PreprintPDF Available

Genotoxic Impact of Aluminum-containing Nanomaterials in Human Intestinal and Hepatic Cells.

Authors:
  • University Rennes1

Abstract and Figures

Background: Exposure of consumers to aluminum-containing nanomaterials (Al NMs) through numerous products is an area of concern for public health agencies since human health risks are not completely elucidated. In addition, the available data on the genotoxicity of Al2O3 and Al⁰ NMs are inconclusive or rare. In order to provide further information, the present study investigated the in vitro genotoxic potential of Al⁰ and Al2O3 NMs in intestinal and liver cell models since these tissues represent organs which would be in direct contact or could experience potential accumulation following oral exposure. Methods: Differentiated human intestinal Caco-2 and hepatic HepaRG cells were exposed to Al⁰ and Al2O3 NMs (0.03 to 80 µg/cm2) and the results were compared with those obtained with the ionic form AlCl3. Several methods, including H2AX labelling, the alkaline comet assay and micronucleus (MN) assays were used. Oxidative stress and oxidative DNA damage were assessed using High Content Analysis (HCA) and the formamidopyrimidine DNA-glycosylase -modified comet assay respectively. Moreover, carcinogenic properties of Al NMs were investigated through the cell transforming assay (CTA) in Bhas 42 cells. Results: The three forms of Al did not induce chromosomal damage when tested in the MN assay. Furthermore, no cell transformation was observed in Bhas 42 cells. However, although no production of oxidative stress was detected in HCA assays, Al2O3 NMs induced oxidative DNA damage in Caco-2 cells in the comet assay following a 24 h treatment. Considerable DNA damage was observed with Al⁰ NMs in both cell lines in the comet assay, although this was likely due to interference with these NMs. Finally, no genotoxic effects were observed with AlCl3. Conclusion: The slight effects observed with Al NMs are therefore not likely to be related to ion release in the cell media.
Content may be subject to copyright.
Page 1/39
Genotoxic Impact of Aluminum-containing
Nanomaterials in Human Intestinal and Hepatic
Cells.
Pégah Jalili
Anses Laboratoire de Fougeres
Sylvie HUET
Anses Laboratoire de Fougeres
Agnès Burel
MRiC
Benjamin-Christoph Krause
Bundesinstitut fur Risikobewertung
Caroline Fontana
Institut National de Recherche et de Securite
Soizic Chevance
Institut des Sciences Chimiques de Rennes
Fabienne Gauffre
Institut des Sciences Chimiques de Rennes
Yves Guichard
Institut National de Recherche et de Securite
Alfonso Lampen
Bundesinstitut fur Risikobewertung
Peter Laux
Bundesinstitut fur Risikobewertung
Andreas Luch
Bundesinstitut fur Risikobewertung
Kevin Hogeveen
Anses Laboratoire de Fougeres
Valerie Fessard ( Valerie.FESSARD@anses.fr )
Anses Laboratoire de Fougeres https://orcid.org/0000-0001-9046-9117
Research
Keywords: nanomaterials, comet assay, micronucleus, cell transformation assay, oxidative stress
Page 2/39
DOI: https://doi.org/10.21203/rs.3.rs-72845/v1
License: This work is licensed under a Creative Commons Attribution 4.0 International License. 
Read Full License
Page 3/39
Abstract
Background: Exposure of consumers to aluminum-containing nanomaterials (Al NMs) through numerous
products is an area of concern for public health agencies since human health risks are not completely
elucidated. In addition, the available data on the genotoxicity of Al2O3 and Al0 NMs are inconclusive or
rare. In order to provide further information, the present study investigated the
in vitro
genotoxic potential
of Al0 and Al2O3 NMs in intestinal and liver cell models since these tissues represent organs which would
be in direct contact or could experience potential accumulation following oral exposure.
Methods: Differentiated human intestinal Caco-2 and hepatic HepaRG cells were exposed to Al0 and
Al2O3 NMs (0.03 to 80 µg/cm2) and the results were compared with those obtained with the ionic form
AlCl3. Several methods, including H2AX labelling, the alkaline comet assay and micronucleus (MN)
assays were used. Oxidative stress and oxidative DNA damage were assessed using High Content
Analysis (HCA) and the formamidopyrimidine DNA-glycosylase -modied comet assay respectively.
Moreover, carcinogenic properties of Al NMs were investigated through the cell transforming assay (CTA)
in Bhas 42 cells.
Results: The three forms of Al did not induce chromosomal damage when tested in the MN assay.
Furthermore, no cell transformation was observed in Bhas 42 cells. However, although no production of
oxidative stress was detected in HCA assays, Al2O3 NMs induced oxidative DNA damage in Caco-2 cells
in the comet assay following a 24 h treatment. Considerable DNA damage was observed with Al0 NMs in
both cell lines in the comet assay, although this was likely due to interference with these NMs. Finally, no
genotoxic effects were observed with AlCl3.
Conclusion: The slight effects observed with Al NMs are therefore not likely to be related to ion release in
the cell media.
1. Introduction
Within the last decade, aluminum (Al)-containing nanomaterials (NMs) have been widely used not only
for industrial applications, but also in consumer products, due to their higher reactivity compared to the
bulk form [1, 2, 3, 4]. Forms of Al, both in the micro-and the nano-size, are present in food and consumer
products [1, 5] due to their use as rming, anticaking, neutralizing, emulsifying and texturizing agents, as
well as for cooking tools [6], waste water treatment [7, 8] and in medical and hygiene products such as
toothpaste [9, 10, 11]. Nevertheless, their potential toxicity has not been fully evaluated, leading to major
concerns from consumers and public health agencies [12].
According to exposure estimates from the European Food Safety Authority (EFSA), consumers can
absorb up to 2.3 mg Al /kg bw/week, more than twice the weekly tolerable intake (1 mg/kg bw/week) [7].
In addition, a recent study has estimated total consumer exposure to Al containing compounds, including
Page 4/39
contributions from products used in food (additives, contact materials) and in cosmetics, and concluded
that adolescents were highly exposed [13].
Few studies on the genotoxicity of nanoscale forms of Al following oral ingestion have been performed,
and most of the published literature has focused on Al2O3 NMs only. DNA damage was reported in
erythrocytes of rats after a single oral treatment with Al2O3 NMs, although at high doses (1,000 mg/kg)
[14, 15]. Genotoxic effects were observed in bone marrow, but not in other organs, after a short-term
treatement with lower doses of Al2O3 NMs [16].
In vivo
effects of Al0 NMs following oral exposure are
mostly lacking, although one study suggested cross-linking effects on DNA in the duodenum of rats [16].
Following oral exposure of rodents with ionic forms of Al, an increase in MN frequency was reported in
bone marrow after a single oral administration [17] and in liver after a 30 day oral treatment [18].
Nevertheless, the induction of MN formation in liver was shown to decrease with an antioxidant treatment
[18, 19]. Consistent with these results, a slight oxidative DNA damage was observed in blood after a short-
term oral exposure [16].
The
in vitro
genotoxicity of Al2O3 NMs has been assessed in several mammalian cell lines including
human peripheral lymphocytes [20], primary human broblasts [21], hepatic HepG2 cells [22], and
Chinese hamster ovary cells [23]. While some studies have not observed genotoxic effects of Al2O3 NMs
[20, 24, 25, 26], others have reported a positive response [21, 22] which may be associated with oxidative
damage [22]. In contrast, no data on the
in vitro
genotoxicity of Al0 NMs has been published so far, and
only some cytotoxicity was detected in rat alveolar macrophages treated with Al0 above 100 µg/ml [27].
For the salt AlCl3, DNA damage has been reported in human peripheral blood lymphocytes, with positive
results in micronucleus and chromosomal aberration tests, as well as in the comet assay [17, 28, 29, 30].
According to an ECHA safety assessment [31], the data available on the genotoxicity of Al2O3 NMs are
inconclusive while few data on the genotoxicity of Al0 NMs has been published so far. In addition to the
direct contact of Al NMs present in food with the intestinal epithelium, Al accumulation in liver has been
shown after oral exposure with Al2O3 NMs [15, 32, 33].
Therefore, the aim of the current study was to evaluate the
in vitro
genotoxic potential of Al0 and Al2O3
NMs in two relevant human cell models of intestine and liver. Several endpoints of genotoxicity were
investigated using the alkaline and Fpg-modied comet assays which detects DNA breakage including
oxidative lesions, DNA double strand breaks were detected through phosphorylated histone H2AX
(γH2AX), and the micronucleus assay which determines chromosome and genome damage.
Furthermore, the capacity of aluminum-containing NMs to initiate or promote carcinogenesis was
assessed by the Cell Transforming Assay (CTA) in Bhas-42 cells.
As these NMs can potentially dissolve in the dispersion solution or in media, the genotoxicity was
compared to that of the metal salt AlCl3. Moreover, the interference of NMs, including with Al-NMs [34],
has been demonstrated in numerous publications using a wide range of biological assays, and stresses
Page 5/39
the necessity to evaluate interference in order to assess the potential effect on the results [35, 36, 37, 38].
In this study, various sources of interference have been taken into account within the different assays.
2. Materials And Methods
2.1 Chemicals and reagents
Dimethylsulfoxide (DMSO), insulin, cytochalasin B, formamidopyrimidine-DNA glycosylase (Fpg), trypan-
blue, 12-
O
-tetradecanoylphorbol-13-acetate (TPA), 3-methylcholanthrene (3-MCA) and menadione (MEN)
were supplied from Sigma (St. Quentin-Fallavier, France). Methylmethanesulfonate (MMS) was
purchased by Acros Organics (Fairlawn, NJ). Dinophysistoxin-2 (DTX-2) was from the National Research
Council Canada (NRCC, Ottawa, Canada). Penicillin, streptomycin, Williams' E medium and Fetal Bovine
Serum Fetalclone II (FBS) were supplied from Invitrogen Corporation (Illkirch, France). For Bhas 42 cell
cultures, Eagle’s minimum essential medium and Dulbecco’s modied Eagle’s medium/Ham’s F12 was
from Invitrogen Corporation (Illkirch, France). Fetal bovine serum was obtained by Dutscher, (Brumath
France). Hydrocortisone hemisuccinate, HycloneTM DMEM/high glucose and fetal bovine serum for Caco-
2 cells were purchased from Upjohn Pharmacia (Guyancourt, France), GE Healthcare Life Science (Logan,
UT, USA) and Capricorn scientic (Ebsdorfergrund, Germany), respectively. The primary and secondary
antibodies (mouse monoclonal anti γH2AX ser139 (ab26350), rabbit monoclonal anti active caspase-3
antibody (ab13847), goat anti-rabbit IgG H&L AlexaFluor 647 (ab150079) and goat anti-mouse IgG H&L
AlexaFluor 647 (ab150115)) were provided from Abcam (Cambridge, UK). CellROX® Deep Red Reagent
was obtained from Invitrogen (Paisley, UK). Formaldehyde and Giemsa were purchased by Fisher (Illkirch-
Graffenstaden, France).
2.2 Dispersion and characterization of NMs
Al0, Al2O3 and ZnO NMs with a similar primary particle size were supplied from IoLiTec (Heilbronn,
Germany). NM characteristics as provided by the supplier are presented in Table 1. AlCl3 (hexahydrate)
was purchased from Sigma Aldrich (Saint Louis, USA). NM dispersion was performed according to the
NANOGENOTOX protocol [39], as described in [16].
The morphology and agglomeration of Al0 and Al2O3 NMs in the stock dispersion solution and in cell
media were determined by transmission electron microscopy (TEM) (Figure S 1). For the characterization
of NMs from stock solutions, TEM grids were prepared immediately after sonication and dilution (100
µg/mL) in the stock dispersion solution. For the characterization of NMs in cell culture media (DMEM
+10% FBS and William’s Medium +5% FBS), the samples were diluted with distilled water to 1.2 µg/mL
prior to grid preparation. The TEM grids were prepared by deposition of a carbon-coated copper grid onto
a drop of the stock solution for 20 s to allow adsorption of the NMs and were observed with an electron
microscope (JEOL 1400 operated at 120 kV and coupled with a 2k-2k camera from Gatan (Orius 1000)).
Page 6/39
The hydrodynamic diameter of Al0 and Al2O3 NMs were measured using a Malvern Zetasizer (Malvern
Instruments, Malvern, UK) equipped with a 633-nm laser diode operating at an angle of 173°. To assess
the stability of NM suspensions, following NM dispersion, samples were diluted to a nal concentration
of 100 µg/ml in the stock dispersion solution or in cell media and measurements were performed at 0
and 24 h. The samples were equilibrated at 25 °C for 120 s prior to measurement. Ten repeated
measurements for each sample were performed in 3 independent experiments. The mean hydrodynamic
diameter Zave was determined using cumulant analysis.
