Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Trends in Analytical Chemistry 84 (2016) 160171

Contents lists available at ScienceDirect

Trends in Analytical Chemistry


j o u r n a l h o m e p a g e : w w w. e l s e v i e r. c o m / l o c a t e / t r a c

Analytical methodologies for nanotoxicity assessment


Encarnacin Caballero-Daz *, Miguel Valcrcel Cases
Department of Analytical Chemistry, University of Crdoba, Annex Marie Curie Building, C.U. Rabanales, E-14071, Crdoba, Spain

A R T I C L E I N F O A B S T R A C T

Keywords:
Comprehensive research on toxicological effects of nanoparticles is of vital importance owing to their
Nanotoxicity
increasing manufacturing and use. This review outlines critical aspects in nanotoxicity assessment, from
Nanoparticles
In vitro
the primordial characterization of physicochemical properties of nanoparticles, to the most important
In vivo analytical strategies/techniques for elucidating their biological behaviour at cellular and organismal levels.
Assays 2016 Elsevier B.V. All rights reserved.
Inuential factors
Nanoparticle characterization
Analytical methods

Contents

1. Introduction ........................................................................................................................................................................................................................................................ 161


2. Cellular pathways for nanoparticle internalization .............................................................................................................................................................................. 161
3. Determining factors in nanoparticle-induced toxicity ......................................................................................................................................................................... 162
3.1. Nanoparticle intrinsic properties ................................................................................................................................................................................................... 162
3.1.1. Chemical composition ....................................................................................................................................................................................................... 162
3.1.2. Size and shape ..................................................................................................................................................................................................................... 162
3.1.3. Surface features ................................................................................................................................................................................................................... 163
3.1.4. Crystalline structure .......................................................................................................................................................................................................... 163
3.1.5. Agglomeration propensity ............................................................................................................................................................................................... 163
3.2. Other external factors ........................................................................................................................................................................................................................ 164
4. Nanotoxicity assessment ................................................................................................................................................................................................................................ 164
4.1. Conventional in vitro assessment protocols ............................................................................................................................................................................... 164
4.1.1. Genotoxicity .......................................................................................................................................................................................................................... 164
4.1.2. Oxidative stress ................................................................................................................................................................................................................... 164
4.1.3. Cell death ............................................................................................................................................................................................................................... 164
4.1.4. Reduced metabolic activity ............................................................................................................................................................................................. 165
4.1.5. Proinammatory cytokine release ................................................................................................................................................................................ 165
4.2. High-throughput screening tests ................................................................................................................................................................................................... 165
4.3. Advanced omics techniques ......................................................................................................................................................................................................... 166
4.3.1. Genomics ............................................................................................................................................................................................................................... 166
4.3.2. Proteomics ............................................................................................................................................................................................................................ 166
4.3.3. Metabolomics ...................................................................................................................................................................................................................... 166

Abbreviations: AFM, atomic force microscopy; BSA, bovine serum albumin; CFME, carbon ber microelectrode; CME, clathrin-mediated endocytosis; CvME, caveolin-
mediated endocytosis; EC50, median effective concentration; EDX, energy dispersive X-ray; EIS, electrochemical impedance sensing; DCFH-DA, dichlorodihydrouorescein
diacetate; DLS, dynamic light scattering; FBS, fetal bovine serum; FCS, fetal calf serum; FTIR, Fourier transform infrared spectroscopy; GSH, glutathione; HCS, high content
screening; HTS, high-throughput screening; ICP, inductively coupled plasma; LDH, lactate dehydrogenase; MDA, malondialdehyde; MTT, 3-(4,5-dimethylthiazolyl-2-yl)-
2,5-diphenyltetrazolium bromide; NBT, nitroblue tetrazolium; NMR, nuclear magnetic resonance; NPs, nanoparticles; NTA, nanoparticle-tracking analysis; PI, propidium
iodide; ROS, reactive oxygen species; RTCA, real time cell analyzer; SEM, scanning electron microscopy; SOD, superoxide dismutase; SRCD, synchrotron radiation circular
dichroism; SRXAS, synchrotron radiation X-ray absorption spectroscopy; SRXRF, synchrotron radiation X-ray uorescence; TEM, transmission electron microscopy; TUNEL,
terminal deoxynucleotidyl transferase deoxyuridine triphosphate nick end labeling; XANES, X-ray absorption near-edge structure; XPS, X-ray photoelectron spectroscopy;
XRD, X-ray diffraction; XRF, X-ray uorescence.
* Corresponding author. Tel.: +34 957218616; Fax: +34 957218616.
E-mail address: nayara104@hotmail.com (E.C. Daz).

http://dx.doi.org/10.1016/j.trac.2016.03.007
0165-9936/ 2016 Elsevier B.V. All rights reserved.
E. Caballero-Daz, M. Valcrcel Cases / Trends in Analytical Chemistry 84 (2016) 160171 161

4.4. Electrochemical techniques ............................................................................................................................................................................................................. 167


4.4.1. Carbon ber microelectrodes (CFMEs) ....................................................................................................................................................................... 167
4.4.2. Electrochemical impedance sensing (EIS) ................................................................................................................................................................. 167
4.5. In vivo nanotoxicity evaluation ...................................................................................................................................................................................................... 167
4.5.1. Nanoparticle uptake and biodistribution ................................................................................................................................................................... 167
4.5.2. Histological analysis .......................................................................................................................................................................................................... 168
4.5.3. Blood chemistry analysis ................................................................................................................................................................................................. 168
4.5.4. Clearance pathways ........................................................................................................................................................................................................... 168
5. Conclusions ......................................................................................................................................................................................................................................................... 169
Conict of interest ............................................................................................................................................................................................................................................ 169
Acknowledgements .......................................................................................................................................................................................................................................... 169
References ............................................................................................................................................................................................................................................................ 169

1. Introduction Finally, there are two stages that comprise in vivo evaluation. Usually,
testing of organ toxicity and other biological responses induced by
Dramatic advances in Nanoscience and Nanotechnology (N&N) NP exposure are performed in model animals.
have provoked a growing manufacturing and use of engineered NPs This review summarizes those more relevant aspects to be con-
(NPs) inevitably leading to direct and indirect emissions into the sidered in nanotoxicity studies, making special reference to the main
environment. As a consequence, evaluation of potential risks for cellular pathways for NP uptake, inuential factors, and different
human health and environment derived from exposure to NPs is a analytical approaches available for NP-induced toxicity assess-
matter of social concern these days. Donaldson et al. [1] coined for ment, from classical evaluation protocols to novel omics techniques.
the rst time in 2004 the term nanotoxicology to establish a new
subcategory of toxicology focuses on lling gaps in knowledge of
NP-induced toxicity, emphasising research efforts on those NPs more 2. Cellular pathways for nanoparticle internalization
likely to come into contact with human being. Fig. 1 illustrates the
principal work stages involve in any nanotoxicity assessment study Different cellular internalization mechanisms have been pro-
[2]. As can be appreciated, NP characterization must be taken into posed to explain the entrance of NPs through plasma membrane.
account in preliminar studies with the purpose of correlating the Fig. 2 shows a general scheme including the main mechanisms re-
biological response obtained with well-characterized and dened sponsible for cellular uptake of NPs.
NPs. Subsequently, cell-based systems are used as in vitro assess- Most research in this eld concludes that endocytic pathways
ment plataforms to elucidate the toxic impact of NPs at cellular level. constitute the main processes of transport of NPs into cells.

Fig. 1. Illustration of work stages involved in research of nanotoxicity. Reproduced with permission of [2].

Fig. 2. Schematic representation of the principal cellular uptake mechanisms of NPs.


