You are on page 1of 27

Nanotoxicology, March 2010; 4(1): 15–41

Zinc oxide nanoparticles in modern sunscreens: An analysis of potential


exposure and hazard

MEGAN J. OSMOND, & MAXINE J. MCCALL

CSIRO Future Manufacturing Flagship, North Ryde, NSW, Australia


(Received 22 September 2009; accepted 20 November 2009)

Abstract
Sunscreens containing metal oxide nanoparticles appear transparent on the skin and provide excellent protection against
sunburn caused by UV radiation. While it is likely that nanoparticles remain on the surface of the skin of healthy adult humans,
Nanotoxicology Downloaded from informahealthcare.com by University of Toronto on 02/19/13

and thus are considered safe for use in sunscreens, there has been no comprehensive assessment of the impact on human health
from exposure to the metal oxide nanoparticles destined for use in sunscreens, either in the workplace during the
manufacturing process, in long-term use across a range of skin conditions, or upon release into the broader environment,
either accidentally or consequent of normal sunscreen use. In this review, we focus on zinc oxide nanoparticles destined for use
in modern sunscreens, and discuss the potential for human exposure and the health hazard at each stage of their manufacture
and use. We highlight where there is a need for further research.

Keywords: Nanotoxicology, nanoparticle, zinc oxide, sunscreen

Introduction impact at the terrestrial level, although the proportion


For personal use only.

of shorter wavelengths reaching the earth’s surface


Prolonged exposure to the sun may cause painful sun- may alter with the depletion of stratospheric ozone
burn. Research has established that over-exposure to attributed to anthropogenic activities (Frederick
ultraviolet (UV) radiation can lead to inflammation, and Snell 1988; Madronich et al. 1998). UVB
immunosuppression, premature photo-ageing, DNA (290–320 nm) is incompletely attenuated by the
damage, and photocarcinogenesis (Marrot and ozone layer such that the terrestrial UV spectrum
Meunier 2008). In addition to direct cellular damage, currently contains approximately 6% of wavelengths
exposure to UV radiation can generate reactive oxygen in this range (Diffey 2002). The remaining 94%
species (ROS) in skin cells, and lead to an impairment of comprise UVA wavelengths (320–400 nm), sub-
endogenous anti-oxidant defence systems (Darr and categorized into UVAI (340–400 nm) and UVAII
Fridovich 1994; Sander et al. 2004; Sakurai et al. 2005). (320–340 nm) (Diffey 2002; Matts 2006).
Protection from over-exposure to UV radiation is Penetration of UV radiation into human skin
encouraged and the use of sunscreen as part of a decreases rapidly with increasing epidermal depth
protection strategy is recommended (e.g., http:// (Bruls et al. 1984). As nucleic acids and proteins
www.cancer.org.au/Healthprofessionals/PositionState- most efficiently absorb wavelengths within the UVB
ments/sunsmart.htm). However, the rise and fall in the (and UVC) range, with relatively low absorbance at
use of para-aminobenzoic acid (PABA) (Wolf et al. wavelengths greater than 300 nm, UVB was initially
2001) exemplifies the potential duality of sun-protection thought to be the primary damaging component of
ingredients: they can attenuate the damaging effects of terrestrial UV radiation. However, more recently, it
over-exposure to UV radiation but may also elicit a range has been appreciated that wavelengths in the UVA
of side-effects that are not necessarily appreciated during region penetrate deeper into the epidermis than UVB,
the first generation of use. inducing oxidative stress linked to indirect DNA
UV wavelengths are broadly categorized by wave- damage and long-term photo-ageing (Forestier 2008;
length into three groups (UVA, UVB and UVC). Marrot and Meunier 2008). It is now accepted that
UVC (200–290 nm) is completely absorbed by the sunscreens must provide broad spectrum UV
ozone layer and therefore has negligible biological protection (Nash et al. 2006; Forestier 2008).

Correspondence: Megan Osmond, CSIRO Future Manufacturing Flagship, PO Box 184, North Ryde, NSW, 2113, Australia. E-mail: megan.osmond@csiro.au

ISSN 1743-5390 print/ISSN 1743-5404 online  2010 Informa UK Ltd.


DOI: 10.3109/17435390903502028
16 M.J. Osmond & M.J. McCall

There are two major classes of active ingredients any material depends both on the biological toxicity
used in modern sunscreens: ‘chemical’ and ‘physical’. associated with the intrinsic properties of the material,
Active ‘chemical’ ingredients typically are molecules and on the degree of exposure of an organism to it.
with an aromatic ring structure bearing two functional The hazard posed may change during the life cycle of
groups, one of which acts as an electron donor and the the material from its time of manufacture through to
other as an electron acceptor following the absorption its demise or transformation into other forms.
of UV radiation (Wolf et al. 2001; Maier and Korting Zinc oxide shows exceptional properties as an
2005). Active chemical ingredients include octyl- active physical ingredient in sunscreens. It absorbs
methoxycinnamate, butyl-methoxydibenzoylmethane UV more effectively than TiO2 over a broad range,
and 3-(4-methylbenzylidene) camphor. particularly in the UVA region (Mitchnick et al. 1999;
Active ‘physical’ ingredients typically are particles Pinnell et al. 2000), and has consequently been used
of the metal oxides, such as zinc oxide (ZnO) and as the sole active ingredient in some broad-spectrum
titanium dioxide (TiO2) having intrinsic UV absorb- sunscreens. In this review, we focus on the use of ZnO
ing properties. Sunscreens containing physical nanoparticles destined for use in modern sunscreens.
blockers of UV have been shown to be highly effective We assess the potential risk at each stage, from pro-
in protecting cells against UV-induced DNA damage duction to use of product and disposal (Figure 1). An
(Cayrol et al. 1999). In normal pigment size ranges effort has been made to identify the gaps in current
Nanotoxicology Downloaded from informahealthcare.com by University of Toronto on 02/19/13

(150–300 nm for TiO2 and 200–400 nm for ZnO) knowledge and consequent research priorities at each
these particles reflect and scatter light, making the stage.
sunscreens appear white. However, as particle sizes
decrease to smaller sub-micron (nano) dimensions
(typically between 20 and 150 nm for TiO2 and
Workplace exposure
40–100 nm for ZnO) they absorb and scatter UV
radiation, and largely absorb visible wavelengths Hazard identification and potential for exposure
(Wolf et al. 2001), making the sunscreens appear
transparent on skin and thus more aesthetically pleas- Controlled chemical reactions such as gas-phase or
ing. In addition to facilitating consumer acceptance, solution-phase syntheses can be used to manufacture
For personal use only.