2.3 Cell culture and treatment
The human colorectal adenocarcinoma Caco-2 cell line was cultured (passages 25–38) until
differentiation after 21 days as described in [40] including for cell seeding in various plate formats
depending on the assay performed. Simarly, HepaRG cells (passages 13-19) were cultured and seeded for
the various assays as previously described [40, 41].
Differentiated Caco-2 and HepaRG cells were treated for 24 h with Al0 and Al2O3 NMs at concentrations
ranging from 0.03 to 80 µg/cm2 and with AlCl3 as ionic salt control at 90 and 128 µg.mL-1 in DMEM +
10% FBS or William’s medium + 5% FBS respectively. For some assays, ZnO NMs at concentrations from
1.5 to 6 µg/cm2 were used as a positive NM control. Equivalence between volume concentration (µg/mL)
and surface concentration (µg/cm2) are shown in Table S 1B. Al content corresponding to the
concentrations of Al-containing NMs and AlCl3 that were used are summarized in Table S 1B.
2.4 Kinetics of nanoparticle sedimentation
The colloidal characterization of the suspended nanomaterials in the conditions of cellular uptake assay
was achieved using the volumetric sedimentation method (VCM) as reported in DeLoid et al [42]. We rst
measured the volume of the potentially agglomerated NM in DMEM and Williams media, at a NM
concentration of 250 µg.mL-1, using a specic centrifugal tube and ruler device. From the measured pellet,
the effective density (eff) is calculated using the following equation:
Where m is the density of the medium in g.cm-3, NP is the density of NP (2.7 g.cm-3 for Al and 3.95 for
Al2O3), MNP the total mass of NM in 1 mL of dispended volume and V the measured volume pellet. SF is
a stacking factor and was set to 0.634, which generally is appropriate for random stacking. The loss of
mass of NMs from ion release was estimated to be lower than 1% and was neglected in the density
calculation. The viscosity of the cell culture media at 37°C was determined using a Nanoparticle Tracking
Page 7/39
Analysis device (Malvern Instrument) by measuring the apparent hydrodynamic radius of 400nm
standard particles in the media. Finally, the kinetics of sedimentation was calculated using the distorded
grid (DG) model available from DeLoid et al [42]. The size of the NPS was taken from Table 2 (Zave).
Other model parameters are h=3.1mm (liquid column height), initial NM concentration : 0.250mg.mL-1,
the dissolution and cell-NMs stickiness are neglected (parameters set to 0).
2.5 Ion release from NMs
Following the dispersion of Al0 and Al2O3 NMs, suspensions were diluted in stock solution (ultra pure
water + 0.05 % BSA) or cell culture media (DMEM +10% FBS and William’s Medium +5% FBS) at
concentrations of 25, 50 and 100 µg/mL. After 24 h, ion release from NMs was determined by
ultracentrifugation at 16,000g for 1h at 4°C (Hettich Zentrifuge Mikro 220R). The supernatants were
processed through acidic hydrolysis (69% HNO3, 180°C for 20 min in an MLS-ETHOS Microwave system)
before detection of Al species with a quadrupole Inductively Coupled Plasma Mass Spectrometry (ICP-
MS) (iCAP Q, Thermo Fisher Scientic GmbH, Dreieich, Germany) equipped with a PFA ST Nebulizer, a
quartz cyclonic spray chamber and a 2.5 mm quartz injector (Thermo Fisher Scientic). The gas ows
were set to 14 L/min, and 0.65 L/min for the cool gas (Ar) and the auxiliary gas (Ar) respectively. The ow
rate of the sample was 0.39 mL/min. Results are given as percentage of the initial Al amount.
2.6 Uptake observations by TEM
Following a 24 h treatment, cells were xed by glutaraldehyde (2.5%) and embedded in DMP30-epon
before cutting ultra-thin sections (90 nm) for TEM observation as described in [40].
2.7 Cellular imaging and High Content Analysis (HCA)
 After 24 h treatment with Al NMs and AlCl3, plates were processed for HCA with an ArrayScan VTI
HCS Reader (Thermo Scientic, Waltham, USA) as described in [40]. Cell numbers were determined from
DAPI staining, active caspase-3 was quantied in the total cell compartment and H2AX in cell nuclei.
Oxidative stress was measured using CellROX Deep Red Reagent (Fisher Scientic, Illkirch, France).
Briey, cells were pre-incubated for 1 h with 5 M CellROX in serum-free media and washed twice with
PBS before treatment with NMs and AlCl3. After 24 h and twice washing with PBS, cells were incubated
with 3 M Hoescht 33342 for 20 min at 37°C. Cells were then washed twice with PBS and were scanned
and analyzed using the Compartimental Analysis module of the Bioapplication software. For each well,
images from 7 elds (20 × magnication) were analyzed for quantication of uorescence at 647 nm.
Page 8/39
2.8 Comet assay
After a 5 h (Figure S 3) or 24 h treatment with Al NMs and AlCl3, the comet assay was performed as
described in [40, 43]. The individual tail intensity of at least 50 cells per slide were analyzed using the
Comet Assay IV software (Perceptive Instruments, Haverhill, UK). Cells were considered as hedgehogs
when DNA damage was too high to score. At least three independent experiments were performed. Methyl
methanesulfonate (MMS) was used as positive control.
The level of oxidized bases was determined with the modied comet assay using the bacterial DNA repair
enzyme Fpg through the formation of single-strand breaks (SSB) induced by the excision of oxidized
purines [44, 45]. Some additional steps to the protocol described above were performed such as
incubation with enzyme buffer (0.1 M KCl, 0.2 mM EDTA, 40 mM HEPES, 0.2 mg/ml BSA) after lysis.
Two slides, one incubated with enzyme buffer (control slide) and the other with 9 U/slide Fpg at 37°C for
30 min, were then processed as described previously.
2.9 Particle interaction with DNA during the comet assay
The interaction of NMs with DNA migration during the comet assay was evaluated as described
previously [35, 40]. Briey, dilutions of Al0 or Al2O3 NMs in 0.5% low-melting point agarose (LMP) were
prepared at nal concentrations of 28 and 128 µg/mL (corresponding to 9 and 40 µg/cm2 conditions).
After trypsinization and centrifugation (2 min, 136 g), untreated Caco-2 and HepaRG cells were
resuspended in the LMP/NM mixture, loaded on pre-coated slides and processed in the alkaline comet
assay as previously described, in the presence or absence of Fpg. A negative control consisting of
untreated cells in LMP-agarose in the absence of NMs was performed in order to compare the results.
2.10 Cytokinesis-block micronucleus assay (CBMN)
The CBMN assay was performed as described in [40] according to the guideline n°487 of the
Organization for Economic Co-operation and Development (OECD) [46]. After staining of the slides with
acridine orange (100 μg/mL), at least 1000 binucleated cells per slide were scored. Three independent
experiments were carried outand each concentration was tested in duplicate. The replication index (RI)
was calculated using the formula recommended by OCDE guideline n°487. MMS and ZnO NM were used
as positive controls.
2.11 Bhas 42 Cell Transformation Assay (CTA)
Originally established from the v-Ha-ras-transfected BALB/c 3T3 cells by Sasaki et al [47], Bhas 42 cells
used in this study (passage 23) were obtained from Harlan Laboratories (Rossdorf, Germany). Both the
CTA and concurrent cell growth assays were performed in their 6-well format and in accordance with a
Page 9/39
guidance document produced by the OECD [48], with some modications. The protocol, including both an
initiation and a promotion assay, was previously described by Fontana et al [49].
In the initiation assay, 24 h after seeding (420 cells/cm2) (Day 1), the cells were treated with Al NMs and
AlCl3 for 72 h (Day 4). Then, the cells were cultivated in fresh medium until Day 21, with medium changes
on Day 7, Day 10 and Day 14. MCA (1 µg/mL) was used as positive control.
In the promotion assay, the cells were seeded (1,500 cells/cm2) and cultured for 4 days without changing
the media. On Day 4, 7, and 10, the culture medium was replaced with fresh media containing Al NMs or
AlCl3. The treatment continued until Day 14. The cells were then cultured in fresh medium in the absence
of NMs until Day 21. TPA (0.05 µg/mL) was used as positive control.
In both assays, the cells were xed with ethanol on Day 21 and stained with a 5 % Giemsa solution. The
morphological criteria recommended by OECD were followed for the evaluation of transformed foci. The
mean of the number of transformed foci was calculated from six replicate wells.
Cell growth assays in both the initiation and promotion conditions were performed on Day 7 using three
replicate wells for each condition. The cells were xed in 4% formaldehyde and stained with 1 µg/mL
DAPI. The number of cell in wells was determined by automated microscopy with an Arrayscan VTi using
the Target Activation module of the BioApplication software. The relative cell growth (%) was calculated
as follows: (number of cells in treated cultures / number of cell in control cultures) x100.
2.12 Statistical analysis
The statistical signicance of HCA results was tested using one-way Analysis of variance (ANOVA)
followed by Dunnett's post-hoc tests with GraphPad Prism 5.
For the comet assay, the one-way Analysis of variance (ANOVA) was used followed by Dunnett's post-hoc
test.
For the micronucleus assay, the percentages of micronucleated cells in treated and solvent control
cultures were compared using the one-way Pearson chi-square test.
For the CTA, data were statistically analysed by multiple comparison using the one-sided Dunnett's test
(p<0.05, upper-sided). The signicance of the positive controls (MCA and TPA) was evaluated relative to
the control (p < 0.05) by the one-sided Student's t-test.
3. Results
3.1 Nanomaterial characterization
Page 10/39
Information concerning the physico-chemical characterization, including the morphology, primary size,
surface specic area (SSA), purity and density of the Al0, Al2O3 and ZnO NMs used in this study are
provided in Table 1. However, in contrast to the information provided by the suppliers, the particle
morphology of Al2O3 NMs in the stock dispersion solution cannot be considered as being spherical, but
rather have a rod-like shape when observed by TEM (Figure S1). Although Al0 particles exhibit a spherical
shape, numerous elongated protrusions are also observed (Figure S1). Therefore, values of “average
particle size” (Table 1) should then be considered with caution. Due to the drying step for preparation of
TEM grids, the crystallization of different components of the culture media did not allow a proper
characterization of the morphology of Al NMs in cell culture media (data not shown).
Particle hydrodynamic diameter and stability in the stock dispersion solution, as well as in cell media,
were assessed by DLS immediately (0 h), as well as after 24 h (Table 2). The hydrodynamic diameters of
Al0, Al2O3 and ZnO NMs in the dispersion stock solution were 254 ± 4 nm for Al0, 168 ± 3 nm for Al2O3
and 233 ± 11 nm for ZnO immediately following dispersion and were stable over time. The stability over
time of these NMs was also observed in cell media (Table 2). Nevertheless, the average hydrodynamic
size of Al0 and Al2O3 NMs were lower in DMEM + 10% FBS at any time of measurement compared to
stock solution and William’s media + 5% FBS medium. The average hydrodynamic size of ZnO NMs was
similar in the stock dispersion solution and in the two media. Globally, although estimated to have a
similar primary size, the average hydrodynamic sizes of Al2O3 NMs are consistently smaller than those
measured for Al0 and ZnO NMs.
The polydispersity index (PdI) of Al0, Al2O3 and ZnO NMs suspensions in stock dispersion solution and in
media were stable over time. Whereas the PdI was quite low (< 0.25) for all NMs in stock dispersion
solution, it increased in cell media for Al2O3 (0.52 ± 0.027 in DMEM and 0.47 ± 0.015 in William’s at 0 h)
as well as for ZnO (0.27 ± 0.010 in DMEM and 0.23 ± 0.030 in William’s at 0 h). This effect of media on
the PdI was not observed for Al0 NMs.
The sedimentation of particles during
in vitro
exposure is critical when considering interactions of cells
with NMs. Indeed, in a typical experiment the NMs are dispensed onto adherent cells in well plates.