162 E. Caballero-Daz, M. Valcrcel Cases / Trends in Analytical Chemistry 84 (2016) 160171

Endocytosis is a form of active transport by which a cell internal- 3. Determining factors in nanoparticle-induced toxicity
izes objects by enclosing them in vesicles or vacuoles that are
generated from its own cytoplasmic membrane [3]. There are two The potential harmful effects associated with NPs have been dem-
general modalities of endocytosis: phagocytosis and pinocytosis. onstrated to be highly varying depending on multiple inuential
Phagocytosis (cell eating) is the predominant mechanism for factors such as physicochemical characteristics of the tested NPs and
uptake of NPs larger than 500 nm by immune response cells (i.e. exposure conditions. This might lead to misleading and little con-
monocytes, macrophages, neutrophils and dendritic cells) [4]. This clusive results that prevent from extrapolating ndings beyond each
entry pathway is rstly activated by opsonization processes in the case of study. In order to overcome this bottleneck, adequate con-
bloodstream, in which foreign substances are recognized by small trols and complementary assays involving multiple cell types and
proteins (opsonins) that make them visible to immune response readouts are strongly required.
cells. Protein-coated NPs are then bound to receptors on the cell
plasma membrane, triggering a signal cascade in the target cell. 3.1. Nanoparticle intrinsic properties
As a result, a cell surface extension is formed engulng NP in an
intracellular vesicle (0.5 to 1 m in size) called phagosome. These It should be highlighted that one of the most important chal-
vesicles will be later fusioned with endosomes and lysosomes for lenges of nanotoxicology research is to consider the highly dynamic
digestion. Once NPs are degraded, receptors are cycled back to the chemical composition and physical properties of NPs. And it is that,
cell membrane [4,5]. Pinocytosis (cell drinking) is an entrance once synthesized, NPs experience an evolution process as a result
route that occurs in most cellular types constantly and indepen- of chemical and physical transformations, what determines their
dently from the needs of cell [4]. Through this mechanism, plasma interaction mechanisms with biological systems and resulting
membrane invaginates and engulfs materials by forming vesicles nanotoxicity. In addition, new generations of NPs are appearing with
0.55 m in size [6]. There are four modes of pinocytosis, namely: a great chemical and structural diversity what leads to highly vari-
macropinocytosis, receptor-mediated endocytosis, adsorptive pi- able modes of interaction with living systems and to a growing
nocytosis, and other alternative routes [5]. In general terms, diculty for establishing standard protocols of evaluation [9]. There
macropinocytosis allows uids and other essential generic mate- is a minimum set of NP properties that are commonly agreed in char-
rials (e.g. salts, glucose and amino acids) to be taken up into cells acterization studies for nanotoxicity assessment purposes. These
in a nonspecic manner [6]. During process, those NPs with a size include chemical composition, size, shape, surface, crystallinity, and
about 1 m and located near the plasma membrane may be also agglomeration/aggregation state.
internalized coincidentally [5]. Macropinocytosis has been de-
scribed as a signicant mechanism for the cellular uptake of 3.1.1. Chemical composition
positively charged NPs [3]. Among highly selective receptor- One of critical physicochemical characteristics inuencing the
mediated mechanisms, clathrin-mediated endocytosis (CME) is NP toxicity is their chemical composition, since elements them-
generally accepted as the primary internalization process for NPs selves show different chemical reactivity and toxicity in biological
in the range of 100120 nm through specic regions of the plasma media. Analytical techniques suitable for analysis of chemical com-
membrane enriched in clathrin proteins [3]. During CME, clathrin position of NPs include X-ray photoelectron spectroscopy (XPS),
proteins bind to proteins known as adaptor proteins 2 (AP-2) forming raman spectroscopy, X-ray uorescence spectroscopy (XRF), energy
vesicles that swallow NPs and transport them into cytoplasm. dispersive X-ray analysis (EDX), inductively coupled plasma (ICP)
After endocytosis, clathrin proteins dissociate from vesicles and analyses, fourier transform infrared spectroscopy (FTIR) and nuclear
return to the cell membrane [6]. Other typical receptor-mediated magnetic resonance (NMR) [10].
pathway in endothelial cells is the caveolin-mediated endocytosis It is logical to think that NPs composed of elements with a low
(CvME). In CvME, the protein caveolin integrates itself within the associated toxicity will induce less harmful effects than those made
cholesterol rafts present in hydrophobic domains of the plasma up of highly toxic elements. In this context, Lanone et al. [11] tested
membrane, whereby encouraging curvature and formation of ask- the toxicity of 24 engineered NPs of similar equivalent spherical di-
shaped invaginations of 5080 nm in size [7]. These vesicles nally ameter but different elemental composition in two human
detach from the membrane bringing their content to compart- pulmonary cell lines. Authors found that among all the tested NPs,
ments called caveosomes [4]. copper and zinc-based NPs presented the highest toxicity whatev-
Unlike these selective and specic mechanisms, NPs may be also er their oxidation status. In contrast, titania, alumina, ceria and
internalized by non-specic interactions with complementary zirconia NPs showed moderate toxicity, while no toxicity was ob-
binding sites on the cell surface. This process is known as adsorp- served for those tungsten carbide-based NPs. Other research works
tive pinocytosis. Cationic NPs show a high binding anity to the also conrmed a higher cytotoxicity for zinc oxide NPs in compar-
negatively charged cell surface and thus are good candidates to be ison with other NPs [12].
taken up through this mechanism [5].
Several endocytic routes seem to be clathrin- and caveolin- 3.1.2. Size and shape
independent and involve regulation by other proteins such as Ras NP size and shape result to be crucial factors as they dene both
homolog family member A, ADP-ribosylation factor 6, or the cell preferential internalization mechanisms and uptake eciency, con-
division control protein 42 homolog, Cdc42 [5]. ditioning thus the toxicological behaviour of NPs in biological
Finally, NPs may also penetrate into cells by non-endocytic environments. A variety of analytical techniques are appropiate for
mechanisms of passive diffusion involving a chemical concentra- NP size and shape determination including transmission electron
tion gradient between intra and extracellular medium. Jiang et al. microscopy (TEM), scanning electron microscopy (SEM), atomic force
[8] recently evaluated the cellular internalization of 2, 4 and 6 nm microscopy (AFM) and X-ray diffraction (XRD) [10,1316].
core gold NPs featuring neutral (zwitterionic), anionic and cat- Size-dependent nanotoxicity of silver NPs has been recently ex-
ionic headgroups. Variable results from these studies were explained amined by Chen et al. [17] in studies with sh red blood cells in
by a modulation of uptake pathway depending on surface proper- order to elucidate their suitability for biomedical applications. While
ties of NPs. According to authorsconclusions, zwitterionic gold NPs silver NPs-50 showed the highest level of adsorption and uptake,
were primarily internalized through passive diffusion, while the in- suggesting an optimal nanoparticle size for passive uptake by red
ternalization of cationic and anionic NPs was dominated by multiple blood cells, smallest NPs (silver NPs-15) were found to be more cy-
endocytic routes. totoxic as a result of oxidative stress phenomena. Research with
E. Caballero-Daz, M. Valcrcel Cases / Trends in Analytical Chemistry 84 (2016) 160171 163

different sized gold NPs revealed that 1.4 nm NPs were the most toxic serum compounds resulting in a reduced endocytosis rate and
for four cell lines compared with 15 nm NPs which were shown to cytotoxicity.
be non-toxic at up to 60-fold higher concentrations [18].
Nanotoxic effects have been also shown to vary depending on
NP shape. Spherocylindrical NPs have demonstrated to exhibit a 3.1.4. Crystalline structure
greater ability of translocation inside cells in comparison with spher- The crystallinity of NPs might also play a relevant role in their
ical, pyramidal, conic, cubic or rod-like NPs [6]. Other demonstration physicochemical behaviour and toxicological impact. Differences in
of the inuence of nanoparticle shape on nanotoxicity was re- the orientation of the atoms result in variations in chemical sta-
ported by Zheng et al. [19] when compared the nanotoxicity induced bility of NPs and in their resulting biological response. XRD is one
by several optical NPs (gold nanospheres and nanorods, silver of the classical and most used techniques to provide information
nanospheres and triangular nanoplates, and quantum dots) to human about the crystallographic structure of NPs, which is dened by the
carcinoma cells. Authors found that silver and gold nanospheres were pattern, position, intensity and shape of the diffraction peaks [21].
more toxic than silver triangular nanoplates and gold nanorods, A vast majority of studies about the inuence of NP crystallin-
respectively. ity on nanotoxicity have been conducted in model aquatic organisms
[30]. In this context, Iswarya et al. [31] investigated the possible toxic
3.1.3. Surface features impact of titania NPs in its two crystalline phases (i.e. anatase and
Surface properties of NPs (i.e. surface area, chemistry and charge) rutile) to freshwater microalgae Chlorella sp. EC50 values (i.e. con-
strongly determine their interaction with cellular membranes as rst centration of a substance that causes a certain effect in 50% of the
step for induced cytotoxicity mechanisms. Analytical techniques for tested organisms) were found to be strongly dependent on the NP
determining surface chemistry of NPs are the same that those pre- crystallinity, resulting to be 3.36 and 6.25 mg L1 for anatase and
viously commented in section 3.1.1 for chemical composition analysis. rutile NPs, respectively.
Among them, ICP analysis constitutes the most appropiate choice
for metal and metal oxide NPs when the quantication of metal ions
3.1.5. Agglomeration propensity
released into cellular environments is imperative [20]. Regarding
The NP aggregation is a quite common phenomenon in biolog-
surface charge, zeta potential of NPs is usually determined by light-
ical media containing salts and proteins that might lead to the
scattering electrophoresis or electro-acoustophoresis methods.
formation of aggregates/agglomerates with much larger hydrody-
Finally, surface area and porosity might be investigated by the
namic diameter, and shape, surface, colloidal stability and
Brunauer-Emmett-Teller analysis method which is based on gas
homogeneity very different to those of the parental material. In an
adsorption/desorption isotherms [21].
extreme case, NPs may precipitate at the bottom of the plate re-
Cytotoxicity values highly dependent on surface area of NPs were
sulting in a local concentration dose distinct from the originally
reported in cellular studies with silicon-based NPs [22]. In terms
intended one [21]. NP-tracking analysis (NTA) and dynamic light
of surface chemistry, NP functionalization might have serious effects
scattering (DLS) are the most utilized techniques for NP sizing in
on the resulting toxicity given that certain functional groups confer
liquids yielding information about changes in NP hydrodynamic
a greater biocompatibility to NPs or, on the contrary, a greater toxic
radius arising from bioconjugation events [13,32]. Whereas DLS pro-
potential. Hauck et al. [23] modulated the mammalian cellular uptake
vides a NP size distribution resulting from combination of signals
of gold nanorods by manipulating the surface charge and function-
of all NPs present in the liquid matrix, NTA allows for visualiza-
al groups located on NP surface. Authors found that the absolute
tion and monitoring of single NPs or aggregates.
quantity of uptake and consequent cytotoxicity, was dependent on
Most of research studies dene the toxicity of aggregated NPs
the chemistry of the molecules adsorbed onto the nanorod surface.
as a function of the tested NP concentration. For instance, the ag-
Other ndings reported for studies with silver NPs demonstrated
gregation behaviour of zinc oxide NPs was correlated with their toxic
that the viability of NIH/3T3 broblasts and cellular uptake were
potential to RAW 264.7 cells in a NP concentration-dependent
both dependent on NP surface chemistry. In addition, further surface
manner. According to authors, the aggregation process reduced the
modication of NPs with PEG molecules was found to decrease the
ion release from NPs and partially covered up their cytotoxic po-
NP internalization and also the cytotoxicity values [24]. As re-
tential to cells. Fig. 3 illustrates the agglomeration effect on the
ported by these authors, other publications have also demonstrated
cytotoxicity induced by zinc oxide NPs as a function of NP concen-
that the functionalization of NPs with PEG corona provides multi-
tration [33].
ple benets such as reducing nonspecic protein adsorption, cellular
uptake and cytotoxicity [25,26].
Concerning nanoparticle surface charge, it is crucial to always
investigate whether the zeta potential of NPs is positively or neg-
atively charged since this fact marks the interactions with biological
systems. Numerous reports have concluded that positively charged
NPs induced more toxic effects on cells than their negatively charged
counterparts [27]. To mention a more detailed example, Baek et al.
[28] determined the correlation between physicochemical proper-
ties of zinc oxide NPs and their cytotoxicity to human lung A549
cells by testing NPs featuring two different sizes (20 nm and 70 nm)
and charges (positive and negative). In vitro assays showed that pos-
itively charged NPs of small size exhibited the highest toxicity
probably due to their greater interaction with the negatively charged
plasma membrane. It is worthy to note in this point that the for-
mation of protein corona surrounding the NP might alter its surface
charge leading to variable biological responses. For instance, Merhi
et al. [29] found that both fetal calf serum (FCS) and bovine serum
albumin (BSA) sharply reduced the positive surface charge of NPs Fig. 3. Implications of zinc oxide NP aggregation on their cytotoxicity to murine mac-
by associations between the cationic surface of NPs and the anionic rophages RAW 264.7 cells. Reproduced with permission of [33].
164 E. Caballero-Daz, M. Valcrcel Cases / Trends in Analytical Chemistry 84 (2016) 160171