nanosized metal oxide particles may also offer greater ZnO nanoparticles from precursors (Perl 1993;
UV protection compared to their microsized counter- Kleinwechter et al. 2002; Viswanathan and Gupta
parts, at least in the UVB region (Popov et al. 2005). 2003; Bauermann et al. 2006; Duffy et al. 2007).
A recent review of 308 sunscreen products revealed Alternatively, ZnO nanoparticles can be prepared
that 3.6% contained physical blockers alone for their using mechanical methods such as high energy attri-
active ingredients, 54.8% contained only chemical tion milling. Where destined for incorporation into
UV-absorbers, whilst the remaining 41.6% contained sunscreens, they may be prepared as powders or
a mixture of both (Wahie et al. 2007) suggesting that suspensions. Although these processes typically take
approximately half of the commercially-available place within closed systems, inherent in their overall
sunscreens in this study contained metal oxide par- production, including post-production processing
ticles as at least one of their active ingredients. Of and packaging, is the possibility that ZnO nanoparti-
these, TiO2 was contained in ~ 45%, ZnO < 5%, but cles may be released incidentally as ‘dust’ or as aerosol
the portion of this percentage containing nanoparti- droplets into the ambient air. Once in the air, they
cles was not reported. may circulate for some time before settling on work-
Currently, metal oxide nanoparticles have not been place surfaces or on workers themselves. Therefore,
comprehensively assessed in regard to potential there may be the potential for exposure of workers via
effects on human health, from exposure (accidental inhalation and ingestion (by swallowing airborne
or otherwise) in the workplace during nanoparticle nanoparticles), as well as possible dermal contact
production or exposure through use in commercial (Figure 1, left).
products (e.g., TiO2 or ZnO sunscreens), or for their Workplace variables that may influence the poten-
effects on ecosystems if released into the environ- tial for exposure during manufacture include the
ment. It has been reported that some nanoparticles emission source, air flow dynamics including move-
such as TiO2 and carbon black, when inhaled by rats ment, current and ventilation rate, and worker mobil-
or instilled in their lungs, can elicit a greater inflam- ity (Brouwer et al. 2004). Particle mobility and a
matory response on an equivalent mass basis than fine changing size distribution over time may also be
or bulk particles of the same material (Baggs et al. important factors in exposure estimates, as airborne
1997; Donaldson et al. 2000; Oberdörster 2000). nanoparticles are unlikely to remain as single particles
However, the risk of adverse impact on health from and may form homogenous or heterogeneous
Analysis of ZnO for sunscreens 17
Nanotoxicology Downloaded from informahealthcare.com by University of Toronto on 02/19/13
For personal use only.

Figure 1. Schematic showing potential exposure to zinc oxide nanoparticles destined for use in sunscreens, from their manufacture in the
workplace and their transport to be incorporated in sunscreen, through to their use by consumers. Possible points of exposure to humans and
the environment are highlighted in red, with unregulated exposure indicated as potential hot-spots.

agglomerates with themselves or other particles staff, cleaners and trades people may be exposed
(Brouwer et al. 2004; Maynard and Kuempel 2005; through circulating or settled nanoparticles.
Tsai et al. 2008). Particle size and composition can
influence the efficiency of deposition in the respiratory
tract as well as the biological impact, particularly as Likelihood of exposure
the lung environment may favor the stabilization of
agglomerates (Kendall et al. 2002). Assessment of the likelihood and degree of workplace
Given that even small amounts of nanoparticulate exposure has been limited by a lack of robust
material contain enormous numbers of individual techniques and procedures for obtaining relevant
particles, low-level chronic exposure may occur in dose metrics for inhalation. The heightened pro-
the workplace via inhalation or ingestion, or by other inflammatory effects of nanoparticles in general
means such as dermal absorption including the eyes. have most often been attributed to their greater sur-
The exposure of those directly involved in the face area relative to the same mass of larger particles
manufacturing process may be mitigated by imple- (Oberdörster 2000; Hohr et al. 2002; Hext et al. 2005;
menting a hierarchy of controls including process Monteiller et al. 2007), and the importance of surface
engineering for handling and storage, and the properties (Tabata and Ikada 1988; Fubini 1997),
provision and use of adequate personal protective particle size (Limbach et al. 2005), and shape at point
equipment (PPE). Depending on the levels of control of contact (Champion and Mitragotri 2006) have also
implemented, other personnel such as administrative been emphasized. Although instruments are available
18 M.J. Osmond & M.J. McCall

to characterize nanoparticulate aerosols on the basis bundles rather than dispersing, and therefore extra-
of mass, particle number or surface area, their effec- polation of levels of airborne particles from a model
tiveness may be limited as the use of static samplers using CNTs may underestimate levels for more dust-
may not accurately reflect personal exposure like nanoparticles such as ZnO.
(Brouwer et al. 2004). In addition, these instruments
cannot quantify specific nanoparticles against a gen-
eral, high background, and so current identification of Potential health effects of workplace exposure
exposure to specific nanoparticles relies on the col-
lection of airborne samples on filters that are subse- Inhalation. Depending on size, deposition of
quently analyzed by electron microscopy. Therefore, inhaled particles can occur in different areas of the
until there are technological advances in measuring respiratory tract, with theoretical modeling of
personal exposure to specific nanoparticles in the nanoparticles suggesting preferential deposition of
workplace, it is difficult to readily estimate the specific particles ~ 10–100 nm in the alveolar and tracheo-
likelihood and degree. bronchial regions, and even smaller nanoparticles
Information on the likelihood of workplace exposure in the nasopharynx region (Oberdörster 2001;
to nanoparticulate ZnO destined for sunscreen use Maynard and Kuempel 2005); however, deposition
during the production phase is not publically available efficiency in the latter region increases when
Nanotoxicology Downloaded from informahealthcare.com by University of Toronto on 02/19/13

but other studies using different nanoparticles may expressed as per unit surface area, as the nasopharynx
provide some insight. A recent workplace study has a smaller surface area than the alveolar region
(Tsai et al. 2008) was carried out during the produc- (Oberdörster 2001). In the former, alveolar macro-
tion of polymer-nanoalumina nanocomposites by twin phages generally phagocytose particles and migrate
screw extrusion (TSE), a commercial method by which via the mucociliary escalator to the tracheobronchial
nanoparticles and polymer melts are compounded. region, where they can be either coughed up or
Release of airborne nanoparticles during the com- swallowed. Lung lining fluid, containing antioxidants,
pounding process was assessed using a Fast Mobility lipids, mucins and proteins, provides further protec-
Particle Sizer (FMPSTM) spectrometer, placed at var- tion against inhaled particles (Kendall et al. 2002). If
ious points around the workplace during production. these mechanisms fail, particles can enter the inter-
For personal use only.