Therefore the amount of particles in contact with cells depends on the rate of sedimentation. We applied
the dosimetry method reported by DeLoid et al [42] to evaluate the sedimentation of Al0 and Al2O3 NMs in
DMEM and Willam’s media in the conditions of cell exposure. When dispersed in culture medium, NMs
may form agglomerates with adsorbed proteins and entrapped uid. The effective density (eff) of these
agglomerates should rst be measured to determine the colloidal behavior of such agglomerates. The
effective densities determined for Al0 NMs are 1.18 and 1.19 in DMEM and William’s media respectively.
Densities of 1.24 and 1.17 were obtained for Al2O3 NMs in DMEM and William’s cell culture media
respectively. Applying the sedimentation model provided in DeLoid et al [42], we calculated the evolution
of the concentration of NMs at the surface of cells with respect to time (Figure 1). The sedimentation of
Al0 NMs was similar in both media, and after 24 h the concentration at the bottom of the well is
Page 11/39
approximately 650 µg/mL. The rate of sedimentation of Al2O3 NMs was relatively slower, and the
difference was signicantly more pronounced in William’s medium, with a deposited concentration of 450
µg/mL at the bottom of the well after 24 h.
3.2 Ion release in stock solution and cell culture media
Ion release from Al NMs was investigated using ICP-MS (Table 3) and results are presented as
percentages with respect to the initial concentration of aluminum. A decrease in the percentage of ion
release from Al0 NMs was observed with increasing NMs concentrations in both the stock dispersion
solution (1.30 % at 25 µg/mL and 0.48 % at 100 µg/mL) and in media (3.88 % to 0.95 % in DMEM, and
2.42 % to 0.68 % in William’s for 25 and 100 µg/mL respectively). Nevertheless, ion release from Al0 NMs
was slightly higher in media when compared to the dispersion stock solution. A concentration-dependent
decrease in ion release was also observed for ZnO NMs (Table 3). Ion release was also higher in media
compared to dispersion stock solution.
In contrast to Al0 and ZnO NMs, for Al2O3 NMs, the percentage of ion release with respect to the initial
concentration was very low, relatively stable and independent of the NM concentration, although ion
release was slightly higher in cell media.
The level of ions from AlCl3 solutions was stable and independent of the concentration in the stock
solution, but decreased with increasing concentration in media. This decrease of ion concentration with
higher concentrations is likely due to the precipitate formed by AlCl3 in cell media.
3.3 Uptake of Al NMs in Caco-2 and HepaRG cells
The uptake and the intracellular distribution of Al0 and Al2O3 NMs following a 24 h treatment in Caco-2
and HepaRG cells were investigated by TEM (Figure 2 and 3). In both cell lines, the majority of Al0 NMs
were found as dense agglomerates of various sizes in the cytoplasm embedded in electron lucent or
dense vesicles which are likely endosomes and lysosomes (Figure 2 and 3, B and C, notched arrows).
Moreover, in certain cases, some Al0 NMs were observed as isolated nanoparticle clusters in the
cytoplasm proximal to the nucleus (Figure 2 and 3 C, full arrows). Observations of Al0 NMs in the nucleus
were very rare, and this result requires further investigation as it may be due to artefacts. The distribution
pattern observed for Al2O3 NMs was similar in both cell lines. While a perinuclear localization of Al2O3
NMs was also seen, this occured less frequently than for Al0 NMs (Figure 2 and 3, D and E). Even at the
lowest concentration, Al0 and Al2O3 NMs were internalized through vesicle formation, most likely
endocytosis, and accumulated in the cytoplasm of Caco-2 and HepaRG cells (Figure S 2).
3.4 Cytotoxicity
Page 12/39
Viability and apoptosis in Caco-2 and HepaRG cells following a 24 h treatment with Al NMs were
investigated by cell counts (Figure 4 A) and active caspase-3 labeling (Figure 4 B) respectively using
automated image analysis. No signicant change in cell numbers and active caspase-3 levels were
observed in either Caco-2 or HepaRG cells treated with Al0 and Al2O3 NMs up to 80 µg/cm2. In addition,
no signicant cytotoxic effects were observed in cells treated with the ionic salt control AlCl3 up to 128
µg/mL Al content (1.16 mg/mL AlCl3).
Similarly, no signicant change in cell numbers were observed in Caco-2 cells treated with ZnO at the
concentrations tested. However, a signicant decrease in cell numbers as well as an increase in active
caspase-3 levels was observed for the highest dose (6 µg/cm2) of ZnO NMs in HepaRG cells.


3.5 Oxidative stress
Quantication of intracellular ROS was used to evaluate oxidative stress in Caco-2 and HepaRG cells
following a 24 h treatment (Figure 5). Intracellular reactive oxygen species (ROS) levels were not
signicantly changed following treatment up to 80 µg/cm2 with Al0 and Al2O3 NMs or the ionic salt
control AlCl3. However, treatment with ZnO NMs signicantly increased levels of ROS at the highest
concentration (6 µg/cm2) in HepaRG cells.
3.6 Genotoxicity
3.6.1 γH2AX
Quantication of γH2AX labeling was used to evaluate the induction of DNA double stand breaks in
Caco-2 and HepaRG cells following a 24 h treatment with Al NMs. Compared to untreated cells, the
γH2AX levels were not affected in the nuclei of Caco-2 cells treated for 24 h with Al0 and Al2O3 NMs up to
80 µg/cm2, with ZnO NMs, or with the ionic salt control AlCl3 up to 128 µg/mL (Figure 6). However in
HepaRG cells, Al0 NMs induced a slight but statistically signicant increase in γH2AX levels at the highest
concentration (80 µg/cm2) tested. ZnO NMs induced signicant increases at 3 and 6 µg/cm2.
3.6.2 Comet assay
The potential for Al0 and Al2O3 NMs to induce DNA damage in Caco-2 and HepaRG cells was investigated
with the alkaline comet assay after a 24 h treatment (Figure 7 A and B). A modied comet assay with the
Fpg enzyme was also performed to detect oxidative DNA damage (Figure 7 C and D).
Page 13/39
In Caco-2 cells, a signicant increase in tail DNA was observed with Al0 NMs from 28 to 80 µg/cm2 in the
alkaline comet assay (Figure 7 A). In contrast, neither Al2O3 and ZnO NMs, or the ionic salt control AlCl3
induced any signicant increase in tail DNA. In the Fpg-modied comet assay, a signicant increase in
tail DNA was observed in cells treated with Al2O3 NMs at 3, 9 and 80 µg/cm2 (Figure 7 C).
In HepaRG cells, tail DNA signicantly increased in a dose-dependent manner in cells treated with Al0
NMs, including a very considerable effect starting at 28 µg/cm2. In contrast, no effect was observed for
cells treated with Al2O3 and ZnO NMs, or the ionic control AlCl3 (Figure 7 B). Similarly, an increase in tail
DNA in the Fpg-modied comet assay was observed for Al0 NMs at all concentrations tested with a very
signicant effect observed at concentrations above 9 µg/cm2. No signicant changes in tail DNA were
detected in HepaRG cells treated with Al2O3 and ZnO NMs or AlCl3 (Figure 7 D).
DNA damage in Caco-2 and HepaRG was also investigated by the alkaline comet assay after a 5 h
treatment (Figure S 3). In Caco-2 cells, a concentration-dependent increase in tail DNA was observed in
cells treated with Al0 NMs from 28 to 80 µg/cm2. No effect was detected in cells treated with Al2O3 and
ZnO NMs, or the ionic salt control AlCl3. In HepaRG cells, a concentration-dependent increase was also
observed with Al0 NMs from 9 to 80 µg/cm2. In the Fpg-modied comet assay, an increase in tail DNA
was detected in Caco-2 cells treated with Al0 NMs from 9 to 80 µg/cm2 and with Al2O3 NMs at 3, 9 and 28
µg/cm2. Interestingly, in HepaRG cells treated for 5 h with Al NMs, results from the Fpg-modied comet
assay showed that at every concentration tested, only hedgehogs were observed for all NMs (data not
shown).
3.6.3 Interaction of NMs with DNA during the comet assay
The interference of NMs with the comet assay was assessed (Figure 8) according to the protocol
described by Bessa et al [35]. Compared to the untreated control, a concentration-dependent increase in %
tail DNA was observed when Al0 NMs are added at nal concentrations of 9 and 40 µg/cm2. A similar
effect was also observed when Fpg was included in the assay. Compared to the negative control, no
difference was detected for Al2O3 NMs in the absence of Fpg, while a slight increase was observed with
Fpg.
3.6.4 Micronucleus assay
In order to evaluate chromosome damage, the cytokinesis-block micronucleus assay was performed in
Caco-2 and HepaRG cells treated for 24 h (Table 4) with Al NMs. No modication of cell viability (RI
value) was observed in either Caco-2 or HepaRG cells exposed to Al0, Al2O3, ZnO NMs and AlCl3.
Compared to the negative control, no signicant increase in the percentage of BNMN cells was detected
Page 14/39
in either cell line. Similarly, no increase in micronucleated mononuleated cells or in polyploid cells was
observed (data not shown).
4. Bhas-42 Cell transforming (CTA) assay
Results of Bhas-42 CTA performed with Al0 and Al2O3 NMs and with AlC13 are shown in Table 5. In the
initiation assay, both Al0 NMs and AlC13 induced a concentration-dependent decrease in cell proliferation
on Day 7, inhibiting around 90% of cell proliferation at the highest concentration (3 µg/cm2 for Al0 NMs
and 28 µg/ml for AlCl3). In contrast, Al2O3 NMs in the initiation assay and the three Al forms in the
promotion assay induced no, or only a moderate, decrease in cell proliferation for all concentrations
tested. No transforming activity was shown with the three Al forms, irrespective of the concentration
tested in both the initiation and promotion assays. In contrast, the number of foci was found lower than
those of controls at some concentrations of Al and Al2O3.
4. Discussion
Exposure of the general population to NMs present in consumer products, including food, has increased
dramatically within the last decade, and a thorough evaluation of the potential adverse effects resulting
from exposure to NMs following ingestion is necessary. Among the toxic effects of Al-containing NMs
that have been shown in several studies, genetic damage is of particular concern [15, 21, 22, 23]. Both
intestine and liver are considered key organs for investigating genotoxic effect of nanomaterials found in
food since they represent the main organ of contact and the main organ of accumulation, respectively.
Nevertheless, in our recent
in vivo
study investigating the genotoxicity of Al NMs, only a very limited
genotoxic response was observed. In fact, only a cross-linking effect was suggested in the rat duodenum
with Al0 NMs [16]. As the
in vivo
treatment duration was rather short (3 administrations over 2 days), and
that it cannot be excluded that the level of NMs in the organs would be low, we chose to complete our
study by investigating the
in vitro
genotoxicity of Al NMs in human intestinal Caco-2 and hepatic HepaRG
cells using complementary tests.
Despite the uptake and presence of Al NMs in Caco-2 and HepaRG cells, no cytotoxicity or apoptotic
response was observed following treatment with Al2O3 NMs. Our results are in agreement with data from
various publications that have reported little or no cytotoxicity in various cell lines [21, 27, 34, 50, 51],
including in Caco-2 cells [52, 53] and HepG2 cells [22].
No induction of chromosomal damage was observed in the micronucleus assay in either Caco-2 or
HepaRG cells exposed to Al2O3 NMs. Moreover, we did not observe a transforming activity in the CBA
assay, supporting the absence of mutagenic potential for Al2O3 NMs. Our results are consistent with two
recent studies that reported a negative response in the chromosomal aberration and the micronucleus
assays in human lymphocytes treated with Al2O3 NMs with a smaller size (3 to 4 nm) than the one used
in this study (20 nm), and for a longer incubation time (72 h) [20, 54]. In contrast, other studies have
Page 15/39
reported an increase in micronucleus formation following a 24 h treatment with Al2O3 NMs in other cell
lines, including CHO cells [23], human broblasts [21] and RAW264 murine macrophages [24].
Interestingly, Al2O3 NMs were shown to inhibit the replication eciency of high-delity DNA polymerase
[55]. Nevertheless, such inhibition did not affect the mutation rate at the single nucleotide level of
replication products compared to controls [55]. Further investigation demonstrated that Al2O3 NMs did
not induce a clastogenic effect but rather chromosome loss and polyploidy, although these effects were
observed only at one concentration [21]. An aneugenic effect of Al NMs was not observed in our study
(data not shown). The discrepancy may be explained by the fact that our tests were performed in non-
proliferating cells.
Similarly, numerical chromosomal damage (aneuploidy and polyploidy) and abnormal metaphases were
reported in the bone marrow of rats 48 hours after a single oral dose of Al2O3 NMs while no effect was
observed with bulk Al2O3 [14, 15]. In addition, induction of micronuclei in erythrocytes was also observed.