3.2. Other external factors 4.1.2. Oxidative stress


One of the most widely reported nanotoxicity mechanisms is the
Some discrepancies have been found in results from in vitro pro- generation of intracellular reactive oxygen species (ROS). The over-
tocols when NPs with similar physicochemical properties are production of ROS overwhelms the antioxidant defenses of cells
evaluated. This fact might be explained by the undeniable impact leading to oxidative stress and other side damages such as alter-
of cell type/line and conditions and constituents of the culture ation of proteins, lipid peroxidation, DNA breakage and gene
medium on colloidal stability of NPs in physiological matrices, what expression modulation [48]. There are two ways to analyze oxida-
generates important consequences on the toxicological prole of tive stress in cells: direct methods that quantify the amount of
identical NPs. Nafee et al. [34] checked the colloidal stability and intracellular ROS, and indirect methods that monitor other
cytotoxicity of chitosan-modied PLGA NPs in culture medium under side effects as a result of a prolonged oxidative stress. Both
variable conditions of study. Cell type was found to be a determin- dichlorodihydrouorescein diacetate (DCFH-DA) and nitroblue tet-
ing factor in all assays, dening a different biological response for razolium (NBT) assays belong to the rst category. DCFH-DA is
same incubation conditions and tested NPs. Equally, different pH oxidized into a green uorescent product in presence of intracel-
values of medium exerted variable cytotoxicity in terms of lactate lular ROS and the obtained uorescence signal might be directly
dehydrogenase (LDH) content. Specically, NPs acquired positive correlated to the ROS level [39]. Copper oxide NPs have been re-
surface charge in acidic conditions what led to a greater harmful cently tested in hepatocellular carcinoma cells by DCFH-DA test and
effect on cells membranes. Authors also observed that the pres- results revealed signicant intracellular ROS generation of up to 242%
ence of FBS (fetal bovine serum) in culture medium increased the in poorly differentiated cells [49]. Less used, the colorimetric NBT
cell viability in treated cells given that FBS formed a protective shell assay is based on the reduction of a tetrazolium salt to formazan
around the NPs shielding their original harmful effects. As demon- crystals in presence of superoxide anions. To give an example, Dubey
strated in these studies, cellular type and line, even its differentiation et al. [50] recently studied the impact of titanium dioxide and zinc
stage, might make cells to show a different sensitivity to the cyto- oxide NPs in cells from gill tissue of Wallago attu using this in vitro
toxic effects of NPs. It has been extensively reported in literature test.
that the induction and type of NP-induced toxicity mechanisms Concerning indirect methods, these basically contemplate the
depend on the tested cells [3537]. evaluation of glutathione (GSH) content, malondialdehyde (MDA)
formation and superoxide dismutase (SOD) activity. While GSH is
an antioxidant and its depletion is indicative of ROS overproduc-
4. Nanotoxicity assessment
tion in cells, the increase of MDA, which is a byproduct of lipid
peroxidation, is clear evidence of cellular oxidative stress [39]. These
Taking into account those challenging factors associated with re-
two biomarkers demonstrated the potential of silica NPs to induce
search in nanotoxicity, that were previously commented in section
oxidative effects in human liver cells in terms of ROS generation,
3.1, nanotoxicologists have opted for two different strategies for
lipid peroxidation and depletion of GSH following exposure [51].
nanotoxicity assessment purposes. In general terms, the rst meth-
Oxidative stress after exposure of aquatic organisms to silver NPs
odology consists in using conventional in vitro protocols through
has been also reported in studies with drosophila melanogaster and
acquisition of commercially available kits that measure a specic
zebrash [52,53]. SOD catalyzes the dismutation of superoxide anion
cellular endpoint likely to be altered by NPs. And the second, and
and its activity is a reection of superoxide production in cells. An
most interesting approach, has as objective to develop and use ad-
interesting study of Gao et al. [54] related the depletion of GSH in
vanced analytical methodologies with a greater degree of adaptation
HL7702 cells, following the treatment with gold NPs, with strong
and specicity to the assessment of nanotoxic effects. Recently,
Au-S bonding interactions between NP surface and antioxidant.
Gunsolus and colleagues [9] have extensively reviewed the analyt-
ical techniques most used in NP toxicity research, making special
4.1.3. Cell death
reference to NP physicochemical properties and transformations,
Apoptosis and necrosis (i.e. programmed and accidental cell
and their biological impact.
death) reect the ability of NPs to trigger intracellular suicide mecha-
nisms or destroy cells. NP-induced cell death assessment focuses
4.1. Conventional in vitro assessment protocols on examining the mitochondrial membrane integrity or potential,
measuring the apoptotic protein levels/activation or evaluating DNA
4.1.1. Genotoxicity fragmentation [55]. In this section, it will be discussed the princi-
The potential of some NPs to damage the cellular DNA is com- pal screening techniques to evaluate the membrane integrity, given
monly evaluated by alkaline comet assay or single-cell gel that these constitute the most common ways to measure cell death-
electrophoresis. The basis of this rapid and sensitive assay lies in related processes.
the separation of DNA fragments from lysed cells in agarose gel due
to their different migration distances. Once separated, DNA frag- 4.1.3.1. Trypan blue and propidium iodide staining tests. These vital
ments are visually analyzed by uorescence microscopy following dyes can freely cross the cell membrane when it is compromised
staining with ethidium bromide dye, mainly. The extent of migra- and stain the intracellular components. Therefore, only damaged
tion is nally interpreted as a reection of the DNA damage [38,39]. cells will appear stained [38]. Fullerol NPs were tested in human
By using this in vitro test, recent studies have demonstrated the retinal epithelial cells by trypan blue (TB) assay. Experimentally,
genotoxic potential of some NPs, such as silver and metal oxide NPs treated cells were rstly stained by TB reagent and then loaded onto
[4042]. TUNEL (terminal deoxynucleotidyl transferase deoxyuridine hematocytometer chamber and analyzed by microscopy, yielding
triphosphate nick end labeling) assay is also useful for detecting DNA information on the number of dead and viable cells. Results con-
damage-induced apoptosis mechanisms. TUNEL assays have been rmed that NP concentrations above 10 M after 17 h treatment
reported for apoptosis assessment and DNA damage in cells exposed resulted to be very cytotoxic to cells [56]. Similar scheme was fol-
to carbon nanotubes, and silver and zinc oxide NPs [4345]. Other lowed by Acar et al. [57] in order to investigate the toxicity of
approaches also available to assess the mutagenic activity of NPs titanium dioxide NPs in human amniotic uid cells.
comprise micronucleus assay, which examines the ability of NPs to Propidium iodide (PI) staining followed by ow cytometry anal-
induce the formation of micronuclei, and the evaluation of expres- ysis have also applied to examine NP-induced apoptosis in terms
sion of proteins related to DNA repair [46,47]. of alteration in cell cycle progression [5860]. Particularly, the
E. Caballero-Daz, M. Valcrcel Cases / Trends in Analytical Chemistry 84 (2016) 160171 165

execution of PI test in MCF-7 cancer cells treated with zinc oxide with controls [72,73]. However, some interferences between this
NPs showed a signicant induction of apoptosis, as determined by assay and various NPs have been reported in literature. In this line,
differences in the percentage of cells in each phase of the cellular the Lupus group [74] conrmed that titanium dioxide NPs induced
cycle compared with control cells [61]. the transformation of MTT to formazan in experiments without cells
due to the photocatalytic properties of these NPs. Equally, Kroll et al.
4.1.3.2. Neutral red staining test. Neutral red dye is only incorpo- [75] demonstrated that both carbon black and titanium dioxide NPs
rated into viable cells, consequently, unstained cells will correspond interfered with the light absorption of formazan product in MTT
to those with compromised membranes [39]. A variation of the con- assays.
ventional neutral red test is the so-called neutral red retention time Cell viability for nanotoxicity assessment purposes might be also
(NRRT) assay that measures the retention of the dye over time. NRRT evaluated by using alamar blue or resazurin assay in which a
assay followed by absorbance measurements has been applied to nonuorescent compound (alamar blue) is converted into a pink
evaluate the toxicity of copper, chromium, cobalt, gold and tita- uorescent product in metabolically active cells. Using this method,
nium NPs [62]. Gliga et al. [76] found cytotoxicity for 10 nm citrate- and PVP-
coated silver NPs in BEAS-2B cells after 24 h exposure.
4.1.3.3. Lactate dehydrogenase content test. In damaged cells, lactate Adenosine triphosphate (ATP) luminiscence assays are also other
dehydrogenase (LDH) is excreted out of cellular membrane and can available alternatives to measure cell viability by quantifying the
be quantied through a coupled enzymatic assay where a tetrazo- luminiscent signal derived from a conversion reaction of luciferin
lium salt is converted into a red formazan product that is detected to oxyluciferin in presence of ATP. Huang et al. [77] reported a sig-
spectrophotometrically. The absorbance of the reaction product is nicant decrease in ATP content of human dermal broblasts in
thus related directly to the membrane integrity disruption [38]. Cell response to gold NPs.
death caused by copper oxide, gold and silver NPs have been
recently examined by using commercially available LDH tests 4.1.5. Proinammatory cytokine release
[6365]. NPs may also trigger immune response in cells by the release
of cytokines that are important biomarkers, mainly expressed by
4.1.3.4. Phosphatidylserine translocation monitored by Annexin macrophages, that play a key role in the regulation of the immune
V. Phosphatidylserine is translocated to the outer leaet of the cel- response, inammatory reaction and phagocytosis [78]. Levels of
lular membrane in apoptotic cells, and can be monitored by using IL-1, IL-6 and TNF- cytokines in culture medium were evalu-
uorescently-labelled annexin V that binds very specically to the ated following the exposure of human monocytic cells to iron oxide
exposed phosphatidylserine allowing for its detection by ow NPs. Results showed no increases in cytokine secretion what was
cytometry and uorescence microscopy [38]. Several examples in attributed to the protective protein corona shield of NPs [79].
literature describe the use of annexin-V/PI double-staining assays
in order to distinguish apoptotic cells from necrotic cells in re- 4.2. High-throughput screening tests
sponse to NPs [66,67]. Annexin-V FITC is commonly used as a marker
of apoptosis, while PI only recognizes necrotic cells. Data from double Taking into consideration the increasing production of NPs with
staining assays are shown as percentages of unstained cells (viable tuneable physicochemical features, and multiple factors that de-
cells), those stained with PI (necrotic cells), those stained with termine their toxicity in real environments, there is an urgent need
annexin-V FITC (apoptotic cells), and dual stained cells (late apop- for methods that allow for testing multiple samples simultane-
tosis) [68]. ously. Current research in nanotoxicology proposes high-throughput
screening (HTS) platforms as an ecient alternative to overcome
4.1.3.5. Mitochondrial membrane potential and apoptotic protein this bottleneck and speed up NP risk assessment. Fig. 4 shows a ow
level/activity. Mitochondrial membrane depolarization after NP ex- chart illustrating the work stages required for performing the high
posure is a clear indicative of iniciation of apoptosis processes. The throughput nanotoxicity screening. Firstly, NPs are acquired from
state of potential is normally examined by the use of membrane- a supplier that usually distributes them as dry powders. NPs are then
permeant uorescent dyes that are able to accumulate in the characterized as-produced, in terms of chemical composition (purity),
mitochondria and modify its uorescence depending on the po- size, shape, crystallinity and surface features. Subsequently, NP dis-
tential state. For example, the typical uorescent probe JC-1 exhibits persions in liquid media (e.g. water or culture medium) are prepared
red uorescence in healthy cells, whereas it shows green uores- and further characterization is required in this stage in order to elu-
cence in cells with impaired mitochondria [69]. Consideration of cidate the colloidal stability of NPs and any change in their surface
changes in activity/level of apoptosis-related proteins might help as a result of interactions with other entities present in solution.
nanotoxicologists to elucidate NP-induced cell death mechanisms. Once characterized, NPs are tested in HTS platforms and nally, the
In this sense, caspase-3 and caspase-7 activation are common in- set of obtained data is mined and processed by advanced statisti-
dicators evaluated in nanotoxicity assays since these enzymes play cal tools [21].
a crucial role in apoptotic cascade. Increased activity/level of caspases Numerous reports in literature have conrmed the usefulness
37 has been reported in cell-based assays following incubation with of HTS assays to simultaneously evaluate multiple cellular injury
nickel oxide and gold NPs [70,71]. pathways across an array of different NPs and incubation condi-
tions (e.g. doses and exposure times) [80,81].
4.1.4. Reduced metabolic activity Available HTS approaches can be classied into two modali-
Cellular damage also might lead to a reduction in metabolic ac- ties: plate reader-based assays that use luminiscence, uorescence
tivity of cells. One of the most common analytical methods is MTT or absorption readouts, and high content screening (HCS) methods.
[3-(4,5-dimethylthiazolyl-2-yl)-2,5-diphenyltetrazolium bromide] For the rst category, NP-induced cytotoxicity might be assessed
assay where a tetrazolium salt is reduced to formazan product as by monitoring the biological response after treatment using a cock-
a result of the mitochondrial activity in alive cells. Formazan is a tail of compatible dyes each one of the which is specic for
colored compound that can be spectrophotometrically quantied examinating a cellular endpoint [21]. Following the scheme of the
at 500600 nm, resulting its absorbance value proportial to per- plate reader-based HTS assays, the toxicity of different metal and
centage of viable cells in samples. The cytotoxicity of several NPs metal oxide NPs was assayed in two cell types and Zebrash embryos
has been assayed by comparing the absorbance values of samples through several mechanisms related to oxidative stress, cationic
166 E. Caballero-Daz, M. Valcrcel Cases / Trends in Analytical Chemistry 84 (2016) 160171