Workplace controls were not specifically described but stitium where they are no longer available for clear-
the presence of an exhaust system attached to the TSE ance via normal pathways. Their presence can then
instrument and a laboratory ventilation system were chronically stimulate cells, leading to inflammation,
noted. Elevated levels of nanoparticles in the ambient fibrosis and disease (Donaldson et al. 1998). Studies
air at the source of production and in the breathing with TiO2 and carbon black nanoparticles suggest that
zone were reported (Tsai et al. 2008). phagocytosis of these types of nanoparticles may be
An earlier workplace study (Maynard et al. 2004) sub-optimal (Churg et al. 1998; Renwick et al. 2001;
assessed the generation of aerosol particles during Geiser et al. 2008) and that nanosized particles may
post-production handling of single-walled carbon show long-term retention in the bronchial airways
nanotubes (SWCNTs) across four facilities. Particle (Kreyling et al. 2006).
handling was carried out in specially-constructed Studies with animal models have suggested that
clean air enclosures and air was monitored using a single exposures to ZnO nanoparticles as freshly gen-
portable condensation particle counter and a size- erated aerosols (60 nm) or in suspension via intra-
discriminating optical particle counter. Filter samples tracheal instillation (50–70 nm) can produce at least a
were also taken and cotton gloves worn by the workers transient inflammatory response (Gordon et al. 1992;
were used to assess dermal exposure. During the Sayes et al. 2007) (Table Ia). In a guinea pig model,
manufacturing process, when it was noted that work- repeated inhalation (3 h/day 6 day) of the industry-
ers took care to move with slow deliberation, airborne relevant dose of 5 mg/m3 of freshly generated ultrafine
nanoparticles were present at only low levels. These ZnO (50 nm) caused lung lesions characterized
levels increased during clean-up and when workers by infiltration of inflammatory cells, interstitial
entered or left the work area (Maynard et al. 2004), thickening, and diminished lung capacity (Lam et al.
indicating that increased circulation at these points 1985), although this response was not observed in a
may have disturbed surface-deposited nanoparticles. previous single-exposure study (Lam et al. 1982).
The study also reported that detectable levels of However, interspecies differences in the response to
SWCNTs deposited on workplace surfaces were inhaled particles in general (Gordon et al. 1992;
transferred to gloves worn by workers. It should Bermudez et al. 2002; Bermudez et al. 2004;
also be noted that due to their surface chemistries, Hext et al. 2005) can make it difficult to reliably
some CNTs can be particularly prone to forming extrapolate the results of these studies to humans.
Analysis of ZnO for sunscreens 19

For example, particle overload can occur in some with a companion study addressing zinc nano-
mammals leading to compromised macrophage- particles. Two weeks after 5 g/kg body weight zinc
mediated particle clearance and mobility, and sub- (not ZnO) nanoparticles (58 nm) and microparticles
sequent development of inflammation, fibrosis and (1.08 mm) were gastrointestinally administered to
cancer (Morrow 1988; Oberdörster et al. 1994; four-week-old mice, more severe anemia and renal
Nikula et al. 1997; Tran et al. 2000), but the rele- damage was observed in animals exposed to the
vance of particle overload in humans has been ques- nanoparticles compared to the microscale particles.
tioned (Borm et al. 2004). Histological examination of two mice that died in the
Metal fume fever, which is characterized by short- first week showed intestinal obstruction caused by
term pulmonary and systemic alterations in humans, aggregation of zinc nanoparticles in the intestine
is caused by the inhalation of freshly generated metal (Wang et al. 2006), although serum biomarkers of
oxide aerosol (Kelleher et al. 2000) and may be a inflammation generally did not differ appreciably
more relevant model for workplace exposure to ZnO between the two groups. In a follow up study also
nanoparticles. A single 10–30 min inhalation of a high in mice, treatment with 1–5 g/kg body weight 20 nm
dose of nanoparticulate ZnO aerosol containing and 120 nm ZnO particles showed relatively little
170 nm aggregates (from 8–40 nm primary particles) toxicity, although some pathological damage was
was sufficient to induce increases in levels of the detected in the stomach, liver, heart and spleen
Nanotoxicology Downloaded from informahealthcare.com by University of Toronto on 02/19/13

inflammatory cytokines interleukin (IL)-6, IL-8, (Wang et al. 2008). It is important to note that the
and tumor necrosis factor (TNF)-a in broncho- doses used in both of these studies were higher than
alveolar lavage (BAL) fluid within three hours of those to which most people are likely to be exposed
exposure in humans (Kuschner et al. 1997). and, when taken together, the studies showed little
A more likely scenario of workplace exposure, data consistent with increased oral toxicity of ZnO
however, might be chronic, low-level exposure. The nanoparticles.
industry threshold limit for human inhalation of ZnO
is 5 mg/m3 for up to 8 h, but it has been shown that
less than half this exposure can induce an adverse Dermal absorption. To our knowledge there have been
response in humans (Gordon et al. 1992; Fine et al. no studies investigating the hazard potential or fate of
For personal use only.

1997). However, humans appear to develop tolerance ZnO nanoparticle dust that may settle on exposed
to inhaled ZnO fumes (Fine et al. 2000), with no skin from the air or from physical contact with other
known long-term effects of metal fume fever, and surfaces. Some studies are available that have assessed
acquired tolerance to inhaled ZnO has also been nanoparticles suspended in emulsions or sunscreen
shown in mice (Wesselkamper et al. 2001a). formulations, but these will be considered in the
One study comparing the effects of ZnO particle Product use section.
size on inflammogenicity reported that there were no
differences in the response of healthy adults to inhaled
nanoparticulate (40 nm) or larger (291 nm) ZnO Potential for translocation
particles (Beckett et al. 2005), although it should
be noted that the dose used was below the threshold Lung and airways. A range of structural and immune
to detect a biological difference between inhaled ZnO cells comprise the barriers of the respiratory system
and filtered air and therefore might not be conclusive (Suzuki et al. 2008). Due to their small size, nano-
as to potential differences between nano-ZnO and particles may retard or evade phagocytosis by macro-
larger ZnO particles. phages thus giving them a greater opportunity to
Although ZnO has only low solubility in water, pH penetrate into the pulmonary interstitium (Churg
can influence solubility (Environmental Protection et al. 1998; Oberdörster 2001; Renwick et al. 2001;
Agency [EPA] 1995) and therefore a further consid- Borm and Kreyling 2004; Geiser et al. 2008;
eration is the possibility that ZnO nanoparticles may Lynch and Dawson 2008). Nanoparticles may also
dissolve in the more acidic environment of the lung form complexes with proteins such that the fate of
lining fluid, potentially leading to transient increases the nanoparticle may be determined by that of the
in the local Zn2+ concentration. The likelihood that protein (Kreyling et al. 2006; Semmler et al. 2004).
this occurs and its possible biological impact have not The biological impact of this ‘protein corona’
yet been investigated. may alter depending on particle size and surface
characteristics as both of these have been shown to
influence the constituents of the corona in human
Ingestion. Only one study has been published on the plasma using a range of polystyrene nanoparticles
effects of ingestion of ZnO nanoparticles (Table Ib), (Lundqvist et al. 2008).
Nanotoxicology Downloaded from informahealthcare.com by University of Toronto on 02/19/13
For personal use only.

20

Table I. A summary of published data relating to potential hazards of zinc oxide nanoparticles.