However, this genotoxic effect on erythrocytes was concomitant with a cytotoxic effect, while no toxicity
was observed in our study [16], or in the study of Zhang et al [56]. In contrast, other results obtained from
in vivo
studies are in agreement with the lack of chromosomal damage observed
in vitro
in our study
following treatment with Al2O3 NMs. In fact, with the same Al2O3 NMs used in this study, we did not
observe an induction of micronuclei in either bone marrow or in the colon of rats after a short-term oral
treatment [16]. Similarly, no induction of micronuclei in the bone marrow of mice was detected following
intraperitoneal injections, irrespective of the size of the Al2O3 particles [56].
The absence of genotoxic activity of Al2O3 NMs in Caco-2 and HepaRG cells was further conrmed in the
H2AX assay as well as the comet assay. We did not observe any increase in H2AX levels in either cell
line, which is in agreement with results from a study by Tsaousi et al [21] in primary human broblasts.
Additionally, Al2O3 NMs did not induce DNA damage in the alkaline comet assay in Caco-2 and HepaRG
cells following a 24 h treatment. Although some studies have reported negative results in the comet
assay in human lymphocytes and in human embryonic kidney cells [20, 26], others have demonstrated
time- and/or concentration-dependent genotoxic effects in Chinese hamster lung broblasts [56], in
RAW264 murine macrophages [24] and in human liver HepG2 cells [22] treated with Al2O3 NMs.
Nevertheless, the increase of DNA fragmentation in these latter studies was probably linked to cell death
detected by Trypan blue exclusion [56] or by apoptotic markers [22, 24].
In vivo
, after a short-term treatment using the same Al2O3 NMs, we only observed an increase in DNA
damage in the comet assay in bone marrow, while no effect was observed in intestine, colon, kidney,
spleen or blood [16]. Balasubramanyam et al [14] showed a time- and concentration-dependent increase
in DNA damage in blood with the comet assay with both bulk and nano Al2O3 forms after a single gavage
but the effect decreased at 48 h before disappearing at 72 h. DNA breakage associated with necrosis
and apoptosis was observed in liver and kidney of rats after a repeated oral treatment for 75 days with 70
mg/kg bw Al2O3 NMs [57]. Therefore, it seems that both the
in vitro
and
in vivo
results with Al2O3 NMs
Page 16/39
support the conclusion that DNA breaks detected by the comet assay were mostly related to cell death
rather than to a clear genotoxicity.
Nevertheless, we have shown that Al2O3 NMs induced oxidative DNA damage in Caco-2 cells following a
24 h treatment, despite no signicant ROS induction. Furthermore, a concentration-dependent trend
towards oxidative damage was observed at 5 h. This could suggest the rapid formation of oxidative DNA
damage which is further repaired, as previously demonstrated [58, 59]. Evidence from
in vitro
experiments
in a variety of different cell lines suggests that treatment with Al2O3 NMs can induce oxidative stress [20,
56, 60, 61] including in Caco2 cells [53]. Interestingly, Alari et al [22] reported positive results in the comet
assay in HepG2 cells which was accompanied by oxidative damage and cell death. In the present study,
no oxidative DNA damage or oxidative stress was observed in HepaRG cells. Differentiated HepaRG cells
represent a model which is more similar to human hepatocytes when compared to HepG2 cells, and could
therefore be less sensitive to oxidative damage resulting from Al2O3 NMs. Similarly, we did not detect
oxidative DNA damage in liver, or in other organs of rats after oral exposure [16]. In contrast, an increase
in oxidative stress was observed in several tissues including liver after acute and repeated oral exposure
of rats with Al2O3 NMs [33].
Similar to the results obtained for Al2O3 NMs, no cytotoxicity or apoptotic response was observed
following treatment with Al0 NMs, despite their presence in the cytoplasm of Caco-2 and HepaRG cells. In
contrast to our results in differentiated Caco-2 and HepaRG cells, Al0 NMs were found to induce a
decrease in viability in rat alveolar macrophages and in BRL3A rat liver cells following 24 h exposure at
concentrations similar to those used in our study [27, 62]. This discrepancy could be explained by a
difference in relative cell density for a similar concentration of Al0 NMs tested with a lower NM:cell ratio
in differentiated Caco2 and HepaRG cells compared to the two other proliferating cell systems.
Despite only a slight increase in H2AX levels observed only in HepaRG cells and only at the highest
concentration tested, a dose-dependent increase in tail DNA was observed in both Caco-2 and HepaRG
cell lines treated with Al0 NMs using the alkaline comet assay after both 5 h and 24 h treatments.
Nevertheless, this result required further investigation due to possible interference of NMs with the
alkaline comet assay that has been widely documented in the literature [35, 36, 63, 64]. Indeed, NMs
present in the cytoplasm of cells following uptake can interact with DNA following the lysis step of the
comet assay, and could therefore induce additional breaks or inhibit DNA migration. In addition, a
dissolution due to the conditions of the comet assay could result in reaction of aluminum ions with DNA,
especially the phosphate backbone, as reported in some studies [65, 66]. Such reactions may then induce
DNA damage revealed during the comet assay as suggested by Zhang et al [67]. Our results clearly
demonstrate that, unlike Al2O3 NMs, Al0 NMs can induce DNA damage when in contact with DNA and
interfere signicantly with the comet assay. Consequently, the positive results in cells treated with Al0
NMs obtained in this study should therefore be treated with caution.
In vivo
, using the same Al0 NMs as
the present study, no genotoxic response was observed in several key tissues, with the exception in rat
duodenum where a cross-linking effect was suggested [16].
Page 17/39
The carcinogenic potential of Al NMs was investigated using the cell transformation assay with Bhas 42
cells. Neither Al0 nor Al2O3 NMs induced cell transformation, although a decrease in the number of
transformed foci was observed. This decrease, observed at concentrations inducing a weak inhibition of
cell proliferation at Day 7, is likely explained by a more pronounced inhibition of cell growth after 21 days
of culture due to the three repeated treatments during the promotion assay. This phenomena was also
observed with amorphous silica NMs [49] as well as with other non-carcinogenic chemicals such as L-
ascorbic acid and caffeine [68].
Ion release from NMs in cell culture media, or in intracellular compartments can contribute to cytotoxic
effects
in vitro
. The soluble fraction of Al0 and Al2O3 NMs measured by ICP-MS demonstrated a very low
solubility of Al0 and Al2O3 NMs in both cell media . However, ion release may occur after cell uptake in
specic compartments with low pH such as lysosomes [69] as suggested for Al2O3 NMs [24]. In such a
scenario, secondary effects affecting mitochondria and resulting in the generation of ROS cannot be
excluded. In the case of Al0, the formation of a passivating oxide layer may inuence its dissolution
behavior [70]. Consequently, effects could be induced by ionic Al released from the NMs rather than
effects related to the particulate form [1]. As a strong oxygen acceptor, the Al ion tends to bind to citrate,
phosphate, and catecholamine, generating oxygen radicals [1, 71]. In addition, Al ions can also bind to
negatively charged phospholipids, which are easily attacked by reactive oxygen species such as O2·,
H2O2, and OH· [72, 73] as well as DNA [66].
No genotoxic effects were observed in differentiated Caco-2 or HepaRG cells treated with AlCl3 at
concentrations up to 128 µg/mL Al content corresponding to 1.16 mg/mL AlCl3. At the concentrations of
AlCl3 tested, no effects were observed in the different assays following 5 or 24 h treatments. Indeed,
negative results were obtained for promotion and initiation, as well as for genotoxic and oxidative stress
responses. Our results are consistent with Villarini et al [74] who observed no genotoxicity in response to
Al ions in neuroblastoma cells with the comet assay, as well as no cytotoxicity or oxidative stress.
However, other studies have reported genotoxicity of AlCl3 in human lymphocytes [17, 29]. Interestingly,
the authors of this study observed the highest level of micronuclei during the G1-phase of the cell cycle.
The differentiated HepaRG and Caco2 cells used in our study are not proliferating, and therefore could
explain the discrepancy between the studies.
In vitro
, chromosomal damage observed in blood cells at
AlCl3 concentrations below 25 µg/mL, was associated with apoptosis [28, 29, 30]. Moreover it was shown
that Al ions can induce oxidative DNA damage irrespective of the cell cycle phase [29]. Indeed, the role of
Al ions in mediating genotoxic effects may be more complex, as it has been suggested that Al ions may
inhibit several DNA repair proteins with zinc nger domains [29, 75].
In our study, as the soluble fraction of AlCl3 was always higher than that for Al0 and Al2O3 NMs, the
effects observed for Al0 and Al2O3 NMs are not likely to be related to ion release in cell media. Although
ECHA emphasized that the difference in the toxicological prole between soluble aluminum compounds
and insoluble aluminum oxide may be explained by lower bioavailability of insoluble test compounds, it
Page 18/39
was recently shown that the content of Al in blood of rats treated orally was higher with Al2O3 NMs than
with AlCl3 [76]. Moreover, the persistence of NMs in organs long after intial exposures has been described,
and the accumulation of Al NMs in organs following repeated exposure could poteniate adverse effects
in tissues in the long term. Further studies are clearly needed to investigate the fate of accumulated NMs
in tissue, including possible effects due to ion release, as well as toxic effects related to particle
accumulation.
5. Conclusion
In summary, despite the uptake and presence of Al NMs in the cytoplasm of differentiated Caco-2 and
HepaRG cells, we have shown that Al2O3 NMs do not induce apoptosis, oxidative stress, or cytotoxic
effects following a 24 h treatment. In addition, Al2O3 NMs were negative in the micronucleus assay, and
in initiation and promotion in the CTA. Nevertheless, oxidative DNA damage was observed in Caco-2 cells.
The assays performed with Al0 NMs and AlCl3 were also negative except a slight increase of H2AX levels
only in HepaRG cells, and only at the highest concentration tested. Considerable DNA damage was
observed with Al0 NMs in both Caco-2 and HepaRG cells in the comet assay, although this was likely
associated with the signicant interference with these NMs, and these results must be taken with caution.
As ion release from Al NMs was shown to be very limited in cell media, the effects are rather due to the
particulate form or to ion release inside the cells. Further investigation is needed to clarify the extent of
intracellular ion release from NMs, its contribution to cytotoxic effects compared to the direct impact of
the presence of intracellular particles.
Declarations
Ethical Approval and Consent to participate
Not applicable.
Consent for publication
Not applicable.
Availability of supporting data
The datasets used and/or analysed during the current study are available from the corresponding author
on reasonable request.
Competing interests
Page 19/39
The authors declare that they have no competing interests.
Funding
This publication arises from the French-German bilateral project SolNanoTOX funded by the German
Research Foundation (DFG, Project ID: DFG (FKZ LA 3411/1-1 respectively LA 1177/9-1) and the French
National Research Agency (ANR, Project ID: ANR-13-IS10-0005).
Authors' contributions
PJ and AB performed the electron microscopy study and analysis. BCK performed the experiments for
dispersion and dissolution characterization. CF and YG performed and analysed the CTA. FG and SC
performed the density and dispersion chracterisation. PJ, SH and KH performed the genotoxicity
experiments. PJ, BCK, FG, YG, KH and VF wrote the manuscript. AB, FG, AlL, PL, AnL, KH and VF wrote the
proposal to obtain funding. All authors read and approved the nal manuscript.
Acknowledgements
The authors would like to thank Rachelle Lanceleur and Marie-Thérèse Lavault for technical assistance.
References
1. Willhite CC, Karyakina NA, Yokel RA, Yenugadhati N, Wisniewski TM, Arnold IM, et al. Systematic
review of potential health risks posed by pharmaceutical, occupational and consumer exposures to
metallic and nanoscale aluminum, aluminum oxides, aluminum hydroxide and its soluble salts.
Critical reviews in toxicology. 2014;44 Suppl 4:1-80; doi: 10.3109/10408444.2014.934439.
2. Som C, Wick P, Krug H, Nowack B. Environmental and health effects of nanomaterials in nanotextiles
and façade coatings. Environment International. 2011;37 6:1131-42; doi:
10.1016/j.envint.2011.02.013.
3. Shepard MN, Brenner S. An Occupational Exposure Assessment for Engineered Nanoparticles Used
in Semiconductor Fabrication. The Annals of Occupational Hygiene. 2013;58 2:251-65; doi:
10.1093/annhyg/met064.