Fig. 4. Flow chart with the different stages in HTS applied to nanotoxicity. Reproduced with permission of [21].

injury and metal ion shedding. Fluorescence readout-based HTS anal- examined the renal effect of cadmium-containing silica NPs in rats
ysis was performed by using a cocktail of three compatible dyes, exposed intratracheally using DNA microarray technology. Renal gene
each one of which was used to examine different cellular end- expression showed that 139 and 153 genes were differentially ex-
points such as mitochondrial depolarization and membrane pressed at 7 and 30 day, respectively, compared with control
permeability. The analysis of uorescence at varying wavelengths organisms. For the shorter exposure time, genes related to acute in-
allowed for obtaining a heat map of responses, where colder and ammation, immunity and defence processes were down-regulated,
warmer colours indicated, respectively, lower and higher percent- while 30 days after dosing, the altered gene expression was asso-
ages of cells positive to the response under different incubation ciated to cell regulation and apoptosis mechanisms.
conditions. Final HTS data demonstrated the highest lethality for
quantum dots and zinc oxide NPs in cell-based assays [82].
4.3.2. Proteomics
HCS assays are proposed as advanced tools in the integration and
Research efforts in proteomics focus on delving into those spe-
automation of quantitative uorescence microscopy and image anal-
cic cellular pathways related to protein expression that explain the
ysis. Jan and colleagues [83] conducted a comprehensive investigation
biological response provoked by NPs. Western blot analysis is the
of cytotoxicity of CdTe quantum dots to HepG2 human hepatocel-
most used approach to investigate the expresion of proteins of in-
lular carcinoma cells using various HCS assays. Fluorescence images
terest, which in most of cases are related to apoptosis processes.
from multiplexed cytotoxicity assays (i.e. cell count, nuclear area,
With this purpose, Qin et al. [90] investigated the cytotoxicity of
mitochondrial membrane potential and intracellular free calcium
graphene quantum dots (GQDs) in THP-1 macrophages. GQDs re-
concentration) were acquired, processed and analyzed by HCS plat-
sulted to decrease the expression level of anti-apoptosis protein Bcl-
form and image analysis software, providing a more sensitive and
2, and to increase the expression of apoptotic proteins Bax and Bad.
informative assessment of nanotoxic mechanisms.
Western blot analysis of Bcl-2 and Bax proteins was also per-
As well as the screening at cellular level, whole-organism screen-
formed for evaluating the harmful effects caused by copper
ing has been succesfully applied to nanotoxicity eld. Recently, it
nanoclusters in C2C12 myoblasts. Findings reported a marked de-
has been described a HTS system to measure population- and
crease in Bcl-2 anti-apoptotic protein expression level, while Bax
organism-level toxicity of 20 types of NPs using Caenorhabditis
apoptotic protein expression level was increased after treatment with
elegans as model organism. Results showed that most of NPs caused
NPs [91].
toxicity with the exception of fullerene, fullerol, titanium dioxide
and cerium oxide NPs [84].
4.3.3. Metabolomics
4.3. Advanced omics techniques Metabolomics constitutes an ideal tool for investigating NP-
organism interaction at different molecular levels by tracking the
Some biomarkers, such as genes, mRNA transcripts, proteins and altered expression pattern of small molecules (e.g. amino acids or
metabolites, might be differentially expressed in an organism when sugars) involved in metabolic and biological processes in re-
it has been exposed to a xenobiotic, yielding information about the sponse to external stimuli. To date, the main analytical techniques
specic pathways that have been altered because of exposure. Mo- used for metabolomic studies are NMR, gas chromatography/mass
lecular indicators not only provide insight into triggered toxicity spectrometry (GC/MS), liquid chromatography/mass spectrom-
mechanisms, but also inform about the modulation of the general etry (LC/MS), and capillary electrophoresis/mass spectrometry (CE/
molecular response of a cell, tissue or organism in response to NPs. MS) [92].
Some advantages of metabolomics over other omics tech-
4.3.1. Genomics niques (i.e. genomics, transcriptomics and proteomics) are: the
The use of global gene expression patterns in microarrays and reduced number of molecules of interest, the possibility of quan-
next-generation sequencing technologies allows for analysis of a mul- titative and qualitative interpretation of the results obtained from
titude of genes and pathways all at once and with no preconceived the other techniques, and the more generic nature of the provided
idea about those more signicant genes [85]. Nanotoxicity has been information [93]. Briey, for metabolomic analysis, biological samples
widely investigated by using gene expression proling as expo- are collected (i.e. biouids, tissues or cells cultures) and metabo-
sure biomarkers [8688]. To cite an example, Coccini et al. [89] lites are extracted and analyzed by spectroscopic detection, NMR
E. Caballero-Daz, M. Valcrcel Cases / Trends in Analytical Chemistry 84 (2016) 160171 167