Particle surface
Authors Particle size (nm) area Surface coating Administration vehicle Experimental regime Results

(a) Inhalation
In vitro
Brunner et al. 19–36 57 m2/g NR1 Nanoparticles Human mesothelioma and Cell viability was reduced after
(2006) suspended in deionized rodent fibroblast cells exposure to 3.75 ppm and was
water then diluted in cell incubated with 1–30 ppm ZnO. ~ 100% at 15 ppm. Toxicity was
culture media. attributed to ZnO solubility.
M.J. Osmond & M.J. McCall

Gojova et al. 100–200 length; 21 m2/g NR Nanoparticles Human aortic endothelial cells Doses at or above 10ug/ml initiated
(2007) 20–70 width suspended in cell exposed to ZnO nanoparticle a dose-dependent inflammatory
culture medium. concentrations ranging from response and decrease in cell
0.001–50 mg/ml in culture viability. Internalization of ZnO
media for 4 h, with cell viability nanoparticles within intracellular
and inflammatory biomarkers vesicles was detected.
measured at 4 h and 24 h.
Gopalan et al. 40–70 NR NR Nanoparticles Primary human sperm cells and DNA damage occurred in sperm
(2009) suspended in DMSO, lymphocytes incubated with cells and lymphocytes compared to
filtered through a 11.5–92.3 mg/ml ZnO. a negative control, but ZnO was
0.2 mm syringe filter Incubation with the particles only weakly photogenotoxic.
and diluted in PBS. was performed in the dark, with
pre- or simultaneous irradiation
with UV light.
Jeng and 50–70 15–25 m2/g NR Nanoparticles Mouse neuroblastoma cells Cell viability decreased at a
Swanson (2006) suspended in deionized incubated for 48 h with threshold dose of 50 mg/ml.
water then added to cell 10–100 mg/ml ZnO particles in
culture media. culture media.
In vivo2
Beckett et al. 40 & 291 NR NR Freshly generated ZnO Inhaltion exposure of human No differences were observed
(2005) vapor diluted in air. subjects to air containing between filtered air and either ZnO
500 mg3 nano- or microparticles sample. The concentration used
ZnO for 2 h with physiological was below the threshold required
endpoints assessed 24 h post- for an acute response.
exposure.
Nanotoxicology Downloaded from informahealthcare.com by University of Toronto on 02/19/13
For personal use only.

Table I (Continued)

Particle surface
Authors Particle size (nm) area Surface coating Administration vehicle Experimental regime Results

Fine et al. (1997) 300 (secondary NR NR Freshly generated ZnO Inhalation exposure of human Symptoms of metal fume fever and
aggregates from vapor diluted in filtered subjects to air containing 0, 2.5 plasma levels of IL-6 were elevated
primary air. and 5 mg/m3 ZnO for 2 h on post-inhalation.
nanoparticles) separate days. Venous blood
was withdrawn 3 and 6 h
post-exposure and subjects
recorded symptoms 0, 3, 6
and 9 h post-exposure.
Fine et al. (2000) 300 (secondary NR NR Freshly generated ZnO Inhalation exposure of naïve Inflammatory biomarkers and
aggregates from vapor diluted in filtered and chronically exposed symptoms of metal fume fever
primary air. human subjects to air initially increased in naïve subjects
nanoparticles) containing 5 mg/m3 ZnO but tolerance was induced by
for 2 h on 1 or 3 days. repeat exposures. Chronically
Bronchoalveolar lavage was exposed subjects reported fewer
performed 20 h post-exposure. symptoms than naïve subjects, but
Venous blood was withdrawn still had elevated levels of plasma
3 and 6 h after each exposure. IL-6 following exposure.
Subjects also recorded
symptoms 0, 3, 6 and 9 h
post-final exposure.
Gordon et al. 60 (primary) NR NR Freshly generated ZnO Inhalation exposure of guinea Dose-dependent elevation of
(1992) vapor diluted in filtered pigs, rats and rabbits to air inflammatory biomarkers as well as
air. containing 2.5 or 5 mg/m3 ZnO increased ZnO particle retention
particles for 3 h. Lungs were was shown in rats and guinea pigs
lavaged 0, 4 or 24 h after but not in rabbits.
treatment. Inhalation exposure Human subjects reported
of human subjects to air symptoms of metal fume fever
containing within 4–8 h of exposure although
5 mg/m3 ZnO particles for 2 h no differences in pulmonary
with lung function assessed function were detected
immediately afterwards and immediately post-exposure.
recorded symptoms 24 h
post-exposure.
Kuschner et al. 170 (secondary NR NR Freshly generated ZnO Inhalation exposure of human Biomarkers for pulmonary
(1997) aggregates from vapor diluted in air. subjects to air containing an inflammation were elevated in
8–40 nm primary average of 33 mg/m3 for 10–30 subjects 3 h post-exposure.
nanoparticles) min. Bronchoalveolar lavage
was performed 3 or 20 h
post-exposure.
Analysis of ZnO for sunscreens
21
Nanotoxicology Downloaded from informahealthcare.com by University of Toronto on 02/19/13
For personal use only.

22

Table I (Continued)

Particle surface
Authors Particle size (nm) area Surface coating Administration vehicle Experimental regime Results

Lam et al. (1982) 50 NR NR Freshly generated ZnO Inhalation exposure of guinea Most parameters of lung function
vapor diluted in air. pigs to air containing 7.8 mg/ assessed were not significantly
m3 ZnO particles alone, or influenced by single 3 h exposure
0.8–6 mg/m3 ZnO in to ZnO alone but were impaired in
combination with SiO2 for 3 h. animals treated with the ZnO in
Lung function was assessed combination with SiO2 in a
within 2 h of treatment. dose-dependent manner.
Lam et al. (1985) 50 NR NR Freshly generated ZnO Inhalation exposure of guinea Lung function was decreased and
vapor diluted in air. pigs to air containing 5 mg/m3 markers for pulmonary
M.J. Osmond & M.J. McCall

ZnO particles 3 h/day for inflammation increased following


6 days. Lung function and treatment and persisted up to 72 h
morphology were assessed at 1, post-inhalation.
24, 48 or 72 h post-inhalation.
Sayes et al. 50–70 12.1 NR Powder suspended in Intratracheal instillation of 1 or ZnO fine and nanoparticles caused
(2007) Mili-Q water, PBS or 5 mg/kg fine or nano-ZnO in a transient inflammatory response
cell culture media rats with lung inflammatory in vivo in rats. Results of
biomarkers assessed 24 h, comparative in vitro studies were
1 wk, 1 month or 3 months not always predictive of the in vivo
post-exposure. response.
Wesselkamper 300 (secondary NR NR Freshly generated ZnO Inhalation exposure of mice to Inflammatory biomarkers in the
et al. (2001a) aggregates from vapor diluted in filtered air containing 1 mg/m3 ZnO lung decreased with repeated
primary air. particles for 3 h/d for 1, 3 or 5 exposures, however total protein
nanoparticles) successive days. A subset of levels and lung pathology were
mice was exposed for 5 higher with increasing exposure,
successive days, followed by and an increase in metallothionein
re-exposure after 5 d rest. mRNA was also observed.
Bronchoalveolar lavage and Acquired tolerance was lost when
morphological assessment exposure was not on consecutive
performed 24 h post-exposure. days.
Wesselkamper 300 (secondary NR NR Freshly generated ZnO Inhalation exposure of Acquired pulmonary tolerance to
et al. (2001b) aggregates from vapor diluted in filtered 11 inbred mouse strains to inhaled ZnO fumes has a genetic
primary air. 1 mg/m2 ZnO for 3 h/day for component in mice.
nanoparticles) 1 or 5 consecutive days.
Bronchoalveolar lavage was
performed 24 h post-exposure.
Nanotoxicology Downloaded from informahealthcare.com by University of Toronto on 02/19/13
For personal use only.