4. Li X, Aldayel AM, Cui Z. Aluminum hydroxide nanoparticles show a stronger vaccine adjuvant activity
than traditional aluminum hydroxide microparticles. J Control Release. 2014;173:148-57; doi:
10.1016/j.jconrel.2013.10.032.
5. Saiyed SM, Yokel RA. Aluminium content of some foods and food products in the USA, with
aluminium food additives. Food additives and contaminants. 2005;22 3:234-44; doi:
10.1080/02652030500073584.
Page 20/39
6. Moradi Z, Esmaiili M, Almasi H. Development and characterization of keran - Al2O3 nanocomposite
lms: Morphological, physical and mechanical properties. International Journal of Biological
Macromolecules. 2019;122:603-9; doi: 10.1016/j.ijbiomac.2018.10.193.
7. European Food Safety A. Safety of aluminium from dietary intake - Scientic Opinion of the Panel on
Food Additives, Flavourings, Processing Aids and Food Contact Materials (AFC). EFSA Journal.
2008;6 7:754; doi: 10.2903/j.efsa.2008.754.
8. Kumar E, Bhatnagar A, Kumar U, Sillanpää M. Deuoridation from aqueous solutions by nano-
alumina: Characterization and sorption studies. Journal of Hazardous Materials. 2011;186 2:1042-9;
doi: 10.1016/j.jhazmat.2010.11.102.
9. Zhang D, Quayle MJ, Petersson G, van Ommen JR, Folestad S. Atomic scale surface engineering of
micro- to nano-sized pharmaceutical particles for drug delivery applications. Nanoscale. 2017;9
32:11410-7; doi: 10.1039/C7NR03261G.
10. Zhao J, Castranova V. Toxicology of nanomaterials used in nanomedicine. Journal of toxicology and
environmental health Part B, Critical reviews. 2011;14 8:593-632; doi:
10.1080/10937404.2011.615113.
11. Narayan RJ, Adiga SP, Pellin MJ, Curtiss LA, Hryn AJ, Stafslien S, et al. Atomic layer deposition-based
functionalization of materials for medical and environmental health applications. Philosophical
transactions Series A, Mathematical, physical, and engineering sciences. 2010;368 1917:2033-64;
doi: 10.1098/rsta.2010.0011.
12. Laux P, Tentschert J, Riebeling C, Braeuning A, Creutzenberg O, Epp A, et al. Nanomaterials: certain
aspects of application, risk assessment and risk communication. Archives of toxicology. 2018;92
1:121-41; doi: 10.1007/s00204-017-2144-1.
13. Tietz T, Lenzner A, Kolbaum AE, Zellmer S, Riebeling C, Gürtler R, et al. Aggregated aluminium
exposure: risk assessment for the general population. Archives of toxicology. 2019;93 12:3503-21;
doi: 10.1007/s00204-019-02599-z.
14. Balasubramanyam A, Sailaja N, Mahboob M, Rahman MF, Hussain SM, Grover P. In vivo genotoxicity
assessment of aluminium oxide nanomaterials in rat peripheral blood cells using the comet assay
and micronucleus test. Mutagenesis. 2009;24 3:245-51; doi: 10.1093/mutage/gep003.
15. Balasubramanyam A, Sailaja N, Mahboob M, Rahman MF, Misra S, Hussain SM, et al. Evaluation of
genotoxic effects of oral exposure to aluminum oxide nanomaterials in rat bone marrow. Mutation
research. 2009;676 1-2:41-7; doi: 10.1016/j.mrgentox.2009.03.004.
16. Jalili P, Huet S, Lanceleur R, Jarry G, Le Hegarat L, Nesslany F, et al. Genotoxicity of Aluminum and
Aluminum Oxide Nanomaterials in Rats Following Oral Exposure. Nanomaterials (Basel). 2020;10
2:305; doi: 10.3390/nano10020305.
17. Paz LNF, Moura LM, Feio DCA, Cardoso MdSG, Ximenes WLO, Montenegro RC, et al. Evaluation of
invivo and invitro toxicological and genotoxic potential of aluminum chloride. Chemosphere.
2017;175:130-7; doi: 10.1016/j.chemosphere.2017.02.011.
Page 21/39
18. Turkez H, Yousef MI, Geyikoglu F. Propolis prevents aluminium-induced genetic and hepatic damages
in rat liver. Food and chemical toxicology : an international journal published for the British Industrial
Biological Research Association. 2010;48 10:2741-6; doi: 10.1016/j.fct.2010.06.049.
19. Turkez H, Geyikoglu F, Tatar A. Borax counteracts genotoxicity of aluminum in rat liver. Toxicology
and industrial health. 2013;29 9:775-9; doi: 10.1177/0748233712442739.
20. Rajiv S, Jerobin J, Saranya V, Nainawat M, Sharma A, Makwana P, et al. Comparative cytotoxicity and
genotoxicity of cobalt (II, III) oxide, iron (III) oxide, silicon dioxide, and aluminum oxide nanoparticles
on human lymphocytes in vitro. Human & experimental toxicology. 2016;35 2:170-83; doi:
10.1177/0960327115579208.
21. Tsaousi A, Jones E, Case CP. The in vitro genotoxicity of orthopaedic ceramic (Al2O3) and metal
(CoCr alloy) particles. Mutation research. 2010;697 1-2:1-9; doi: 10.1016/j.mrgentox.2010.01.012.
22. Alari S, Ali D, Alkahtani S. Nanoalumina induces apoptosis by impairing antioxidant enzyme
systems in human hepatocarcinoma cells. Int J Nanomedicine. 2015;10:3751-60; doi:
10.2147/IJN.S82050.
23. Di Virgilio AL, Reigosa M, Arnal PM, Fernández Lorenzo de Mele M. Comparative study of the
cytotoxic and genotoxic effects of titanium oxide and aluminium oxide nanoparticles in Chinese
hamster ovary (CHO-K1) cells. Journal of Hazardous Materials. 2010;177 1:711-8; doi:
10.1016/j.jhazmat.2009.12.089.
24. Hashimoto M, Imazato S. Cytotoxic and genotoxic characterization of aluminum and silicon oxide
nanoparticles in macrophages. Dental Materials. 2015;31 5:556-64; doi:
10.1016/j.dental.2015.02.009.
25. McKenna DJ, Gallus M, McKeown SR, Downes CS, McKelvey-Martin VJ. Modication of the alkaline
Comet assay to allow simultaneous evaluation of mitomycin C-induced DNA cross-link damage and
repair of specic DNA sequences in RT4 cells. DNA Repair. 2003;2 8:879-90; doi: 10.1016/S1568-
7864(03)00086-7.
26. Demir E, Burgucu D, Turna F, Aksakal S, Kaya B. Determination of TiO2, ZrO2, and Al2O3
nanoparticles on genotoxic responses in human peripheral blood lymphocytes and cultured
embyronic kidney cells. Journal of toxicology and environmental health Part A. 2013;76 16:990-1002;
doi: 10.1080/15287394.2013.830584.
27. Wagner AJ, Bleckmann CA, Murdock RC, Schrand AM, Schlager JJ, Hussain SM. Cellular Interaction
of Different Forms of Aluminum Nanoparticles in Rat Alveolar Macrophages. The Journal of
Physical Chemistry B. 2007;111 25:7353-9; doi: 10.1021/jp068938n.
28. Lima PDL, Leite DS, Vasconcellos MC, Cavalcanti BC, Santos RA, Costa-Lotufo LV, et al. Genotoxic
effects of aluminum chloride in cultured human lymphocytes treated in different phases of cell cycle.
Food and Chemical Toxicology. 2007;45 7:1154-9; doi: 10.1016/j.fct.2006.12.022.
29. Lankoff A, Banasik A, Duma A, Ochniak E, Lisowska H, Kuszewski T, et al. A comet assay study
reveals that aluminium induces DNA damage and inhibits the repair of radiation-induced lesions in
Page 22/39
human peripheral blood lymphocytes. Toxicology letters. 2006;161 1:27-36; doi:
10.1016/j.toxlet.2005.07.012.
30. Banasik A, Lankoff A, Piskulak A, Adamowska K, Lisowska H, Wojcik A. Aluminum-induced
micronuclei and apoptosis in human peripheral-blood lymphocytes treated during different phases of
the cell cycle. Environmental toxicology. 2005;20 4:402-6; doi: 10.1002/tox.20125.
31. ECHA. Registration dossier Aluminum oxide. https://echa.europa.eu/fr/registration-
dossier/-/registered-dossier/16039/7/7/1.
32. Park EJ, Sim J, Kim Y, Han BS, Yoon C, Lee S, et al. A 13-week repeated-dose oral toxicity and
bioaccumulation of aluminum oxide nanoparticles in mice. Archives of toxicology. 2015;89 3:371-9;
doi: 10.1007/s00204-014-1256-0.
33. Shrivastava R, Raza S, Yadav A, Kushwaha P, Flora SJS. Effects of sub-acute exposure to TiO2, ZnO
and Al2O3 nanoparticles on oxidative stress and histological changes in mouse liver and brain. Drug
and Chemical Toxicology. 2014;37 3:336-47; doi: 10.3109/01480545.2013.866134.
34. Monteiro-Riviere NA, Oldenburg SJ, Inman AO. Interactions of aluminum nanoparticles with human
epidermal keratinocytes. Journal of applied toxicology : JAT. 2010;30 3:276-85; doi:
10.1002/jat.1494.
35. Bessa MJ, Costa C, Reinosa J, Pereira C, Fraga S, Fernández J, et al. Moving into advanced
nanomaterials. Toxicity of rutile TiO(2) nanoparticles immobilized in nanokaolin nanocomposites on
HepG2 cell line. Toxicology and applied pharmacology. 2017;316:114-22; doi:
10.1016/j.taap.2016.12.018.
36. Azqueta A, Dusinska M. The use of the comet assay for the evaluation of the genotoxicity of
nanomaterials. Front Genet. 2015;6:239-; doi: 10.3389/fgene.2015.00239.
37. Ferraro D, Anselmi-Tamburini U, Tredici IG, Ricci V, Sommi P. Overestimation of nanoparticles-induced
DNA damage determined by the comet assay. Nanotoxicology. 2016;10 7:861-70; doi:
10.3109/17435390.2015.1130274.
38. Di Bucchianico S, Cappellini F, Le Bihanic F, Zhang Y, Dreij K, Karlsson HL. Genotoxicity of TiO2
nanoparticles assessed by mini-gel comet assay and micronucleus scoring with ow cytometry.
Mutagenesis. 2017;32 1:127-37; doi: 10.1093/mutage/gew030.
39. Hartmann NB, Jensen KA, Baun A, Rasmussen K, Rauscher H, Tantra R, et al. Techniques and
Protocols for Dispersing Nanoparticle Powders in Aqueous Media-Is there a Rationale for
Harmonization? Journal of toxicology and environmental health Part B, Critical reviews. 2015;18
6:299-326; doi: 10.1080/10937404.2015.1074969.
40. Jalili P, Gueniche N, Lanceleur R, Burel A, Lavault M-T, Sieg H, et al. Investigation of the in vitro
genotoxicity of two rutile TiO2 nanomaterials in human intestinal and hepatic cells and evaluation of
their interference with toxicity assays. NanoImpact. 2018;11:69-81; doi:
10.1016/j.impact.2018.02.004.
41. Aninat C, Piton A, Glaise D, Le Charpentier T, Langouët S, Morel F, et al. Expression of cytochromes
P450, conjugating enzymes and nuclear receptors in human hepatoma HepaRG cells. Drug
Page 23/39
Metabolism and Disposition. 2006;34 1:75; doi: 10.1124/dmd.105.006759.
42. DeLoid GM, Cohen JM, Pyrgiotakis G, Demokritou P. Preparation, characterization, and in vitro
dosimetry of dispersed, engineered nanomaterials. Nature protocols. 2017;12 2:355-71; doi:
10.1038/nprot.2016.172.
43. Le Hégarat L, Huet S, Fessard V. A co-culture system of human intestinal Caco-2 cells and
lymphoblastoid TK6 cells for investigating the genotoxicity of oral compounds. Mutagenesis.
2012;27 6:631-6; doi: 10.1093/mutage/ges028.
44. Dušinská M, Collins A. Detection of Oxidised Purines and UV-induced Photoproducts in DNA of
Single Cells, by Inclusion of Lesion-specic Enzymes in the Comet Assay. Alternatives to Laboratory
Animals. 1996;24 3:405-11; doi: 10.1177/026119299602400315.