mainly [94]. Raw data are then processed by advanced chemometric 4.4.2. Electrochemical impedance sensing (EIS)
analysis and differential small molecules are identied and dened In view of the interferences reported between some types of NPs
as biomarkers. Changes in biomarker concentrations before and after and classical endpoint methods with colorimetric and uorimet-
treatment enable researchers to map the main metabolic path- ric readouts, impedance-based systems have appeared as label-
ways involved in the perturbations caused by exposure to stimuli free and high throughput alternatives providing dynamic responses
[93]. in real time for nanotoxicity screening purposes. Real time cell ana-
As of now, applications of metabolomics to nanotoxicity eld have lyzer (RTCA) systems measure the electrical impedance through
been barely described in literature in spite of the fact that this an- microelectrodes integrated on the bottom of culture plates. The
alytical technique yields crucial information concerning specic NP- sensing mechanism is based on the measurement of electrode
induced toxicity mechanisms at molecular level. In this line, Lei et al. surface impedance which is sensitive to cellular changes that affect
[94] investigated the biochemical effects of copper NPs by analyz- to cell adhesion degree to the plate, such as cell number, viability
ing urine, serum, and liver and kidney tissues from exposed rats by and morphological alterations. The applicability of RTCAs have been
a NMR-based metabolomic approach. Biomarkers were correlated successfully demonstrated to evaluate the cytotoxicity of multiple
with mitochondrial failure, enhanced ketogenesis, fatty acid NPs [101103]. Hondroulis et al. [104] fabricated a EIS chip to assess
-oxidation, and glycolysis, what was nally associated with hepa- the toxicity of graphene to blood brain barrier (BBB) cells. EIS chip
totoxicity and nephrotoxicity mechanisms. Toxicity of several metal was designed with an array of eight gold electrodes inserted in the
oxide NPs have been also assayed by comparison of metabolic pro- bottom of well plate where cells were later seeded and incubated
les of controls and samples [92,95]. More specically, metabolomic with NPs.
abnormalities in the concentration of various cellular amino acids
and gluthatione antioxidant revealed oxidative stress mecha- 4.5. In vivo nanotoxicity evaluation
nisms in MRC-5 human fetal lung broblasts after treatment with
silica NPs [96]. Although most of research has been devoted to in vitro
nanotoxicity evaluation, biological effects observed in cells neces-
sarily require further corroboration in in vivo models where new
4.4. Electrochemical techniques factors and mechanisms come into play and contribute to dene
the toxicological prole of NPs. The concept of pharmacokinetics
Analytical electrochemistry has arisen as a simple and useful emcompasses those biological processes which NPs are subjected
methodology for shedding light on those biochemical and cellular to, such as absorption, distribution, metabolism and excretion.
events involved in nanotoxicological response. Additionally, elec-
trochemical methods also provide insight into evaluation of 4.5.1. Nanoparticle uptake and biodistribution
physicochemical properties of NPs that inuence in their biologi- Analysis of uptake and biodistribution of NPs in organisms enables
cal reactivity, such as their surface, size, oxidation state and researchers to elucidate the target tissues and organs. For this
dissolution kinetics. However, up to date, electrochemical ap- purpose, conventional microscopy techniques are considered as
proaches have been sparingly dedicated to the nanotoxicity useful analysis tools to quantify and map NPs in treated animals.
framework despite of multiple advantages of these methodolo- Particularly, electron microscopy techniques (i.e. SEM and TEM) are
gies offer over conventional evaluation protocols. In addition, most good candidates to examine the uptake and presence of NPs in
of related publications have been devoted to in vitro nanotoxicity, tissues, however, their low sensitivity and potential interferents of
while applications to in vivo models are still lacking [97]. biological samples make uorescence microscopy a more appropiate
approach for analysis. However, uorescence techniques also exhibit
some handicaps for in vivo analysis including reduced light
4.4.1. Carbon ber microelectrodes (CFMEs) penetration, interfering compounds, and photobleaching and de-
The principal application of carbon ber microelectrodes (CFMEs) stabilization of the dyes [105]. Microscopy techniques have been
is monitored electrochemical species related to physiological and extensively used for investigating the tracking and uptake of mul-
neurological processes which have been altered by NPs. CFMEs allow tiple NPs [106108]. ICP analytical techniques have been also proven
for observation of biochemical changes and exocytosis processes in for monitoring metal and metal oxide NPs in in vivo models, yield-
single cells or model animals with high temporal and spatial res- ing information on the elemental content in tissues and organs
olution [97]. zel and colleagues [98] quantied electrochemically [109,110]. Whereas these techniques provide low detection limits,
the intestinal nitric oxide (NO) concentration in live zebrash using multielemental analysis, high precision and possibility of monitor-
a CFME implanted in the intestine of embryos. Changes in NO con- ing changes over time, they are also limited when there are native
centration following exposure to copper oxide and cerium oxide NPs elements in biological systems that coincide with those from the
were monitored and correlated with results from conventional assays tested NPs [105].
based on uorescent dyes specic for NO. Copper oxide NPs were New analytical approaches, such as radiotracer and synchro-
found to increase NO concentration to almost 4 times higher com- tron techniques have arisen as potential tools for monitoring the
pared with unexposed embryos, suggesting thus an intestinal biodistribution of NPs in animals. In radiotracer techniques, NPs are
oxidative damage in organisms, while low concentrations of cerium rstly labelled with radiactive isotopes (e.g. 125I) and then moni-
oxide NPs showed scavenging effect against NO. tored in vivo with a gamma-ray detector. By using radiotracer
Concerning the application of CFME amperometry to cell- technique, distribution of silver NPs, nanodiamonds and graphene
based analyses, most of publications focus on examining effects of oxide has been shown in model organisms [111113]. Concerning
NPs on exocytosis function [99]. In this context, the Maurer-Jones synchrotron radiation techniques, there are three different modali-
group [100] evaluated the exocytosis perturbation in mast cells as ties: synchrotron radiation X-ray uorescence (SRXRF), synchrotron
a result of exposure to titanium dioxide NPs by using CFMEs. Real- radiation X-ray absorption spectroscopy (SRXAS) or also known as
time electrochemical measurements revealed that NP-induced X-ray absorption near-edge structure (XANES), and synchrotron ra-
oxidative stress under UV light exposure was correlated to pertur- diation circular dichroism (SRCD). The principal attribute of these
bations in exocytotic cellular function so more ROS present caused techniques is the use of a tuneable, very bright and focused light
a greater decrease in the number of serotonin molecules being source that improves the signal to noise ratio, reduces analysis time
released. and provides single cell resolution [114]. Plainly, the former
168 E. Caballero-Daz, M. Valcrcel Cases / Trends in Analytical Chemistry 84 (2016) 160171

modality focuses on investigating the microdistribution of trace el- in mice after administration of titanium dioxide NPs. Fluores-
ements in biological matrices. SRXAS or XANES yields insight into cence images obtained by SRXRF clearly indicate how NPs
the chemical speciation of NPs in biological environments for a better preferentially accumulate in lungs.
understanding of their interactions with other biological entities.
And nally, SRCD is useful to examine protein-NP interactions [115]. 4.5.3. Blood chemistry analysis
These techniques have been already applied to explore the physi- Any positive or negative deviation in blood constituents of NP-
cochemical changes experimented by quantum dots in vivo, the treated animals compared with control groups is a clear nanotoxicity
biodistribution of titanium dioxide NPs in mice, and the interac- indicator [121]. Within this context, Jenkins et al. [122] analyzed
tions of protein corona with gold nanorods [116118]. blood samples from mice injected with gold-iron oxide NPs 14 days
after administration. As blood chemistry markers, researchers se-
4.5.2. Histological analysis lected Na+, K+, glucose, creatinine and urea nitrogen. In addition, other
Histological examination of tissues allows for investigation of parameters were also contemplated for analysis including, total bili-
damages, inammation or lesions in organs as a consequence of NP rubin, white blood cell count, mean corpuscular count and mean
treatment. As a general rule, organ sections are rstly xed with a corpuscular hemoglobin. Findings conrmed no toxicity of these NPs
chemical xative (e.g. formalin), embedded in paran, cut in thin towards animals under the treatment conditions.
sections with a microtome, and then mounted on glass slides. Finally,
sections are stained with a dye, commonly hematoxylin and eosin 4.5.4. Clearance pathways
(H&E), and examined under microscopy [119]. The most used organs Of special importance for nanomaterial biosafety is to take into
for histological analysis comprise brain, kidney, stomach, liver, lung, account the excretion routes from the body, since this might de-
heart and spleen. Song et al. [120] reported histological abnormali- termine the unexpected appearance of long-term toxic effects or,
ties in gills of three species of freshwater sh following incubation on the contrary, the reversibility of the observed acute toxicity. Moni-
with copper NPs. Fig. 5 shows images from biodistribution study toring of NP excretion can be performed by ICP analyses of
and histological analysis conducted by Zhang and colleagues [117] urine and feces samples collected at different time points after

Fig. 5. Distribution of titanium element in lungs (A and B), liver (C and D) and spleen (E and F) after 1 day of administration in mice at a dose of 1 mg Kg1. X-ray uores-
cence image (right) is paired with the corresponding histological image (left) obtained with H&E staining. Reproduced with permission of [117].
E. Caballero-Daz, M. Valcrcel Cases / Trends in Analytical Chemistry 84 (2016) 160171 169