Table I (Continued)

Particle surface
Authors Particle size (nm) area Surface coating Administration vehicle Experimental regime Results

(b) Ingestion
In vitro
No data available
In vivo
Wang et al. 20 & 120 4.4  105 & 9.1 NR Powder suspended in Nanoparticle suspension Little difference in toxicity between
(2008)  104 cm2/g 1% sodium carboxy administered 20 and 120 nm ZnO particles was
methyl cellulose. gastrointestianally into mice at observed, but a lower dose of the
1-, 2-, 3-, 4-, and 20 nm particles induced greater
5-g/kg body weight. pathology than higher doses.
Liver, spleen heart, pancreas and
bone were identified as target
organs for ZnO particles.
(c) Dermal
In vitro
Dufour et al. 100 (average) NR Uncoated Stock solutions of ZnO Chinese hamster ovary cells Cells were more sensitive to ZnO-
(2006) powder added to cell incubated with ZnO under dark induced clastogenic effects with
culture medium. conditions, or pre- or pre- or simultaneous UV
simultaneously irradiated with irradiation, but ZnO itself was not
UV light then assessed for strongly photo-clastogenic.
DNA damage.
Hidaka et al. NR NR NR ZnO particles extracted 1 mg/ml extracted particles ZnO particles extracted from
(2006) from sunscreens and incubated in vitro with plasmid sunscreens were found to be highly
suspended in deionized DNA in the presence of UV photoactive and caused rapid DNA
water. light and assessed for DNA damage under in vitro conditions.
strand breakage.
Rampaul et al. 14–27 NR Uncoated or dimethicone ZnO particles washed Photobleaching of methylene ZnO particles extracted from
(2007) from sunscreen or blue by ZnO extracted particles commercially available sunscreens
obtained directly from was assessed under light and showed photocatalytic activity
the manufacturer. dark conditions. comparable to uncoated TiO2.
Analysis of ZnO for sunscreens
23
Nanotoxicology Downloaded from informahealthcare.com by University of Toronto on 02/19/13
For personal use only.

24

Table I (Continued)

Particle surface
Authors Particle size (nm) area Surface coating Administration vehicle Experimental regime Results

Yang et al. 20 NR NR Powder suspended in Primary mouse embryo ZnO nanoparticles exerted
(2008) FBS then diluted in cell fibroblast cells incubated with comparatively more cytotoxic and
culture medium. ZnO nanoparticles, carbon oxidative stress on cells than other
black, single-walled carbon nanoparticles measured, but was
nanotubes, and silicon dioxide less genotoxic than single-walled
nanoparticles. carbon nanotubes. Cytotoxicic
effects were attributed to particle
M.J. Osmond & M.J. McCall

composition while genotoxicity


was attributed to particle shape.
Ex vivo
Cross et al. 15–30 NR Polymethylsilsesquioxane 60% ZnO in caprylic Sunscreen applied to full- Negligible penetration through the
(2007) capric triglyceride or thickness human abdominal stratum corneum.
20% ZnO in caprylic skin sections mounted on
capric triglyceride diffusion cells for
incorporated into 24 h. Also viewed by electron
sunscreen formulation. microscopy.
Dussert et al. 117 NR NR 60% dispersion of Sunscreen applied to human No penetration through stratum
(1997) zinc oxide in mineral abdominal skin and viewed by corneum.
oil/triglyceride electron microscopy.
incorporated into
sunscreen formulation.
Gamer et al. 80, with 90% NR Uncoated 10.3% wt ZnO in Emulsion applied to full- Negligible penetration through the
(2006) < 160 nm oil/water emulsion. thickness porcine abdominal stratum corneum.
skin samples mounted on
diffusion cells for
24 h. Skin stripping was also
performed.
Kuo et al. (2009) 10 NR Uncoated Nanoparticles Single application of ZnO nanoparticles detected
suspended in solutions nanoparticle solution onto 20–30 mm from the skin surface
of PBS, oleic acid, hairless mouse dorsal skin when applied to skin samples in the
ethanol or oleic samples mounted on diffusion presence of oleic acid and/or
acid-ethanol. cells and viewed by ethanol but did not penetrate
multiphoton microscopy. through stratum corneum in
control solution (PBS).
Nanotoxicology Downloaded from informahealthcare.com by University of Toronto on 02/19/13
For personal use only.

Table I (Continued)

Particle surface
Authors Particle size (nm) area Surface coating Administration vehicle Experimental regime Results

In vivo
Zvyagin et al. 26–30 NR NR Commercially available One application to forearm, ZnO nanoparticles settled into skin
(2008) sun protective cheek, shoulder and feet of four folds and hair follicles but did not
formulation. subjects of different ethnic penetrate through the stratum
background. Penetration corneum.
assessed by multiphoton
microscopy and SEM/EDX. Ex
vivo human skin samples also
assessed.
Roberts et al. 26–30 NR Polymethylsilsesquioxane Commercially available One application of formulation ZnO nanoparticles found in skin
(2008) suspension of coated rubbed into forearms of 60 folds but no penetration into viable
ZnO nanoparticles in human volunteers of different epidermis.
caprylic/capric age, race and sex for 5 min.
triglycerides. Penetration assessed by
multiphoton microscopy and
SEM/EDX.
Gulson et al. 20 & > 100 NR Uncoated 18% wt 68ZnO in Sunscreens containing Not yet available.
(2009) sunscreen formulation. traceable 68ZnO applied to
human skin twice daily for five
days with exposure to water and
UV radiation.
(d) Environment
Adams et al. 480 nm–4 mm NR NR Powder suspended in E. coli and B. subtilis incubated B. subtilis more sensitive to ZnO
(2006a) (in suspension) Milli-Q water and with 10–5000 ppm overnight than E. coli, with negligible
diluted in liquid under light or dark conditions difference in the presence of
bacterial media. then plated on agar and sunlight compared to dark
colonies counted. incubation for B. subtilis, but more
pronounced light-induced growth
inhibition observed for E. coli.
Adams et al. 480 nm–4 mm NR NR Powder suspended in E. coli or B. subtilis incubated Nanoparticles formed micron-
(2006b) (in suspension) Milli-Q water and with 10–5000 ppm nano- or sized aggregates in suspension and
diluted in bacterial bulk-sized in sunlight or dark were not significantly more toxic to
media. conditions. Cultures were gram-positive or gram-negative
plated on agar and colonies bacteria than particles with a
counted. primary size in the micron-range.
D. magna exposed to B subtilis was more sensitive to ZnO
0.2–1 ppm ZnO in than E. coli. Toxicity was observed
spring water. for B. subtilis under both light and
Analysis of ZnO for sunscreens

dark conditions but E. coli was less


sensitive under dark conditions
25
Nanotoxicology Downloaded from informahealthcare.com by University of Toronto on 02/19/13
For personal use only.