45. Collins AR, Duthie SJ, Dobson VL. Direct enzymic detection of endogenous oxidative base damage in
human lymphocyte DNA. Carcinogenesis. 1993;14 9:1733-5; doi: 10.1093/carcin/14.9.1733.
46. OCDE. Test No. 487: In Vitro Mammalian Cell Micronucleus Test. 2010.
47. Sasaki K, Mizusawa H, Ishidate M. Isolation and characterization of ras-transfected BALB/3T3 clone
showing morphological transformation by 12-O-tetradecanoyl-phorbol-13-acetate. Jpn J Cancer Res.
1988;79 8:921-30; doi: 10.1111/j.1349-7006.1988.tb00056.x.
48. OECD. Guidance document on the in vitro Bhas 42 cell transformation assay. Series on Testing &
Assessment No. 231. 2017; doi:
http://www.oecd.org/ocialdocuments/publicdisplaydocumentpdf/?
cote=ENV/JM/MONO(2016)1&doclanguage=en.
49. Fontana C, Kirsch A, Seidel C, Marpeaux L, Darne C, Gaté L, et al. In vitro cell transformation induced
by synthetic amorphous silica nanoparticles. Mutation research. 2017;823:22-7; doi:
10.1016/j.mrgentox.2017.08.002.
50. Radziun E, Dudkiewicz Wilczynska J, Ksiazek I, Nowak K, Anuszewska EL, Kunicki A, et al.
Assessment of the cytotoxicity of aluminium oxide nanoparticles on selected mammalian cells.
Toxicology in vitro : an international journal published in association with BIBRA. 2011;25 8:1694-
700; doi: 10.1016/j.tiv.2011.07.010.
51. Simon-Deckers A, Gouget B, Mayne-L’Hermite M, Herlin-Boime N, Reynaud C, Carrière M. In vitro
investigation of oxide nanoparticle and carbon nanotube toxicity and intracellular accumulation in
A549 human pneumocytes. Toxicology. 2008;253 1:137-46; doi: 10.1016/j.tox.2008.09.007.
52. Ivask A, T, Tiina,, V, Meeri, Vija H, K, Aleksandr,, Sihtmae M, et al. Toxicity of 11 Metal Oxide
Nanoparticles to Three Mammalian Cell Types <i>In Vitro</i>. Current Topics in Medicinal Chemistry.
2015;15 18:1914-29; doi: 10.2174/1568026615666150506150109.
53. Song Z-M, Tang H, Deng X, Xiang K, Cao A, Liu Y, et al. Comparing Toxicity of Alumina and Zinc
Oxide Nanoparticles on the Human Intestinal Epithelium In Vitro Model. Journal of Nanoscience and
Nanotechnology. 2017;17 5:2881-91; doi: 10.1166/jnn.2017.13056.
54. Akbaba GB, Turkez H. Investigation of the Genotoxicity of Aluminum Oxide, beta-Tricalcium
Phosphate, and Zinc Oxide Nanoparticles In Vitro. International journal of toxicology. 2018;37 3:216-
Page 24/39
22; doi: 10.1177/1091581818775709.
55. Gao C-H, Mortimer M, Zhang M, Holden PA, Cai P, Wu S, et al. Impact of metal oxide nanoparticles on
in vitro DNA amplication. PeerJ. 2019;7:e7228-e; doi: 10.7717/peerj.7228.
56. Zhang Q, Wang H, Ge C, Duncan J, He K, Adeosun SO, et al. Alumina at 50 and 13 nm nanoparticle
sizes have potential genotoxicity. Journal of applied toxicology : JAT. 2017;37 9:1053-64; doi:
10.1002/jat.3456.
57. Yousef MI, Mutar TF, Kamel MAE. Hepato-renal toxicity of oral sub-chronic exposure to aluminum
oxide and/or zinc oxide nanoparticles in rats. Toxicology reports. 2019;6:336-46; doi:
10.1016/j.toxrep.2019.04.003.
58. Sliwinska A, Kwiatkowski D, Czarny P, Milczarek J, Toma M, Korycinska A, et al. Genotoxicity and
cytotoxicity of ZnO and Al2O3 nanoparticles. Toxicology Mechanisms and Methods. 2015;25 3:176-
83; doi: 10.3109/15376516.2015.1006509.
59. Sadiq R, Khan QM, Mobeen A, Hashmat AJ. In vitro toxicological assessment of iron oxide,
aluminium oxide and copper nanoparticles in prokaryotic and eukaryotic cell types. Drug and
Chemical Toxicology. 2015;38 2:152-61; doi: 10.3109/01480545.2014.919584.
60. Shah SA, Yoon GH, Ahmad A, Ullah F, Amin FU, Kim MO. Nanoscale-alumina induces oxidative stress
and accelerates amyloid beta (Aβ) production in ICR female mice. Nanoscale. 2015;7 37:15225-37;
doi: 10.1039/C5NR03598H.
61. Li X, Zhang C, Zhang X, Wang S, Meng Q, Wu S, et al. An acetyl-L-carnitine switch on mitochondrial
dysfunction and rescue in the metabolomics study on aluminum oxide nanoparticles. Particle and
bre toxicology. 2016;13:4-; doi: 10.1186/s12989-016-0115-y.
62. Hussain SM, Hess KL, Gearhart JM, Geiss KT, Schlager JJ. In vitro toxicity of nanoparticles in BRL 3A
rat liver cells. Toxicology in Vitro. 2005;19 7:975-83; doi: 10.1016/j.tiv.2005.06.034.
63. Magdolenova Z, Collins A, Kumar A, Dhawan A, Stone V, Dusinska M. Mechanisms of genotoxicity. A
review of in vitro and in vivo studies with engineered nanoparticles. Nanotoxicology. 2014;8 3:233-78;
doi: 10.3109/17435390.2013.773464.
64. Karlsson HL, Di Bucchianico S, Collins AR, Dusinska M. Can the comet assay be used reliably to
detect nanoparticle-induced genotoxicity? Environmental and molecular mutagenesis. 2015;56 2:82-
96; doi: 10.1002/em.21933.
65. Heli H. A study of double stranded DNA adsorption on aluminum surface by means of
electrochemical impedance spectroscopy. Colloids and Surfaces B: Biointerfaces. 2014;116:526-30;
doi: 10.1016/j.colsurfb.2014.01.046.
66. Zhang R-Y, Liu Y, Pang D-W, Cai R-X, Qi Y-P. Spectroscopic and Voltammetric Study on the Binding of
Aluminium(III) to DNA. Analytical Sciences. 2002;18 7:761-6; doi: 10.2116/analsci.18.761.
67. Zhang F, Cao Q, Cheng J, Zhang C, An N, Bi S. Electrochemical and Spectrometric Studies of Double-
Strand Calf Thymus Gland DNA Denatured by Al(III) at Neutral pH. Analytical Sciences. 2009;25
8:1019-23; doi: 10.2116/analsci.25.1019.
Page 25/39
68. Sakai A, Sasaki K, Muramatsu D, Arai S, Endou N, Kuroda S, et al. A Bhas 42 cell transformation
assay on 98 chemicals: The characteristics and performance for the prediction of chemical
carcinogenicity. Mutation Research/Genetic Toxicology and Environmental Mutagenesis. 2010;702
1:100-22; doi: 10.1016/j.mrgentox.2010.07.007.
69. Sabella S, Carney RP, Brunetti V, Malvindi MA, Al-Juffali N, Vecchio G, et al. A general mechanism for
intracellular toxicity of metal-containing nanoparticles. Nanoscale. 2014;6 12:7052-61; doi:
10.1039/C4NR01234H.
70. Krause B, Meyer T, Sieg H, Kästner C, Reichardt P, Tentschert J, et al. Characterization of aluminum,
aluminum oxide and titanium dioxide nanomaterials using a combination of methods for particle
surface and size analysis. RSC Advances. 2018;8 26:14377 - 88; doi: 10.1039/c8ra00205c.
71. Harris WR, Berthon G, Day JP, Exley C, Flaten TP, Forbes WF, et al. Speciation of aluminum in
biological systems. Journal of Toxicology and Environmental Health. 1996;48 6:543-68; doi:
10.1080/009841096161069.
72. Verstraeten SV, Oteiza PI. Effects of Al3+ and Related Metals on Membrane Phase State and
Hydration: Correlation with Lipid Oxidation. Archives of Biochemistry and Biophysics. 2000;375
2:340-6; doi: 10.1006/abbi.1999.1671.
73. Verstraeten SV, Golub MS, Keen CL, Oteiza PI. Myelin Is a Preferential Target of Aluminum-Mediated
Oxidative Damage. Archives of Biochemistry and Biophysics. 1997;344 2:289-94; doi:
10.1006/abbi.1997.0146.
74. Villarini M, Gambelunghe A, Giustarini D, Ambrosini MV, Fatigoni C, Rossi R, et al. No evidence of DNA
damage by co-exposure to extremely low frequency magnetic elds and aluminum on
neuroblastoma cell lines. Mutatation Research. 2017;823:11-21; doi:
10.1016/j.mrgentox.2017.09.001.
75. Hanas JS, Gunn CG. Inhibition of transcription factor IIIA-DNA interactions by xenobiotic metal ions.
Nucleic Acids Res. 1996;24 5:924-30; doi: 10.1093/nar/24.5.924.
76. Krause BC, Kriegel FL, Rosenkranz D, Dreiack N, Tentschert J, Jungnickel H, et al. Aluminum and
aluminum oxide nanomaterials uptake after oral exposure - a comparative study. Sci Rep. 2020;10
1:2698-; doi: 10.1038/s41598-020-59710-z.
Tables
Table 1: Summary of NM characteristics as reported by the supplier.
Page 26/39
NM NM-code Average particle
sizea
(nm)
SSAb
(m2/g)
Purity
cBulk density
True
densityd
(g/cm3)
Morphology
Al0 NM-0015-
HP 18 40-60 > 99% 2.70
0.008-0.20
spherical
Al2O3 NM-0036-
HP 20 <200 99% -
0.9
spherical
ZnO NM-0011-
HP 20 50 99.5% 5.6
0.3-0.45
Nearly
spherical
a Average particle size was determined by TEM
b Average specic surface area (SSA) was determined by Brunauer-Emmet-Teller (BET)
c Purity was determined by X-ray Powder Diffraction (XRD)
d Density was assessed by normal volumetric test
Table 2: Physico-chemical characterization of Al0, Al2O3 and ZnO NMs.
Page 27/39
Sample
(100 µg/ml)
PdI Z-Ave (d.nm) PdI Z-Ave (d.nm)
Dispersion solution (0h) Dispersion solution (24 h)
Al0 0.173 ± 0.004 254 ± 4 0.159 ± 0.026 253 ± 12
Al2O3 0.235 ± 0.015 168 ± 3 0.186 ± 0.021 160 ± 2
ZnO 0.104 ± 0.038 233 ± 11 0.112 ± 0.045 189 ± 18
DMEM 10% FBS (0h) DMEM 10% FBS (24h)
Al0 0.176 ± 0.011 197 ± 2 0.156 ± 0.011 201 ± 1
Al2O3 0.521 ± 0.027 81 ± 1 0.337 ± 0.041 108 ± 1
ZnO 0.262 ± 0.010 198 ± 4 0.178 ± 0.019 156 ± 9
William’s 5% FBS (0h) William’s 5% FBS (24h)
Al0 0.158 ± 0.008 240 ± 14 0.152 ± 0.007 246 ± 12
Al2O3 0.466 ± 0.015 107 ± 2 0.442 ± 0.018 120 ± 2
ZnO 0.233 ± 0.030 208 ± 6 0.165 ± 0.011 182 ± 11
The mean hydrodynamic diameter (z-Ave) and polydispersity index (PdI) were determined in the stock
dispersion solution and cell media (DMEM + 10 % FBS and William’s + 5 % FBS) after 0 h and 24 h at a
concentration of 100 µg/ml. Three independent experiments were performed. Results are expressed as
mean ± SD.
Table 3: Ion release from Al0, Al2O3, ZnO NMs and AlCl3.