administration. Inductively coupled plasma optical emission spec- [4] A. Panariti, G. Miserocchi, I. Rivolta, The effect of nanoparticle uptake on cellular
behavior: disrupting or enabling functions?, Nanotechnol. Sci. Appl. 5 (2012)
trometry (ICP-OES) is normally used to measure the elemental
87100.
content from NPs in biological samples. For example, Fu et al. [123] [5] H. Kettiger, A. Schipanski, P. Wick, J. Huwyler, Engineered nanomaterial uptake
analyzed by ICP-OES the silicon content in liver, spleen, kidney, lung, and tissue distribution: from cell to organism, Int. J. Nanomed. 8 (2013)
muscle, intestine, feces and urine samples 24 h and 7 days after ad- 32553269.
[6] C.M. Beddoes, C.P. Case, W.H. Briscoe, Understanding nanoparticle cellular
ministration of silica NPs to mice. Data demonstrated that the vast entry: a physicochemical perspective, Adv. Colloid Interface Sci. 218 (2015)
majority of NPs were excreted through feces. In other study, ICP- 4868.
MS data of samples from mice treated with different-sized gold NPs [7] K.G. Rothberg, J.E. Heuser, W.C. Donzell, Y.-S. Ying, J.R. Glenney, R.G.W.
Anderson, Caveolin, a protein component of caveolae membrane coats, Cell
revealed that the primary clearance via for all NPs was the renal 68 (1992) 673682.
route given that, urine gold concentration was signicantly high at [8] Y. Jiang, S. Huo, T. Mizuhara, R. Das, Y.-W. Lee, S. Hou, et al., The interplay of
2 and 5 h after administration, while the gold concentration in feces size and surface functionality on the cellular uptake of sub-10 nm gold NPs,
ACS Nano 9 (2015) 99869993.
remained low throughout the study [124]. [9] I.L. Gunsolus, C.L. Haynes, Analytical aspects of nanotoxicology, Anal. Chem.
(2015) doi:10.1021/acs.analchem.5b04221 Just accepted manuscript.
[10] K.-M. Kim, J.H. Song, M.-K. Kim, S.-T. Chung, J. Jeong, J.-Y. Yang, et al.,
5. Conclusions
Physicochemical analysis methods for nanomaterials considering their
toxicological evaluations, Mol. Cell. Toxicol. 10 (2014) 347360.
Properly evaluating of risks associated with NP exposure is a [11] S. Lanone, F. Rogerieux, J. Geys, A. Dupont, E. Maillot-Marechal, J. Boczkowski,
et al., Comparative toxicity of 24 manufactured NPs in human alveolar
matter of social concern due to the growing use and applications
epithelial and macrophage cell lines, Part. Fibre Toxicol. 6 (2009) 14.
of nanomaterials in the current society, what becomes likely the [12] H. Liu, D. Yang, H. Yang, H. Zhang, W. Zhang, Y. Fang, et al., Comparative study
fact that human being can come into contact with NPs somehow. of respiratory tract immune toxicity induced by three sterilisation
Much research efforts have been dedicated to design and synthe- nanoparticles: silver zinc oxide and titanium dioxide, J. Hazard. Mater. 248249
(2013) 478486.
size safer NPs for which, it is strongly required to evaluate the effects [13] M.V.D.Z. Park, W. Annema, A. Salvati, A. Lesniak, A. Elsaesser, C. Barnes, et al.,
of the manufactured NPs on human health and environment and In vitro developmental toxicity test detects inhibition of stem cell
to propose alternative synthesis methods that allow for obtaining differentiation by silica nanoparticles, Toxicol. Appl. Pharm. 240 (2009)
108116.
more biocompatible NPs. As a general rule, before performing any [14] R.G. Mendes, B. Koch, A. Bachmatiuk, A.A. El-Gendy, Y. Krupskaya, A. Springe,
nanotoxicity analysis, it is mandatory to execute a previous and ex- et al., Synthesis and toxicity characterization of carbon coated iron oxide
haustive characterization study of the NPs of interest, where a nanoparticles with highly dened size distributions, Biochim. Biophys. Acta
2014 (1840) 160169.
minimum set of physicochemical features should be tested, includ- [15] D. Sekar, M.L. Falcioni, G. Barucca, G. Falcioni, DNA damage and repair following
ing chemical composition, size, shape, surface, crystallinity and in vitro exposure to two different forms of titanium dioxide nanoparticles on
colloidal stability. Equally, multiple inuential factors such as con- trout erythrocyte, Environ. Toxicol. 29 (2014) 117127.
[16] M. Baalousha, A. Prasad, J.R. Lead, Quantitative measurement of the
ditions of culture medium, or the cell type should be considered
nanoparticle size and number concentration from liquid suspensions by atomic
for their impact on NP-induced biological response. Numerous an- force microscopy, Environ. Sci. Process. Impacts 16 (2014) 13381347.
alytical methodologies are currently used for nanotoxicity assessment [17] L.Q. Chen, L. Fang, J. Ling, C.Z. Ding, B. Kang, C.Z. Huang, Nanotoxicity of silver
nanoparticles to red blood cells: size-dependent adsorption, uptake and
purposes comprising from the more conventional testing proto-
hemolytic activity, Chem. Res. Toxicol. 28 (2015) 501509.
cols, most of which are commercially available kits for determining [18] Y. Pan, S. Neuss, A. Leifert, M. Fischler, F. Wen, U. Simon, et al., Size-dependent
a specic cellular endpoint, to advanced analytical techniques, such cytotoxicity of gold nanoparticles, Small 3 (2007) 1941.
as omics techniques (i.e. genomics, transcriptomics, proteomics [19] Q. Zheng, D. Shao, W. Ji, J. Li, L. Chen, J. Song, The nanotoxicity investigation
of optical nanoparticles to cultured cells in vitro, Toxicol. Rep. 1 (2014)
and metabolomics), or high throughput electrochemical screen- 137144.
ing approaches. While most of publications in this eld focus on [20] Y. Araj, T. Miyayama, S. Hirano, Difference in the toxicity mechanism between
investigating cellular responses based on in vitro studies, further cor- ion and nanoparticle forms of silver in the mouse lung and in macrophages,
Toxicology 328 (2015) 8492.
roboration of the observed effects with in vivo models would provide [21] R. Damoiseaux, S. George, M. Li, S. Pokhrel, Z. Ji, B. France, et al., No time to
further understanding on the toxicological pattern of NPs in living lose-high throughput screening to assess nanomaterial safety, Nanoscale 3
systems. Overall, there is an urgent need for: (i) new advances in (2011) 13451360.
[22] V. Rabolli, L.C. Thomassen, F. Uwambayinema, J.A. Martens, D. Lison, The
analytical techniques for testing nanotoxicity, (ii) consideration of cytotoxic activity of amorphous silica nanoparticles is mainly inuenced by
inuential factors, (iii) special attention to the limitations of the surface area and not by aggregation, Toxicol. Lett. 206 (2011) 197203.
current assessment protocols, and (iv) appropiate controls and com- [23] T.S. Hauck, A.A. Ghazani, W.C.W. Chan, Assessing the effect of surface chemistry
on gold nanorod uptake, toxicity, and gene expression in mammalian cells,
plementary studies. All these factors will contribute to establish a
Small 4 (2008) 153159.
better evaluation scenario yielding reliable information on poten- [24] E. Caballero-Daz, C. Pfeiffer, L. Kastl, P. Rivera-Gil, B. Simonet, M. Valcrcel,
tial risks of NPs. et al., The toxicity of silver nanoparticles depends on their uptake by cells
and thus on their surface chemistry, Part. Part. Syst. Char. 30 (2013) 1079
1085.
Conict of interest [25] M. Piazza, M. Colombo, I. Zanoni, F. Granucci, P. Tortora, J. Weiss, et al., Uniform
lipopolysaccharide (LPS)-loaded magnetic nanoparticles for the investigation
of LPS/TLR4 signaling, Angew. Chem. Int. Ed. Engl. 50 (2011) 622626.
The authors have declared no conict of interest. [26] A. Orlando, M. Gregori, E. Cazzaniga, M. Colombo, A. Panariti, D. Prosperi, et al.,
Iron oxide nanoparticles surface coating and cell uptake affect biocompatibility
and inammatory responses of endothelial cells and macrophages, J. Nanopart.
Acknowledgements Res. 17 (2015) 351.
[27] S. Bhattacharjee, I.M.C.M. Rietjens, M.P. Singh, T.M. Atkins, T.K. Purkait, Z. Xu,
Authors would like to express their gratitude to the Spains Min- et al., Cytotoxicity of surface-functionalized silicon and germanium
nanoparticles: the dominant role of the surface charges, Nanoscale 5 (2013)
istry of Innovation and Science for Proyect CTQ2014-52939R.
48704883.
[28] M. Baek, M.K. Kim, H.J. Cho, J.A. Lee, J. Yu, H.E. Chung, et al., Factors inuencing
References the cytotoxicity of zinc oxide nanoparticles: particle size and surface charge,
J. Phys. Conf. Ser. 304 (2011) 012044.
[29] M. Merhi, C.Y. Dombu, A. Brient, J. Chang, F. Le Curieux, D. Marzin, et al., Study
[1] K. Donaldson, V. Stone, C.L. Tran, W. Kreyling, P.J.A. Borm, Nanotoxicology, of serum interaction with cationic nanoparticle: implication for in vitro
Occup. Environ. Med. 61 (2004) 727728. endocytosis, cytotoxicity and genotoxicity, Int. J. Pharm. 423 (2012) 3744.
[2] S.W. Shin, I.H. Song, S.H. Um, Role of physicochemical properties in [30] F. Seitz, R.R. Rosenfeldt, S. Schneider, R. Schulz, M. Bundschuh, Size-, surface-
nanoparticle toxicity, Nanomaterials 5 (2015) 13511365. and crystalline structure composition-related effects of titanium dioxide
[3] F. Zhao, Y. Zhao, Y. Liu, X. Chang, C. Chen, Y. Zhao, Cellular uptake, intracellular nanoparticles during their aquatic life cycle, Sci. Total Environ. 493 (2014)
tracking, and cytotoxicity of nanomaterials, Small 7 (2011) 13221337. 891897.
170 E. Caballero-Daz, M. Valcrcel Cases / Trends in Analytical Chemistry 84 (2016) 160171