26

Table I (Continued)

Particle surface
Authors Particle size (nm) area Surface coating Administration vehicle Experimental regime Results

than light. All concentrations used


showed toxicity towards D. magna.
Aruoja et al. 30 NR NR Powder suspended in OECD 201 algal growth Bulk and nano ZnO showed similar
(2009) algal medium. inhibition test guidelines toxicity to P. subcapitata and
followed to assess toxicity of toxicity was attributed largely to
ZnO nanoparticles to dissolved zinc ions.
P. subcapitata.
M.J. Osmond & M.J. McCall

Brayner et al. 8–14 NR NR Particles ± various Bacteriological tests on E. coli Concentration dependent
(2006) dispersing agents on solid agar plates with varying bactericidal activity observed, with
suspended in bacterial concentrations of ZnO internalisation of particles and
medium. nanoparticles. disruption of cellular membranes.
Franklin et al. 30 NR NR Stock solutions P. subcapitata incubated with a Nanoparticles were no more toxic
(2007) prepared in algal test range of ZnO concentrations in to P. subcapitata than bulk ZnO or
medium. algal media and cell counts dissolved ZnCl2. Toxicity
obtained. attributed to dissolved zinc ions.
Heinlaan et al. 50–70 NR NR Powder suspended in V. fischeri, D. magna and Toxicity and growth inhibition was
(2008) Milli-Q water and T. platyurus incubated with detected for all ZnO preparations,
diluted in test media. bulk and nano-sized ZnO. with negligible difference between
bulk and nano ZnO, and zinc ions
Toxicity was attributed to
dissolved zinc ions.
Huang et al. 60 NR Polyvinyl alcohol Particles suspended in S. agalactiae and S. aureus Concentration-dependent
(2008) aqueous ethylene glycol incubated with ZnO bactericidal activity and damage to
media and diluted in nanoparticles in solid and cellular membranes.
bacterial medium. liquid media.
Jiang et al. (2009) 1448 (secondary NR NR Powder suspended in Bacteria incubated with ZnO in ZnO nanoparticles were more toxic
aggregates from filter-sterilized liquid media then plated on to gram positive and negative
primary 20 nm deionized water agar and colonies counted. bacteria compared to bulk ZnO,
nanoparticles) then diluted in 1 g/l with toxicity attributed largely to
NaCl. particle exposure rather than
dissolved zinc ions.
Jones et al. 50–70 NR NR Powder suspended in Gram-positive and negative Toxicity observed towards gram
(2008) sterile double-distilled bacteria incubated with ZnO in negative and gram positive
water. liquid media then plated on bacteria, with greater growth
agar and colonies counted. inhibition observed with
decreasing particle size.
Nanotoxicology Downloaded from informahealthcare.com by University of Toronto on 02/19/13
For personal use only.

Table I (Continued)

Particle surface
Authors Particle size (nm) area Surface coating Administration vehicle Experimental regime Results
2
Lin and Xing 20 58 m /g NR Powder suspended in Seeds from six higher plant Exposure to ZnO nanoparticles
(2007) deionized water. species incubated in decreased seed germination and
nanoparticle or control root elongation in a number of
suspensions then germinated higher plant species.
on filter paper under dark
incubation conditions.
Reddy et al. 13 NR NR Powder suspended in E. coli and S. aureus grown on Concentration-dependent toxicity
(2007) liquid or solid bacterial solid media containing ZnO to gram negative and gram positive
media. nanoparticles or incubated in bacteria at lower doses than
liquid media containing 0–10 primary human T lymphocytes.
mM ZnO nanoparticles.
Yamamoto 100–800 26–0.85 m2/g NR Powder suspended in Bacteria incubated with ZnO Toxicity increased with decreasing
(2001) saline. nanoparticles across a range of particle size and increasing
concentrations. concentration.
Zhu et al. (2008) 180 (in NR NR Powder suspended in Zebrafish 96-h embryo-larval Concentration dependent toxicity
suspension, Milli-Q water. bioassay was used to assess largely attributed to dissolved zinc
from 20 nm toxicity of ZnO to D. rerio with ions, but residual toxicity was
nanoparticles) toxicological endpoints attributed to nanoparticles
assessed within 96 h exposure. themselves.
Zhu et al. (2009) 20 ‡ 90 m2/g Uncoated Powder suspended in Developing zebrafish incubated ZnO nanoparticles in water settle
zebrafish culture with settled aggregates of ZnO out of water column into
medium. nanoparticles for 96 h followed aggregates within 48 h. Dose
by assessment of developmental dependent toxicity to zebrafish
endpoints. attributed to combination of intact
ZnO nanoparticles and dissolved
Zn2+. Toxicity of nanoparticles was
mitigated in the presence of
formulated sediment.
1
NR, Not reported.
2
Only a subset of published literature associated with metal fume fever has been included here.
Analysis of ZnO for sunscreens
27
28 M.J. Osmond & M.J. McCall

Particle retention and response in animals may secondary organs dependent on size and surface
not necessarily be predictive of that in humans characteristics (Hussain et al. 2001; reviewed by
(Nikula et al. 1997; Nemmar et al. 2004) and the Hagens et al. 2007). However, data showing the
majority of translocation studies performed in animals specific translocation of ZnO nanoparticles from the
have not specifically investigated ZnO. Nevertheless, gastrointestinal tract are not available although ZnO
a number of these studies have indicated that other nanoparticles have been shown to have only low
types of inhaled nanoparticles such as carbon, man- solubility in artificial gastric fluid (Wang et al. 2008).
ganese oxide and silver can translocate relatively
swiftly from the lung to extrapulmonary organs, the
central nervous system, and lung-associated lymph Is more research required?
nodes (Takenaka et al. 2001; Oberdörster et al. 2002;
Elder et al. 2006). TiO2 and gold nanoparticles have A range of epidemiological data exists for the health
also been shown to penetrate into red blood cells effects and biomarkers for human exposure to ultra-
in vitro in a size-dependent manner (Rothen- fine combustion particles via air pollution (Lewtas
Rutischauser et al. 2006), with the subsequent 2007), which may provide some insight into the
biodistribution of gold nanoparticles after intravenous possible effects of inhaled manufactured nanoparti-
administration also showing size-dependence (de cles in general. However, we can find scant informa-
Nanotoxicology Downloaded from informahealthcare.com by University of Toronto on 02/19/13

Jong et al. 2008). Additionally, nanoparticles can tion relevant to potential workplace hazards from
permeate the blood-brain barrier depending on sur- inhalation, dermal exposure to dust, or oral ingestion
face characteristics and charge, as well as concentra- posed by ZnO nanoparticles manufactured for
tion (Kreuter 2004; Lockman et al. 2004). In contrast sunscreens (Table Ia, Ib). The number of studies
to these studies, a much lower proportion of systemic investigating the possible translocation of nanoparti-
uptake and translocation to extrapulmonary organs cles in general from lungs or the gastrointestinal tract
of insoluble iridium nanoparticles was reported into systemic circulation is also small and is non-
in rats, reaching a maximum after one week and existent for ZnO nanoparticles. While it is known
steadily clearing thereafter (Kreyling et al. 2002; that inhalation of ZnO aerosols containing nanopar-
Semmler et al. 2004).
For personal use only.