Page 28/39
NMs NM concentration
(µg/mL) Dispersion stock solution
[%] DMEM + 10% FBS
[%] William’s +
5% FBS [%]
Al0 25 1.30 ± 0.06 3.88 ± 0.13 2.42 ± 0.07
50 0.85 ± 0.05 1.94 ± 0.09 1.26 ± 0.07
100 0.48 ± 0.02 0.95 ± 0.04 0.68 ± 0.01
Al2O3 25 0.24 ± 0.04 0.40 ± 0.03 0.32 ± 0.11
50 0.18 ± 0.08 0.48 ± 0.06 0.35 ± 0.07
100 0.15 ± 0.01 0.37 ± 0.02 0.27 ± 0.02
AlCl3 25 89.60 ± 5.97 57.16 ± 5.97 23.35 ±
0.26
50 81.72 ± 0.77 30.09 ± 0.44 12.86 ±
0.28
100 94.31 ± 6.61 17.54 ± 0.72 6.47 ± 0.52
ZnO 25 29.21 ± 0.53 85.09 ± 5.30 54.99 ±
1.03
50 15.07 ± 0.10 57.97 ± 0.68 28.54 ±
1.18
100 7.40 ± 0.05 30.21 ± 0.23 14.74 ±
0.21
Ion release was determined by ICP-MS in the stock dispersion solution and the cell media (DMEM + 10 %
FBS and William’s + 5 % FBS) after 24 h at concentrations of 25, 50 or 100 µg/mL. Data are presented as
the mean ± SD of three independent experiments.
Table 4: Detection of chromosomal damage in differentiated Caco-2 and HepaRG cells treated with Al-
containing NMs.
Page 29/39
 Caco2 HepaRG
%BNMN RI (%) % BNMN RI (%)
Control 0 5.6 ± 0.3 100 ± 0 3.6 ± 0.5 100 ± 0
Al0 NMs
[µg/cm2]
3 5.7 ± 0.4 97 ± 0.8 3.2 ± 0.3 97 ± 5.7
9 5.7 ± 0.5 99 ± 3 3.2 ± 0.2 99 ± 3.1
28 5.5 ± 0.4 103 ± 3.3 2.4 ± 0.2 101 ± 6.7
40 5.9 ± 0.6 98 ± 2.6 3.5 ± 0.9 101 ± 5.5
80 6 ± 0.5 94 ± 0.6 2.2 ± 0.3 110 ± 4.5
Al2O3 NMs
[µg/cm2]
3 6.4 ± 1 102 ± 4 3.6 ± 0.6 99 ± 3.3
9 5.6 ± 1.4 100 ± 2.9 3.3 ± 0.3 102 ± 5.2
28 6.3 ± 1.4 96 ± 2.2 4.5 ± 0.8 109 ± 7.2
40 6.4 ± 0.7 98 ± 0.7 3.5 ± 0.5 106 ± 6.8
80 6.4 ± 1 102 ± 4 3.6 ± 0.6 99 ± 3.3
AlCl3
[µg/ml]
28 6.9 ± 0.5 103 ± 1.5 4.7 ± 1.5 114 ± 3.8
40 6.9 ± 0.8 100 ± 1 4.3 ± 0.8 113 ± 3.3
ZnO NMs
[µg/cm2]
3 7 ± 0.8 100 ± 4.1 3.7 ± 0.4 96 ± 3.5
6 8 ± 0.7 96 ± 3.8 4.1 ± 0.4 90 ± 3.4
MMS [µg/mL] 12.4 ± 0.6 ** 39 ± 5.7 ** 11.2 ± 2.4 ** 96 ± 4.9
Cells were treated with increasing concentrations of Al0, Al2O3 and ZnO NMs, and with the ionic salt
control AlCl3. MMS was used as a positive control (25 µg/ml for Caco-2 cells and 30 µg/ml for HepaRG
cells). Results are presented as means (±SEM) of the percentage of binucleated micronucleated cells
(BNMN) scored from 1000 binucleated cells per slide. Two slides per concentration were scored per
experiment. Viability was calculated by the replicative index (RI). Each concentration was tested in
duplicate,
n
= 3. The percentages of BNMN cells were compared using the one-way Pearson chi-square
test.***p<0.01.
Table 5: Effects of Al-containing NMs on cell growth and foci numbers in the CTA assay in Bhas-42 cells.
Cells were treated from day 1 to 4 (initiation assay) or from day 4 to 14 (promotion assay) with Al0 and
Al2O3 NMs, and with AlC13 and post-cultivated in fresh medium until Day 21. MCA (1 µg/mL) and TPA
(0.05 µg/ml) were included as positive controls. The mean of the cell growth (CG) and the number of
Page 30/39
transformed foci per well (foci/well) were measured from 3 and 6 replicate cultures respectively,
according to the OECD.
Page 31/39
 
Initiation assay Promotion assay
 
Chemical Concentration CG a Foci/well b CG a Foci/well b
   
Al 0 c (0.005% BSA) 100 5.3±1.5 100 8.7±3.1
0.03 μg/cm2 107 6.5±1.4 99 4.7±2.1 *
0.1 μg/cm2 105 5.3±2.4 97 0.7±0.8 *
0.3 μg/cm2 96 4.2±0.8 89 0.7±0.8 *
1 μg/cm2 62 2.7±1.6 * 83 Toxic d
3 μg/cm2 14 1.5±0.8 * 75 Toxic d
   
Al2O3 0 c (0.005% BSA) 100 5.3±1.5 100 8.7±3.1
0.3 μg/cm2 106 3.0±2.0 * 103 5.7±2.0 *
1 μg/cm2 94 3.3±1.5 * 95 7.0±1.3
3 μg/cm2 97 3.8±0.8 93 5.8±1.6 *
9 μg/cm2 74 3.5±1.9 83 4.0±1.5 *
28 μg/cm2 72 2.3±1.6 * 84 0.7±0.5 *
   
AlCl3 0 c (2.5% H2O) 100 4.7 ± 1.6 100 6.8 ±1. 7
3 μg/ml 96 4.7±1.0 96 Toxic d
9 μg/ml 78 4.3±2.4 88 Toxic d
28 μg/ml 16 4.0±1.3 80 Toxic d
   
MCA 0 c (0.1% DMSO) 100 5.8±1.9
1 µg/ml 17 14.7±2.9  
   
TPA 0 c (0.1% DMSO) 100 8.8±2.1
0.05 µg/ml 112 20.3 ±3.0
Page 32/39
   
a % of cell growth compared to that of vehicle control.
b Average number of transformed foci/well±SD.
c Vehicle control: nal vehicle concentration of the working culture media in parentheses.
d Absence of cells in well.
*
p
<0.05; Dunnett test,
vs
vehicle control.
p
<0.05;
t
-test,
vs
DMSO (the vehicle of MCA and TPA).
Figures
Figure 1
Page 33/39
Time evolution over 24 h of the well-bottom concentration of (a) Al0 NMs in DMEM; (b) Al2O3 NMs in
DMEM; (c) Al0 NMs in William’s medium; (d) Al2O3 NMs in Williams medium. Model parameters : bulk
NM concentration 250 µg.mL-1; T=37°C.
Figure 2
TEM images of differentiated Caco-2 cross-sections showing the uptake of Al0 NMs (B, C) at 21 µg/cm2
and Al2O3 NMs (D,E) at 39 µg/cm2, after 24 h treatment compared to the negative control (A).
Concentrations of 21 µg/cm2 Al0 NMs and 39 µg/cm2 Al2O3 NMs were used to ensure equivalent Al
content per well. C and E represent enlarged pictures from B and D respectively. C: Al0 NMs were rarely
Page 34/39
seen in proximity to the nucleus (full arrow). C, E: NMs are present either in small clusters in the
cytoplasm or near the nucleus (open arrow) either as large agglomerates integrated into lucent or dense
vesicles (notched arrows). Scale bar 1µm, C: Cytoplasm, N: nucleus.
Figure 3
TEM images of differentiated HepaRG cross sections showing the uptake of Al0 (B, C) at 21 µg/cm2 and
Al2O3 (D,E) at 39 µg/cm2, after 24 h cell treatment compared to the negative control (A). Concentrations
of 21 µg/cm2 Al0 NMs and 39 µg/cm2 Al2O3 NMs were used to ensure equivalent Al content per well. C
and E represent enlarged pictures from B and D respectively. C: Al0 NMs were rarely seen near the nucleus
Page 35/39
(full arrow). E: NMs are included in vesicles containing large agglomerates (notched arrows) or as dense
vesicles containing small clusters in the cytoplasm (open arrow) or as isolated clusters. Scale bar 1µm, C:
Cytoplasm, N: nucleus.
Figure 4
Effects of Al-containing NMs on cell numbers and active caspase-3 levels in differentiated Caco-2 and
HepaRG cells. Cells were treated for 24 h with Al0 NMs, Al2O3 NMs and AlCl3 as ionic salt control. ZnO
and DTX-2 (20 nM for Caco-2 cells, 15 nM for HepaRG) were used as positive controls. Cell numbers (A)
and active-caspase-3 (B) from automated image analysis are presented as fold changes relative to
untreated cells. Representative images of active caspase-3 (green) in the cytoplasm of HepaRG cells are
shown. C) negative control, D) Al0 NMs 80 µg/cm2, E) Al2O3 NMs 80 µg/cm2, F) AlCl3 128 µg/ml, G)
ZnO NMs 6 µg/cm2. Data are presented as the means ± SEM of 3 (Caco-2) or 4 (HepaRG) independent
experiments. ****p< 0.0001. White bar = 100 µm
Page 36/39
Figure 5
Effects of Al-containing NMs on ROS levels in differentiated Caco-2 and HepaRG cells. A) Cells were
treated for 24 h with Al0 NMs, Al2O3 NMs and AlCl3 as ionic salt control. ZnO and Menadione (MEN) (25
µM for HepaRG, 50 µM for Caco-2 cells) were used as positive controls. Representative images of ROS
detection (yellow) in the cytoplasm of HepaRG cells are shown. B) negative control, C) Al0 NMs 80
µg/cm2, D) Al2O3 NMs 80 µg/cm2, E) AlCl3 (128 µg/ml), F) ZnO NMs 6 µg/cm2. Data are presented as
the means ± SEM of 5 (Caco-2) or 6 (HepaRG) independent experiments. *p< 0.05, ***p<0.001. White bar
= 100 μm.
Page 37/39
Figure 6
Effects of Al-containing NMs on γH2AX level in Caco-2 and HepaRG Cells. A) Cells were treated for 24 h
with Al0 NMs, Al2O3 NMs and AlCl3 as ionic salt control, or positive controls (MMS at 60 or 30 µg/ml
respectively, and ZnO NMs). Representative images of γH2AX detection (red) in the nuclei of HepaRG
cells: B) negative control, C) Al0 NMs 80 µg/cm2, D) Al2O3 NMs 80 µg/cm2, E) AlCl3 128 µg/ml, F) ZnO
NMs 6 µg/cm2. Data are presented as the mean ± SEM of 3 independent experiments. *p< 0.05,
***p<0.001, ****p<0.0001. White bar=100 µm.
Page 38/39
Figure 7
Detection of DNA damage in differentiated Caco-2 and HepaRG cells treated 24 h with Al-containing NMs
with the alkaline and Fpg-modied comet assays. DNA damage (A, B) and oxidative DNA damage (C, D)
were assessed in differentiated Caco-2 and HepaRG cells treated for 24 h with Al0, Al2O3 and ZnO NMs,
and with the ionic salt control AlCl3. MMS was used as a positive control (30 µg/ml). Values are
presented as the mean percentage ± SEM of 3 independent experiments. *p < 0.05, **p < 0.01, ***p<0.001,
****p<0.0001.
Page 39/39
Figure 8
Interference of Al0 and Al2O3 NMs with the alkaline and Fpg-modied comet assays. The interference of
Al0 and Al2O3 NMs with DNA migration was assessed with untreated HepaRG cells. NMs were added in
LMP when cells are deposited on slides and compared with control (cells without addition of NMs. Values
are presented as the mean percentage ± SEM of 2 independent experiments.
Supplementary Files
This is a list of supplementary les associated with this preprint. Click to download.
SuppdatPFT.ppt
Article
Nanoparticles (NPs) are present in many daily life products with particular physical-chemical properties (size, density, porosity, geometry …) giving very interesting technological properties. Their use is continuously growing and NPs represent a new challenge in terms of risk assessment, consumers being multi-exposed. Toxic effects have already been identified such as oxidative stress, genotoxicity, inflammatory effects, and immune reactions, some of which are leading to carcinogenesis. Cancer is a complex phenomenon implying multiple modes of action and key events, and prevention strategies in cancer include a proper assessment of the properties of NPs. Therefore, introduction of new agents like NPs into the market creates fresh regulatory challenges for an adequate safety evaluation and requires new tools. The Cell Transformation Assay (CTA) is an in vitro test able of highlighting key events of characteristic phases in the cancer process, initiation and promotion. This review presents the development of this test and its use with NPs. The article underlines also the critical issues to address for assessing NPs carcinogenic properties and approaches for improving its relevance.