[31] V. Iswarya, M. Bhuvaneshwari, S.A. Alex, S. Iyer, G. Chaudhuri, P.T. [59] Y.-X. Yang, Z.-M. Song, B. Cheng, K. Xiang, X.-X. Chen, J.-H. Li, et al., Evaluation
Chandrasekaran, et al., Combined toxicity of two crystalline phases (anatase of the toxicity of food additive silica nanoparticles on gastrointestinal cells,
and rutile) of titania nanoparticles towards freshwater microalgae: chlorella J. Appl. Toxicol. 34 (2014) 424435.
sp, Aquat. Toxicol. 161 (2015) 154169. [60] J.-H. Kim, J. Sanetuntikul, S. Shanmugam, E. Kim, Necrotic cell death caused
[32] D. Bartczak, P. Vincent, H. Goenaga-Infante, Determination of size- and by exposure to graphitic carbon-coated magnetic nanoparticles, J. Biomed.
number-based concentration of silica nanoparticles in a complex biological Mater. Res. A 103 (2015) 28752887.
matrix by online techniques, Anal. Chem. 87 (2015) 54825485. [61] R. Wahab, M.A. Siddiqui, Q. Saquib, S. Dwivedi, J. Ahmad, J. Musarrat, et al.,
[33] N. Tripathy, T.-K. Hong, K.-T. Ha, H.-S. Jeong, Y.-B. Hahn, Effect of ZnO ZnO nanoparticles induced oxidative stress and apoptosis in HepG2 and MCF-7
nanoparticles aggregation on the toxicity in RAW 264.7 murine macrophage, cancer cells and their antibacterial activity, Colloid. Surface. B 117 (2014)
J. Hazard. Mater. 270 (2014) 110117. 267276.
[34] N. Nafee, M. Schneider, U.F. Schaefer, C.-M. Lehr, Relevance of the colloidal [62] W. Hu, S. Culloty, S. Lynch, J. Davenport, S. Ramirez-Garcia, K. Dawson, et al.,
stability of chitosan/PLGA nanoparticles on their cytotoxicity prole, Int. J. Neutral red retention time assay in determination of toxicity of nanoparticles,
Pharm. 381 (2009) 130139. Mar. Environ. Res. 111 (2015) 158161.
[35] P.J. Chueh, R.-Y. Liang, Y.-H. Lee, Z.-M. Zheng, S.-M. Chuang, Different cytotoxic [63] X. Jing, J.H. Park, T.M. Peters, P.S. Thorne, Toxicity of copper oxide nanoparticles
effects of gold nanoparticles in different mammalian cell lines, J. Hazard. Mater. in lung epithelial cells exposed at the air-liquid interface compared with in
264 (2014) 303312. vivo assessment, Toxicol. In Vitro 29 (2015) 502511.
[36] I.-Y. Kim, E. Joachim, H. Choi, K. Kim, Toxicity of silica nanoparticles depends [64] M.S.P. Boyles, T. Kristl, A. Andosch, M. Zimmermann, N. Trac, E. Casals, et al.,
on size, dose, and cell type, J. Nanomed. Nanotechnol. 11 (2015) 14071416. Chitosan functionalisation of gold nanoparticles encourages particle uptake
[37] B.B. Manshian, A. Al-Ali, J.S. Jenkins, S.H. Doak, Cell type-dependent changes and induces cytotoxicity and pro-inammatory conditions in phagocytic cells,
in CdSe/ZnS quantum dot uptake and toxic endpoints, Toxicol. Sci. 144 (2015) as well as enhancing particle interactions with serum components, J.
246258. Nanobiotechnol. 13 (2015) 84.
[38] J.L. Luque-Garcia, R. Sanchez-Daz, I. Lopez-Heras, P. Martin, C. Camara, [65] F. Sambale, S. Wagner, F. Stahl, R.R. Khaydarov, T. Scheper, D. Bahnemann,
Bioanalytical strategies for in-vitro and in-vivo evaluation of the toxicity Investigations of the toxic effect of silver nanoparticles on mammalian cell
induced by metallic nanoparticles, Trends Analyt. Chem. 43 (2013) 254268. lines, J. Nanomater. 2015 (2015) 136765.
[39] E. Caballero-Daz, M. Valcrcel, Toxicity of gold nanoparticles, in: M. Valcrcel, [66] L. Wei, J. Tang, Z. Zhang, Y. Chen, G. Zhou, T. Xi, Investigation of the cytotoxicity
A.I. Lpez-Lorente (Editors), Gold nanoparticles in analytical chemistry, mechanism of silver nanoparticles in vitro, Biomed. Mater. 5 (2010) 044103.
Elsevier, Netherlands, 2014, pp. 207254. [67] K. Solarska, A. Gajewska, G. Bartosz, K. Mitura, Induction of apoptosis in human
[40] K.K. Awasthi, R. Verma, A. Awasthi, K. Awasthi, I. Soni, P.J. John, In vivo endothelial cells by nanodiamond particles, J. Nanosci. Nanotechnol. 12 (2012)
genotoxic assessment of silver nanoparticles in liver cells of Swiss albino mice 51175121.
using comet assay, Adv. Mater. Lett. 6 (2015) 187193. [68] W. Chunyan, S. Valiyaveettil, Correlation of biocapping agents with cytotoxic
[41] M. Khan, A.H. Naqvi, M. Ahmad, Comparative study of the cytotoxic and effects of silver nanoparticles on human tumor cells, RSC Adv. 3 (2013) 14329.
genotoxic potentials of zinc oxide and titanium dioxide nanoparticles, Toxicol. [69] L.A. Austin, S. Ahmad, B. Kang, K.R. Rommel, M. Mahmoud, M.E. Peek, et al.,
Rep. 2 (2015) 765774. Cytotoxic effects of cytoplasmic-targeted and nuclear-taretes gold and silver
[42] K.K. Awasthi, A. Awasthi, R. Verma, N. Kumar, P. Roy, K. Awasthi, et al., nanoparticles in HSC-3 cells-A mechanistic study, Toxicol. In Vitro 29 (2015)
Cytotoxicity, genotoxicity and alteration of cellular antioxidant enzymes in 694705.
silver nanoparticles exposed CHO cells, RSC Adv. 5 (2015) 3492734935. [70] W.-X. Duan, M.-D. He, L. Mao, F.-H. Qian, Y.-M. Li, H.-F. Pi, et al., NiO
[43] S.L. Montes-Fonseca, E. Orrantia-Borunda, A. Aguilar-Elguezabal, C. nanoparticles induce apoptosis through repressing SIRT1 in human bronchial
Gonzlez-Horta, P. Talams-Rohana, B. Snchez-Ramrez, Cytotoxicity of epithelial cells, Toxicol. Appl. Pharm. 286 (2015) 8091.
functionalized carbon nanotubes in J774A macrophages, J. Nanomed. [71] M. Sathishkumar, S. Pavagadhi, A. Mahadevan, R. Balasubramanian,
Nanotechnol. 8 (2012) 853859. Biosynthesis of gold nanoparticles and related cytotoxicity evaluation using
[44] V. Sharma, P. Singh, A.K. Pandey, A. Dhawan, Induction of oxidative stress, DNA A549 cells, Ecotoxicol. Environ. Saf. 114 (2015) 232240.
damage and apoptosis in mouse liver after sub-acute oral exposure to zinc [72] I.-L. Hsiao, Y.-J. Huang, Effects of various physicochemical characteristics on
oxide nanoparticles, Mutat. Res-Gen. Tox. En. 745 (2012) 8491. the toxicities of ZnO and TiO2 nanoparticles toward human lung epithelial
[45] L. Huo, R. Chen, L. Zhao, X. Shi, R. Bai, D. Long, et al., Silver nanoparticles activate cells, Sci. Total Environ. 409 (2011) 12191228.
endoplasmic reticulum stress signaling pathway in cell and mouse models: [73] M.S. Stan, I. Memet, C. Sima, T. Popescu, V.S. Teodorescu, A. Hermenean, A.
the role in toxicity evaluation, Biomaterials 61 (2015) 307315. Dinischiotu, Si/SiO2 quantum dots cause cytotoxicity in lung cells through redox
[46] P.V. Asharani, G.L.K. Mun, M.P. Hande, S. Valiyaveettil, Cytotoxicity and homeostasis imbalance, Chem. Biol. Interact. 220 (2014) 102115.
genotoxicity of silver nanoparticles in human cells, ACS Nano 3 (2009) [74] A.R. Lupu, T. Popescu, The noncellular reduction of MTT tetrazolium salt by
279290. TiO2 nanoparticles and its implications for cytotoxicity assays, Toxicol. In Vitro
[47] C.-T. Ng, L.Y.L. Yung, H.L.-F. Swa, R.W.-Y. Poh, J. Gunaratne, B.H. Bay, Altered 27 (2013) 14451450.
protein expression prole associated with phenotypic changes in lung [75] A. Kroll, D. Hahn, M.H. Pillukat, J. Schnekenburger, Interference of engineered
broblasts co-cultured with gold nanoparticle treated small airway epithelial nanoparticles with in vitro toxicity assays, Arch. Toxicol. 86 (2012) 11231136.
cells, Biomaterials 39 (2015) 3138. [76] A.R. Gliga, S. Skoglund, I.O. Wallinder, B. Fadeel, H.L. Karlsson, Size-dependent
[48] P.P. Fu, Q. Xia, H.-M. Hwang, P.C. Ray, H. Yu, Mechanisms of nanotoxicity: cytotoxicity of silver nanoparticles in human lung cells: the role of cellular
generation of reactive oxygen species, J. Food Drug Anal. 22 (2014) 6475. uptake, agglomeration and Ag release, Part. Fibre Toxicol. 11 (2014) 11.
[49] M.-L. Kung, S.-L. Hsieh, C.-C. Wu, T.-H. Chu, Y.-C. Lin, B.-W. Yeh, et al., Enhanced [77] Y. Huang, X. L, Y. Qu, Y. Yang, S. Wu, MicroRNA sequencing and molecular
reactive oxygen species overexpression by CuO nanoparticles in poorly mechanisms analysis of the effects of gold nanoparticles on human dermal
differentiated hepatocellular carcinoma cells, Nanoscale 7 (2015) 18201829. broblasts, Biomaterials 37 (2015) 1324.
[50] A. Dubey, M. Goswami, K. Yadav, D. Chaudhary, Oxidative stress and nano- [78] B. Mostaghaci, J. Susewind, G. Kickelbick, C.-M. Lehr, B. Loretz, Transfection
toxicity induced by TiO2 and ZnO on WAG cell line, PLoS ONE 10 (2015) system of amino-functionalized calcium phosphate nanoparticles: in vitro
e0127493. ecacy, biodegradability, and immunogenicity study, ACS Appl. Mater.
[51] J. Ahmad, M. Ahamed, M.J. Akhtar, S.A. Alkokayan, M.A. Siddiqui, J. Musarrat, Interfaces 7 (2015) 51245133.
et al., Apoptosis induction by silica nanoparticles mediated through reactive [79] V. Escamilla-Rivera, M. Uribe-Ramrez, S. Gonzlez-Pozos, O. Lozano, S. Lucas,
oxygen species in human liver cell line HepG2, Toxicol. Appl. Pharm. 259 (2012) A. De Vizcaya-Ruis, Protein corona acts as a protective shield against Fe3O4-PEG
160168. inammation and ROS-induced toxicity in human macrophages, Toxicol. Lett.
[52] M. Ahamed, R. Posgai, T.J. Gorey, M. Nielsen, S.M. Hussain, J.J. Rowe, Silver 240 (2016) 172184.
nanoparticles induced heat shock protein 70, oxidative stress and apoptosis [80] T. Tong, C.T.T. Binh, J.J. Kelly, J.-F. Gaillard, K.A. Gray, Cytotoxicity of commercial
in Drosophila melanogaster, Toxicol. Appl. Pharm. 242 (2010) 263269. nano-TiO2 to Escherichia coli assessed by high-throughput screening: effects
[53] J.E. Choi, S. Kim, J.H. Ahn, P. Youn, J.S. Kang, K. Park, et al., Induction of oxidative of environmental factors, Water Res. 47 (2013) 23522362.
stress and apoptosis by silver nanoparticles in the liver of adult zebrash, [81] S. Lin, Y. Zhao, Z. Ji, J. Ear, C.H. Chang, H. Zhang, et al., Zebrash high-throughput
Aquat. Toxicol. 100 (2010) 151159. screening to study the impact of dissolvable metal oxide NPs on the hatching
[54] W. Gao, K. Xu, L. Ji, B. Tang, Effect of gold nanoparticles on glutathione enzyme, ZHE1, Small 9 (2013) 17761785.
depletion-induced hydrogen peroxide generation and apoptosis in HL7702 [82] S. George, T. Xia, R. Rallo, Y. Zhao, Z. Ji, S. Lin, et al., Use of a high-throughput
cells, Toxicol. Lett. 205 (2011) 8695. screening approach coupled with in vivo zebrash embryo screening to
[55] S.A. Love, M.A. Maurer-Jones, J.W. Thompson, Y.-S. Lin, C.L. Haynes, Assessing develop hazard ranking for engineered nanomaterials, ACS Nano 5 (2011)
nanoparticle toxicity, Annu. Rev. Anal. Chem. (Palo Alto Calif) 5 (2012) 181 18051817.
205. [83] E. Jan, S.J. Byrne, M. Cuddihy, A.M. Davies, Y. Volkov, Y.K. Gunko, et al.,
[56] A.R. Wielgus, B. Zhao, C.F. Chignell, D.-N. Hu, J.E. Roberts, Phototoxicity and High-content screening as a universal tool for ngerprinting of cytotoxicity
cytotoxicity of fullerol in human retinal pigment epithelial cells, Toxicol. Appl. of nanoparticles, ACS Nano 2 (2008) 928938.
Pharm. 242 (2010) 7990. [84] S.-K. Jung, X. Qu, B. Aleman-Meza, T. Wang, C. Riepe, Z. Liu, et al., A multi-
[57] M.S. Acar, Z.B. Bulut, A. Ates, B. Nami, N. Koak, B. Yildiz, Titanium dioxide endpoint, high-throughput study of nanomaterial toxicity in Caenorhabditis
nanoparticles induce cytotoxicity and reduce mitotic index in human amniotic elegans, Environ. Sci. Technol. 49 (2015) 24772485.
uid-derived cells, Hum. Exp. Toxicol. 34 (2015) 7482. [85] R. Klaper, D. Arndt, J. Bozich, G. Dominguez, Molecular interactions of
[58] J.A. Kim, C. Aberg, G. de Crcer, M. Malumbres, A. Salvati, K.A. Dawson, Low nanomaterials and organisms: dening biomarkers for toxicity and high-
dose of amino-modied nanoparticles induces cell cycle arrest, ACS Nano 7 throughput screening using traditional and next-generation sequencing
(2013) 74837494. approaches, Analyst 139 (2014) 882895.
E. Caballero-Daz, M. Valcrcel Cases / Trends in Analytical Chemistry 84 (2016) 160171 171