ticles can lead to adverse health effects in humans,


In humans, particle translocation from lungs after specific research into the potential translocation and
inhalation has not specifically been investigated using secondary health effects of ZnO nanoparticles is lack-
ZnO nanoparticles. In studies addressing other types ing. Currently, one must extrapolate from data
of nanoparticles, inhaled radioactive carbon nanopar- obtained for other kinds of nanoparticles, which is
ticles were reported to rapidly translocate from the problematic given not all nanoparticles exhibit the
lung into systemic circulation (Nemmar et al. 2002), same behaviors and toxicity. Further research is
although the stability of the label in biological fluids required to clarify if translocation occurs, and whether
has been questioned making it difficult to distinguish is likely to be only at low levels and subject to clear-
between translocation of free label and particle-bound ance mechanisms. Specifically, research priorities
label (Wiebert et al. 2006b). Later studies, using more should include: (i) whether workers manufacturing
stable radio-labeled carbonaceous particles in healthy or handling ZnO nanoparticles are at any risk from
or asthmatic humans, showed little evidence of trans- accidental or incidental exposure in the workplace;
location from the lungs to systemic circulation (ii) whether inhaled ZnO nanoparticles in the pulmo-
(Brown et al. 2002; Wiebert et al. 2006a, 2006b). nary or gastrointestinal systems remain as particles or
However, even if the rate of uptake and/or transloca- dissolve in biological fluids; (iii) whether ZnO nano-
tion is low and subject to subsequent clearance, over particles in the body translocate from their primary
periods of chronic and sustained exposure particle site of entry; and (iv) whether they are subject to
accumulation may occur (Kreyling et al. 2006). adequate clearance mechanisms in the body.

Gastrointestinal tract. Orally-ingested, solid micro-


Transport and storage of ZnO nanoparticles
particles that do not dissolve can pass between
prepared for sunscreens
the epithelial cells of the digestive tract and translocate
throughout the body via the lymph or circu- Hazard identification and potential for exposure
latory systems, eventually being eliminated in urine
(Volkheimer 1974). Some types of nanoparticles can After production and packaging, ZnO nanoparticles
pass through the gastrointestinal mucosa and enter the typically are transported from the site of manufacture
bloodstream, where they may undergo translocation to to the site of their formulation into sunscreen. At this
Analysis of ZnO for sunscreens 29

stage they may be in the form of a powder or dispersed those encountered during the manufacture of the
in an oil or aqueous medium depending on their nanoparticles, and they will not be reiterated here.
surface coating. Assuming the nanoparticles are
enclosed within appropriate packaging, their transport
and storage would likely involve only low risk of Product use
personal exposure or release into the environment,
apart from the potential risk of accidental spillage Hazard identification and potential for exposure
(Figure 1, left). It is difficult to assess the possible
differences in hazard potential when transporting Percutaneous absorption of topically applied sub-
nanoparticulate forms of ZnO compared to non- stances can be divided into two stages: penetration
nano due to a lack of experience and knowledge. through the epidermal layers and absorption into the
During sunscreen formulation (Figure 1, centre), blood stream (Harvell and Maibach 1994). The func-
the hazard and potential exposure for workers from tion of the epidermis (Figure 2) is to minimize water
accidental or incidental release of ZnO nanoparticles loss whilst maintaining a barrier against external
as powder or nanoparticulate aerosol are similar to organisms and particles (Goldstein and Abramovits
Nanotoxicology Downloaded from informahealthcare.com by University of Toronto on 02/19/13
For personal use only.

Stratum corneum
Stratum lucidum

Epidermis
Stratum granulosum
Stratum spinosum

Stratum basale
Hair shaft

Sebaceous gland

Bulge
Dermis

Matrix cells

Melanocytes

Dermal papilla

Blood vessels
Sub cutaneous

Connective tissue
Adipose tissue

Figure 2. Schematic of the structure of human skin. The epidermis consists of five thin layers, with the outmost layer, the stratum corneum,
largely providing the vital barrier function of the skin. Hair follicles are embedded in the skin, well into the viable dermis. The outer sheath of
the hair follicle is continuous with the epidermis. Blood vessels feed the skin cells as well as the hair follicle.
30 M.J. Osmond & M.J. McCall

2003; Elias 2005; Brown et al. 2006). The outermost Titanium dioxide nanoparticles (Lademann et al.
layer, the stratum corneum, presents the most formi- 1999; Nanoderm 2007) and ZnO nanoparticles
dable penetration barrier (Benson 2005) and is a (Roberts et al. 2008; Zvyagin et al. 2008) in sun-
relatively impermeable two-compartment system of screens have been shown to settle into skin furrows
10–15 layers of keratinocyte-rich corneocyte cells and hair follicles, and it has been assumed that the
embedded within a lipid-rich extracellular matrix majority in the latter locations are most likely
(Elias 1983; Benson 2005). This tissue is a self- displaced by sebum production and hair growth
renewing layer, with cells moving up from the deeper (Nohynek et al. 2007). Interestingly, though, hair
layers into the upper layers of the epidermis, differ- follicles are being investigated as a target site for
entiating and losing transcriptional and metabolic delivery of drugs (Lademann et al. 2005; Vogt et
activity, eventually reaching the stratum corneum as al. 2005). Although the outer root sheath of a hair
non-viable, protein-dense corneocytes (Goldstein and follicle is continuous with the epidermis, very few
Abramovits 2003). ZnO nanoparticles contact this differentiated corneocytes are present in the lower
surface layer during normal use of sunscreens follicular infundibulum and consequently the
containing them (Figure 1, right). However, the epidermis is relatively permeable at this point (Vogt
effects of different skin ages, skin types, or health et al. 2005). Vogt et al. (2006) showed that 40 nm
conditions on the potential for deep penetration of fluorescent beads, but not larger particles, penetrated
Nanotoxicology Downloaded from informahealthcare.com by University of Toronto on 02/19/13

ZnO nanoparticles from sunscreens have not yet been deeply into the hair follicle, although they were not
established, and so it is not known if they can reach detected below the base of the follicle in the dermis
the deeper living tissues under some conditions, and, (Vogt et al. 2006). The penetrability of a hair follicle
if so, what the impact of that penetration might be. can be influenced by sebum production and/or hair
A number of characteristics of the stratum corneum growth (Lademann et al. 2001) as well as exfoliation
can alter across a lifetime (Cerimele et al. 1990; (Otberg et al. 2004). When considering the potential
Hermanns-Le et al. 2004; Waller and Maibach routes of entry of ZnO nanoparticles through the
2005). The degree to which innate barrier function epidermis, it is important to consider typical human
may become impaired with age is equivocal as data on behaviors prior to the application of sunscreens. For
this subject are contradictory (Cerimele et al. 1990; example, skin on the face or legs may be exfoliated,
For personal use only.