Article
Full-text available
The knowledge about a potential in vivo uptake and subsequent toxicological effects of aluminum (Al), especially in the nanoparticulate form, is still limited. This paper focuses on a three day oral gavage study with three different Al species in Sprague Dawley rats. The Al amount was investigated in major organs in order to determine the oral bioavailability and distribution. Al-containing nanoparticles (NMs composed of Al⁰ and aluminum oxide (Al2O3)) were administered at three different concentrations and soluble aluminum chloride (AlCl3·6H2O) was used as a reference control at one concentration. A microwave assisted acid digestion approach followed by inductively coupled plasma mass spectrometry (ICP-MS) analysis was developed to analyse the Al burden of individual organs. Special attention was paid on how the sample matrix affected the calibration procedure. After 3 days exposure, AlCl3·6H2O treated animals showed high Al levels in liver and intestine, while upon treatment with Al⁰ NMs significant amounts of Al were detected only in the latter. In contrast, following Al2O3 NMs treatment, Al was detected in all investigated organs with particular high concentrations in the spleen. A rapid absorption and systemic distribution of all three Al forms tested were found after 3-day oral exposure. The identified differences between Al⁰ and Al2O3 NMs point out that both, particle shape and surface composition could be key factors for Al biodistribution and accumulation.
Article
Full-text available
Due to several gaps remaining in the toxicological evaluation of nanomaterials (NMs), consumers and public health agencies have shown increasing concern for human health protection. In addition to aluminum (Al) microparticles, Al-containing nanomaterials (Al NMs) have been applied by food industry as additives and contact materials. Due to the limited amount of literature on the toxicity of Al NMs, this study aimed to evaluate the in vivo genotoxic potential of Al0 and Al2O3 NMs after acute oral exposure. Male Sprague-Dawley rats were administered three successive gavages at 6, 12.5 and 25 mg/kg bw. A comparison with AlCl3 was done in order to assess the potential effect of dissolution into Al ions. Both DNA strand breaks and oxidative DNA damage were investigated in six organs/tissues (duodenum, liver, kidney, spleen, blood and bone marrow) with the alkaline and the Fpg-modified comet assays. Concomitantly, chromosomal damage was investigated in bone marrow and colon with the micronucleus assay. The comet assay only showed DNA damage with Al2O3 NMs in bone marrow (BM), while AlCl3 induced slight but non-significant oxidative DNA damage in blood. No increase of chromosomal mutations was observed after treatment with the two Al MNs either in the BM or in the colons of rats.
Article
Full-text available
Aluminium is one of the most abundant elements in earth’s crust and its manifold uses result in an exposure of the population from many sources. Developmental toxicity, effects on the urinary tract and neurotoxicity are known effects of aluminium and its compounds. Here, we assessed the health risks resulting from total consumer exposure towards aluminium and various aluminium compounds, including contributions from foodstuffs, food additives, food contact materials (FCM), and cosmetic products. For the estimation of aluminium contents in foodstuff, data from the German “Pilot-Total-Diet-Study” were used, which was conducted as part of the European TDS-Exposure project. These were combined with consumption data from the German National Consumption Survey II to yield aluminium exposure via food for adults. It was found that the average weekly aluminium exposure resulting from food intake amounts to approx. 50% of the tolerable weekly intake (TWI) of 1 mg/kg body weight (bw)/week, derived by the European Food Safety Authority (EFSA). For children, data from the French “Infant Total Diet Study” and the “Second French Total Diet Study” were used to estimate aluminium exposure via food. As a result, the TWI can be exhausted or slightly exceeded—particularly for infants who are not exclusively breastfed and young children relying on specially adapted diets (e.g. soy-based, lactose free, hypoallergenic). When taking into account the overall aluminium exposure from foods, cosmetic products (cosmetics), pharmaceuticals and FCM from uncoated aluminium, a significant exceedance of the EFSA-derived TWI and even the PTWI of 2 mg/kg bw/week, derived by the Joint FAO/WHO Expert Committee on Food Additives, may occur. Specifically, high exposure levels were found for adolescents aged 11–14 years. Although exposure data were collected with special regard to the German population, it is also representative for European and comparable to international consumers. From a toxicological point of view, regular exceedance of the lifetime tolerable aluminium intake (TWI/PTWI) is undesirable, since this results in an increased risk for health impairments. Consequently, recommendations on how to reduce overall aluminium exposure are given.
Article
Full-text available
Polymerase chain reaction (PCR) is used as an in vitro model system of DNA replication to assess the genotoxicity of nanoparticles (NPs). Prior results showed that several types of NPs inhibited PCR efficiency and increased amplicon error frequency. In this study, we examined the effects of various metal oxide NPs on inhibiting PCR, using high- vs. low-fidelity DNA polymerases; we also examined NP-induced DNA mutation bias at the single nucleotide level. The effects of seven major types of metal oxide NPs (Fe2O3, ZnO, CeO2, Fe3O4, Al2O3, CuO, and TiO2) on PCR replication via a low-fidelity DNA polymerase (Ex Taq) and a high-fidelity DNA polymerase (Phusion) were tested. The successfully amplified PCR products were subsequently sequenced using high-throughput amplicon sequencing. Using consistent proportions of NPs and DNA, we found that the effects of NPs on PCR yield differed depending on the DNA polymerase. Specifically, the efficiency of the high-fidelity DNA polymerase (Phusion) was significantly inhibited by NPs during PCR; such inhibition was not evident in reactions with Ex Taq. Amplicon sequencing showed that the overall error rate of NP-amended PCR was not significantly different from that of PCR without NPs (p > 0.05), and NPs did not introduce single nucleotide polymorphisms during PCR. Thus, overall, NPs inhibited PCR amplification in a DNA polymerase-specific manner, but mutations were not introduced in the process.
Article
Full-text available
Aluminum oxide nanoparticles (Al2O3NPs) and zinc oxide nanoparticles (ZnONPs) have been involved in many industries and they are extensively abundant in many aspects of human life. Consequently, concerns have been raised about their potentially harmful effects. However the toxicities of Al2O3NPs and ZnONPs are well documented, the effect of co-exposure to both nanoparticles remains strictly obscure. Therefore, the present study was undertaken to address this issue. Four groups of male Wistar rats (10 rats each) were used; control, Al2O3NPs treated, ZnONPs treated and Co-treated groups. Rats were orally administered their respective treatment daily for 75 days. The effects of each nanoparticle alone or in combination were assessed at different levels including; hepatic and renal function, structure, and redox status, nuclear DNA fragmentation, hepatic expression of mitochondrial transcription factor A (mtTFA) gene and peroxisome proliferator-activated receptor gamma-coactivator 1α (PGC-1α), systemic inflammation, and hematologic parameters. The results confirmed the hepatorenal toxicities of each nanoparticle used at the level of all parameters with suppression of the hepatic expression of mtTFA and PGC-1α. The co-exposure to both nanoparticles results in synergistic effects. From these results, we can conclude that co-exposure to aluminum oxide nanoparticles and zinc oxide nanoparticles results in more pronounced hepatorenal toxicities and systemic inflammation.
Article
Full-text available
The application of appropriate analytical techniques is essential for nanomaterial (NM) characterization. In this study, we compared different analytical techniques for NM analysis. Regarding possible adverse health effects, ionic and particulate NM effects have to be taken into account. As NMs behave quite differently in physiological media, special attention was paid to techniques which are able to determine the biosolubility and complexation behavior of NMs. Representative NMs of similar size were selected: aluminum (Al⁰) and aluminum oxide (Al2O3), to compare the behavior of metal and metal oxides. In addition, titanium dioxide (TiO2) was investigated. Characterization techniques such as dynamic light scattering (DLS) and nanoparticle tracking analysis (NTA) were evaluated with respect to their suitability for fast characterization of nanoparticle dispersions regarding a particle's hydrodynamic diameter and size distribution. By application of inductively coupled plasma mass spectrometry in the single particle mode (SP-ICP-MS), individual nanoparticles were quantified and characterized regarding their size. SP-ICP-MS measurements were correlated with the information gained using other characterization techniques, i.e. transmission electron microscopy (TEM) and small angle X-ray scattering (SAXS). The particle surface as an important descriptor of NMs was analyzed by X-ray diffraction (XRD). NM impurities and their co-localization with biomolecules were determined by ion beam microscopy (IBM) and confocal Raman microscopy (CRM). We conclude advantages and disadvantages of the different techniques applied and suggest options for their complementation. Thus, this paper may serve as a practical guide to particle characterization techniques.
Article
Full-text available
Development and market introduction of new nanomaterials trigger the need for an adequate risk assessment of such products alongside suitable risk communication measures. Current application of classical and new nanomaterials is analyzed in context of regulatory requirements and standardization for chemicals, food and consumer products. The challenges of nanomaterial characterization as the main bottleneck of risk assessment and regulation are presented. In some areas, e.g., quantification of nanomaterials within complex matrices, the establishment and adaptation of analytical techniques such as laser ablation inductively coupled plasma mass spectrometry and others are potentially suited to meet the requirements. As an example, we here provide an approach for the reliable characterization of human exposure to nanomaterials resulting from food packaging. Furthermore, results of nanomaterial toxicity and ecotoxicity testing are discussed, with concluding key criteria such as solubility and fiber rigidity as important parameters to be considered in material development and regulation. Although an analysis of the public opinion has revealed a distinguished rating depending on the particular field of application, a rather positive perception of nanotechnology could be ascertained for the German public in general. An improvement of material characterization in both toxicological testing as well as end-product control was concluded as being the main obstacle to ensure not only safe use of materials, but also wide acceptance of this and any novel technology in the general public.
Article
The aim of this study was to investigate the genotoxicity of aluminum oxide (Al2O3), β-tricalcium phosphate (β-TCP) (Ca3(PO4)2), and zinc oxide (ZnO) nanoparticles (NPs) that were 4.175, 9.058, and 19.8 nm sized, respectively, on human peripheral blood lymphocytes using micronucleus (MN) and chromosome aberration (CA) techniques. Aluminum oxide and β-TCP NPs did not show genotoxic effects on human peripheral blood cultures in vitro, even at the highest concentrations; therefore, these materials may be suitable for use as biocompatible materials. It was observed that, even at a very low dose (≥12.5 ppm), ZnO NPs had led to genotoxicity. In addition, at high concentrations (500 ppm and above), ZnO NPs caused mortality of lymphocytes. For these reasons, it was concluded that ZnO NPs are not appropriate for using as a biocompatible biomaterial.
Article
TiO2 nanomaterials (NM) have a wide range of industrial applications, including their use in food products. The incorporation of these NMs in consumer products represents a clear concern for public health safety agencies and consumers, and further investigation of the potential impact of these products on human health is necessary. Indeed, since human oral exposure to TiO2 NMs is expected to increase in the years to come, there exist legitimate concerns about the risk assessment of these nanomaterials present in food products. A considerable amount of studies investigating the adverse effects of TiO2 NMs have focused on the genotoxic effects of these NMs, and more recently they have been classified by the International Agency for Research on Cancer (IARC) as carcinogen group 2B following inhalation studies (IARC, 2010). While numerous data are available for anatase or mixes of anatase/rutile forms, the toxicity and the genotoxicity of rutile TiO2 NMs have been rarely investigated. The aim of our study was therefore to investigate the cytotoxic and genotoxic effects of two rutile TiO2 NMs, differing in surface coating, NM103 (hydrophobic) and NM104 (hydrophilic), on intestinal and hepatic cell models. Following 3 or 24 h treatments with concentrations of TiO2 NMs from 1.2 to 80 μg/cm², we have assessed the genotoxicity of these NMs with γH2AX, alkaline comet assay and micronucleus (MN) assays. Cellular viability and effects on oxidative stress were also evaluated. Although TEM imaging demonstrated the presence of the two TiO2 NMs within the cytoplasm, no significant cytotoxic or genotoxic were observed in either cell model. We have also evaluated and taken into account a variety of potential sources of interference of NMs with cellular assays. TiO2 NMs present in the cytoplasm introduce uncertainty in the scoring of micronuclei, and therefore this assay is not recommended for the evaluation of the genotoxicity of TiO2 NMs, or other NMs demonstrating similar interference. The unique properties of TiO2 NMs introduce additional complexity for genotoxicity testing, and caution must be taken in order to obtain reliable results necessary for accurate hazard assessment.