[86] G. Gao, Y. Ze, B. Li, X. Zhao, T. Zhang, L. Sheng, et al., Ovarian dysfunction and [105] X. He, Y. Ma, M. Li, P. Zhang, Y. Li, Z. Zhang, Quantifying and imaging engineered
gene-expressed characteristics of female mice caused by long-term exposure nanomaterials in vivo: challenges and techniques, Small 9 (2013) 1482
to titanium dioxide nanoparticles, J. Hazard. Mater. 243 (2012) 1927. 1491.
[87] H.C. Poynton, H.J. Allen, J.M. Lazorchak, A. Loguinov, C.A. Impelliteri, J.L. [106] X. Huang, L. Li, T. Liu, N. Hao, H. Liu, D. Chen, et al., The shape effect of
Heckman, et al., Toxicogenomic responses of nanotoxicity in daphnia magna mesoporous silica nanoparticles on biodistribution, clearance, and
exposed to silver nitrate and coated silver nanoparticles, Environ. Sci. Technol. biocompatibility in vivo, ACS Nano 5 (2011) 53905399.
46 (2012) 62886296. [107] G. Xie, C. Wang, J. Sun, G. Zhong, Tissue distribution and excretion of
[88] S.I.L. Gomes, S.C. Novais, J.J. Scott-Fordsmand, W. De Coen, A.M.V.M. Soares, intravenously administered titanium dioxide nanoparticles, Toxicol. Lett. 205
M.J.B. Amorim, Effect of Cu-nanoparticles versus Cu-salt in Enchytraeus albidus (2011) 5561.
(Oligochaeta): differential gene expression through microarray analysis, Comp. [108] C.P. Vignardi, F.M. Hasue, P.V. Sartorio, C.M. Cardoso, A.S.D. Machado, T.C.A.
Biochem. Physiol. C Toxicol. Pharmacol. 155 (2012) 219227. Santos, et al., Genotoxicity, potential cytotoxicity and cell uptake of titanium
[89] T. Coccini, E. Roda, M. Fabbri, M.G. Sacco, L. Gribaldo, L. Manzo, Gene expression dioxide nanoparticles in the marine sh Trachinotus carolinus (Linnaeus, 1766),
proling in rat kidney after intratracheal exposure to cadmium-doped Aquat. Toxicol. 158 (2015) 218229.
nanoparticles, J. Nanopart. Res. 14 (2012) 110. [109] D.S. Anderson, E.S. Patchin, R.M. Silva, D.L. Uyeminami, A. Sharmah, T. Guo,
[90] Y. Qin, Z.-W. Zhou, S.-T. Pan, Z.-X. He, X. Zhang, J.-X. Qiu, et al., Graphene et al., Inuence of particle size on persistence and clearance of aerosolized
quantum dots induce apoptosis, autophagy, and inammatory response via silver nanopartices in the rat lung, Toxicol. Sci. 144 (2015) 366381.
p38 mitogen-activated protein kinase and nuclear factor-kB mediated signaling [110] P. Prabu, W.S. Vedakumari, T.P. Sastry, Time-dependent biodistribution,
pathways in activated THP-1 macrophages, Toxicology 327 (2015) 6276. clearance and biocompatibility of magnetic brin nanoparticles: an in vivo
[91] Y. Liu, J. Liang, Q. Wang, Y. He, Y. Chen, Copper nanoclusters trigger muscle study, Nanoscale 7 (2015) 96769685.
cell apoptosis and atrophy in vitro and in vivo, J. Appl. Toxicol. (2015) Article [111] C.-M. Zhao, W.-X. Wang, Biokinetic uptake and eux of silver nanoparticles
in press. in Daphnia magna, Environ. Sci. Technol. 44 (2010) 76997704.
[92] Y. Liu, Y. Zhang, W. Jia, Y. Cheng, T. Chen, X. Wang, et al., GC/TOFMS analysis [112] X. Zhang, J. Yin, C. Kang, J. Li, Y. Zhu, W. Li, et al., Biodistribution and toxicity
of endogenous metabolites in mouse broblast cells and its application in TiO2 of nanodiamonds in mice after intratracheal instillation, Toxicol. Lett. 198
nanoparticle-induced cytotoxicity study, Chromatographia 75 (2012) 1301 (2010) 237243.
1310. [113] B. Li, X.Y. Zhang, J.Z. Yang, Y.J. Zhang, W.X. Li, C.H. Fan, et al., Inuence of
[93] M. Lv, W. Huang, Z. Chen, H. Jiang, J. Chen, Y. Tian, et al., Metabolomics polyethylene glycol coating on biodistribution and toxicity of nanoscale
techniques for nanotoxicity investigations, Bioanalysis 7 (2015) 15271544. graphene oxide in mice after intravenous injection, Int. J. Nanomed. 9 (2014)
[94] R. Lei, C. Wu, B. Yang, H. Ma, C. Shi, Q. Wang, et al., Integrated metabolomic 46974707.
analysis of the nano-sized copper particle-induced hepatotoxicity and [114] Y.-F. Li, J. Zhao, Y. Qu, Y. Gao, Z. Guo, Z. Liu, et al., Synchrotron radiation
nephrotoxicity in rats: a rapid in vivo screening method for nanotoxicity, techniques for nanotoxicology, J. Nanomed. Nanotechnol. 11 (2015) 1531
Toxicol. Appl. Pharm. 232 (2008) 292301. 1549.
[95] M. Tang, T. Zhang, Y. Xue, S. Wang, M. Huang, Y. Yang, et al., Dose dependent [115] B. Wang, Z. Wang, W. Feng, M. Wang, Z. Hu, Z. Chai, et al., New methods for
in vivo metabolic characteristics of titanium dioxide nanoparticles, J. Nanosci. nanotoxicology: synchrotron radiation-based techniques, Anal. Bioanal. Chem.
Nanotechnol. 10 (2010) 85758583. 398 (2010) 667676.
[96] S.-M. Huang, X. Zuo, J.J. Li, S.F.Y. Li, B.H. Bay, C.N. Ong, Metabolomics studies [116] Y. Qu, W. Li, Y. Zhou, X. Liu, L. Zhang, L. Wang, et al., Full assessment of fate
show dose-dependent toxicity induced by SiO2 nanoparticles in MRC-5 human and physiological behaviour of quantum dots utilizing Caenorhabditis elegans
fetal lung broblasts, Adv. Healthc. Mater. 1 (2012) 779784. as a model organism, Nano Lett. 11 (2011) 31743183.
[97] R.E. zel, X. Liu, R.S.J. Alkasir, S. Andreescu, Electrochemical methods for [117] J. Zhang, B. Li, Y. Zhang, A. Li, X. Yu, Q. Huang, et al., Synchrotron radiation
nanotoxicity assessment, Trends Analyt. Chem. 59 (2014) 112120. X-ray uorescence analysis of biodistribution and pulmonary toxicity of
[98] R.E. zel, R.S.J. Alkasir, K. Ray, K.N. Wallace, S. Andreescu, Comparative nanoscale titanium dioxide in mice, Analyst 138 (2013) 65116516.
evaluation of intestinal nitric oxide in embryonic zebrash exposed to metal [118] L. Wang, J. Li, J. Pan, X. Jiang, Y. Ji, Y. Li, et al., Revealing the binding structure
oxide nanoparticles, Small 9 (2013) 42504261. of the protein corona on gold nanorods using synchrotron radiation-based
[99] S.A. Love, Z. Liu, C.L. Haynes, Examining changes in cellular communication techniques: understanding the reduced damage in cell membranes, J. Am.
in neuroendocrine cells after noble metal nanoparticle exposure, Analyst 137 Chem. Soc. 135 (2013) 1735917368.
(2012) 3004. [119] Z. Khatun, M. Nurunnabi, D.Y. Lee, Y.-J. Kim, Y. Byun, K.J. Cho, et al., Optical
[100] M.A. Maurer-Jones, J.R. Christenson, C.L. Haynes, TiO2 nanoparticle-induced imaging, biodistribution and toxicity of orally administered quantum dots
ROS correlates with modulated immune cell function, J. Nanopart. Res. 14 loaded heparin-deoxycholic acid, Macromol. Res. 23 (2015) 686695.
(2012) 1291. [120] L. Song, M.G. Vijver, W.J.G.M. Peijnenburg, T.S. Galloway, C.R. Tyler, A
[101] L. Otero-Gonzlez, R. Sierra-Alvarez, S. Boitano, J.A. Field, Application and comparative analysis on the in vivo toxicity of copper nanoparticles in three
validation of an impedance-based real time cell analyzer to measure the species of freshwater sh, Chemosphere 139 (2015) 181189.
toxicity of nanoparticles impacting human bronchial epithelial cells, Environ. [121] B.J. Marquis, S.A. Love, K.L. Braun, C.L. Haynes, Analytical methods to assess
Sci. Technol. 46 (2012) 1027110278. nanoparticle toxicity, Analyst 134 (2009) 425439.
[102] X. Zhu, E. Hondroulis, W. Liu, C.-Z. Li, Biosensing approaches for rapid [122] J.M. Jenkins, D.L. Halaney, K.V. Sokoloc, L.L. Ma, H.J. Shipley, S. Mahajan, et al.,
genotoxicity and cytotoxicity assays upon nanomaterial exposure, Small 9 Excretion and toxicity of gold-iron nanoparticles, J. Nanomed. Nanotechnol.
(2013) 18211830. 9 (2013) 356365.
[103] A. Pandey, R.S. Chouhan, Y. Gurbuz, J.H. Niazi, A. Qureshi, S. cerevisiae [123] C. Fu, T. Liu, L. Li, H. Liu, D. Chen, F. Tang, The absorption, distribution, excretion
whole-cell based capacitive biochip for the detection of toxicity of different and toxicity of mesoporous silica nanoparticles in mice following different
forms of carbon nanotubes, Sensor. Actuat. B-Chem. 218 (2015) 253260. exposure routes, Biomaterials 34 (2013) 25652575.
[104] E. Hondroulis, Z. Zhang, C. Chen, C.Z. Li, Impedance based nanotoxicity [124] H. Yang, L. Du, X. Tian, Z. Fan, C. Sun, Y. Liu, et al., Effects of nanoparticle size
assessment of graphene nanomaterials at the cellular and tissue level, Anal. and gestational age on maternal biodistribution and toxicity of gold
Lett. 45 (2012) 272282. nanoparticles in pregnant mice, Toxicol. Lett. 230 (2014) 1018.

You might also like