuschel H, Sydlik U, Grether-Beck S, Felsner I, Stöck-
46. Val S, Hussain S, Boland S, Hamel R, Baeza-Squiban A, mann D, Jakob S, Kroker M, Haendeler J, Gotić
Marano F. Carbon black and titanium dioxide nanopar- M, Bieschke C, et al. Carbon nanoparticles induce
ticles induce pro-inflammatory responses in bronchial ceramide- and lipid raft-dependent signalling in lung
epithelial cells: need for multiparametric evaluation due epithelial cells: a target for a preventive strategy against
to adsorption artifacts. Inhal Toxicol 2009, 21(Suppl environmentally-induced lung inflammation. Part Fibre
1):115–122. Toxicol 2012, 9:48.

FURTHER READING
Casciano DA, Sahu SC, eds. Systems Toxicology: From Omics Technology to Nanotechnology. Hoboken, New Jersey, USA:
John Wiley and Sons Ltd.; 2011.
Bhushan B, ed. Encyclopedia of Nanotechnology. Heidelberg, Germany: Springer; 2011.
Houdy P, Lahmani M, Marano F, eds. Nanoethics and Nanotoxicology. Heidelberg, Germany: Springer; 2011.
Brayner R, Fievet F, Cordani T, eds. Nanomaterials: A Danger or a Promise? Heidelberg, Germany: Springer; 2013.
Monteiro-Riviere NA, Tran CL, eds. Nanotoxicology. 2nd ed. Boca Raton, Florida, USA: CRC Press; 2014.
Chen Y, Capco D, eds. Nanomaterial: Impacts on Cell Biology and Medicine. Heidelberg, Germany: Springer; 2014.

652 © 2014 Wiley Periodicals, Inc. Volume 6, November/December 2014


Copyright of WIREs: Nanomedicine & Nanobiotechnology is the property of Wiley-
Blackwell and its content may not be copied or emailed to multiple sites or posted to a listserv
without the copyright holder's express written permission. However, users may print,
download, or email articles for individual use.

You might also like