Harvell and Maibach 1994) but it is recognized that shaved or waxed just prior to sunscreen application. If
barrier integrity can be more easily disrupted in the hair follicles are a site of deposition of nanoparticles
aged compared to the young, and that recovery occurs and are additionally vulnerable to penetration under
more slowly (Ghadially et al. 1995). Skin penetrability some circumstances, then they may provide a route of
can also show large intra- and inter-individual vari- entry for nanoparticles in sunscreens to reach living
ability (Hostynek 2003). For example, human skin skin cells.
may be more susceptible to penetration in areas of the It is also relevant to consider the potential impact of
body where it is thinner, or at sites of flexing, such as incidental ingestion of low levels of sunscreen from
joints (Tinkle et al. 2003; Rouse et al. 2007). Con- hand to mouth transfer, and also from the use of ZnO
sequently, barrier disruption can be influenced by nanoparticles in sun-protective lip balms. These
anatomical location (Breternitz et al. 2007). Stratum exposures may make the gastrointestinal tract a
corneum pH is also integral to normal skin-barrier secondary route of exposure to ZnO nanoparticles
function (Hachem et al. 2003) and can vary in an age in sunscreen, albeit most likely at very low levels
(Fluhr et al. 2004; Choi et al. 2007), gender and site- (Figure 1, right).
specific manner (Wagner et al. 2003). The presence of
skin pathologies such as atopic dermatitis can also
impair barrier function (Bouwstra and Ponec 2006). Likelihood of exposure
Additionally, exposure of skin to a threshold UV dose
may cause a delayed, temporary decrease in its barrier Particles of ZnO in sunscreen are not expected to
function (Haratake et al. 1997; Gelis et al. 2002; Jiang penetrate the stratum corneum (Rostan et al. 2002)
et al. 2007; Mortensen et al. 2008) and alterations in and the majority of studies on samples of healthy skin
humidity and hydration status can also lead to a conducted to date support this view (Nohynek et al.
temporary decline in skin barrier function (Zhai 2007; Environmental Working Group [EWG] 2009)
and Maibach 2001; Sato et al. 2002; Benson 2005; (Table Ic). In humans, the percutaneous absorption
Elias 2005). Many of these factors are likely to be of zinc from ZnO (size not given), following 72-h
relevant to the assessment of skin penetration by metal topical application of ZnO emulsions or ointments
oxide particles, as consumers use sunscreens out- onto ex vivo skin samples, was shown to be less than
doors and often in aqueous environments. 2% of the applied dose, although still detectable
Analysis of ZnO for sunscreens 31

irrespective of the vehicle (Pirot et al. 1996). Within a used in ZnO (and TiO2) nanoparticle penetration
3-h period, only a low, statistically-insignificant studies to date (Wu et al. 2009). The authors also
increase in serum zinc levels was observed in healthy reported a list of biological effects in hairless mice
male subjects on whom 100 g 40% ZnO ointments after 60 day exposure to topically applied TiO2
(size not given) were applied (Derry et al. 1983). Near including decreased body weight, skin damage and
nanosized (~117 nm) ZnO particles applied to ex vivo organ damage (Wu et al. 2009). This study highlights
skin samples from healthy adults remained on top of the importance of realistic exposure periods in asses-
the stratum corneum (Dussert et al. 1997). Consis- sing the long-term use of nanoparticles, such as ZnO
tent with this, penetration into tissue was not detected in sunscreen. For example, it is possible that a build-
in experiments with diffusion cells or by electron up of a secondary diffusion barrier due to the for-
microscopy following a single 24-h exposure of mation of stable bonds between electrophilic metals
ex vivo human skin samples to sunscreen containing and proteins of the skin may retard dermal penetra-
26–30 nm ZnO (Cross et al. 2007). Similarly, within a tion by as much as days (Hostynek 2003). Currently
24-h time-frame, ZnO nanoparticles did not pene- there is no information on whether similar interac-
trate past the first few layers of full-thickness ex vivo tions occur between ZnO nanoparticles and skin
porcine skin samples (Gamer et al. 2006). Under in surface proteins, and whether such interactions
vivo conditions using multiphoton microscopy, may influence the results of penetration studies
Nanotoxicology Downloaded from informahealthcare.com by University of Toronto on 02/19/13

Zvyagin et al. (2008) and Roberts et al. (2008) that only address short-term exposures, such as those
showed that ZnO nanoparticles accumulated into that are available to date.
skin folds and hair follicles when applied to the Some early studies have shown that the state of the
forearms of human volunteers, but did not skin, as well as the delivery vehicle, can influence the
penetrate the stratum corneum (Roberts et al. percutaneous absorption of ZnO microparticles
2008; Zvyagin et al. 2008). (Agren 1991) and suggest that these factors may
In contrast, quantum dots with a range of physio- influence the absorption of ZnO nanoparticles, too.
chemical characteristics penetrated through ex vivo In impaired skin models, serum zinc increased after
porcine skin mounted on flow-through diffusion cells application of ZnO (size not given) to wounded rat
following 8- or 24-h exposure (Ryman-Rasmussen (Hallmans 1978) and human (Hallmans 1977a) skin,
For personal use only.

et al. 2006), and maghemite and iron nanoparticles while the human study also showed elevated levels of
were detected in the stratum granulosum and viable zinc in urine. These studies did not differentiate
epidermis of full-thickness human skin samples also between zinc ions released from ZnO and ZnO par-
mounted on diffusion cells (Baroli et al. 2007), ticles, per se. Nevertheless, they serve to illustrate that
showing that nanoparticle penetration can occur skin can be penetrated by zinc, and that the potential
under some circumstances. ZnO nanoparticles were for such penetration can be influenced by the state of
also shown to pass through the stratum corneum the skin to which the ZnO is applied. More recently,
when they were applied hairless mouse skin mounted UV exposure has been implicated in enhanced pen-
on diffusion cells (Kuo et al. 2009), but the use of this etration of quantum dot nanoparticles through mildly
model which is known to be far more penetrable than sunburned murine skin into the dermis in vivo
human skin (Scott et al. 1986; van Ravenzwaay and (Mortensen et al. 2008), albeit still at a very low level.
Leibold 2004; Scientific Committee on Consumer A recent study has been conducted in which sun-
Products [SCCP] 2006), particularly in the presence screen containing traceable ZnO particles was applied
of chemical enhancers (oleic acid and/or ethanol), twice daily for five consecutive days to humans
may make the relevance of these findings to dermal undergoing normal activities at a Sydney beach
penetration of ZnO nanoparticles during normal sun- (Gulson et al. 2009). The ZnO was enriched to
screen use somewhat questionable. > 99.9% with a stable zinc isotope, 68Zn, with the
Although Roberts et al (2008) used a relatively intention that measurements of altered isotopic
large number of subjects (60) of different age, race ratios of zinc in blood and urine samples by multi-
and gender, the conclusions from all the studies collector ICP-MS (inductively-coupled plasma mass-
described here are limited by the use of a single or spectrometry) would distinguish any zinc dermally
few ZnO applications, small application areas and absorbed from sunscreen from that naturally present
short exposure periods. From this perspective, it is in the body. Two sunscreens, one with 68ZnO nano-
interesting that TiO2 nanoparticles (4 nm) were particles (20 nm) and the other with 68ZnO particles
shown to penetrate through ear skin of pigs when (> 100 nm), were used to determine if dermal absorp-
applied for 30 consecutive days compared with no tion of ZnO was dependent on particle size. The data
penetration after only 24 h using ex vivo skin samples from this study, when available, may provide informa-
mounted on diffusion cells, a common technique tion on potential dermal absorption of uncoated ZnO
32 M.J. Osmond & M.J. McCall

particles in a specific sunscreen formulation, under Even if ZnO nanoparticles in sunscreens do not
conditions of normal use by humans. penetrate into living tissues, there remains the possi-
bility that dissolved Zn2+ may. Zinc is the second-
most abundant, essential trace-metal after iron
Potential health effects of dermal exposure (St. Croix et al. 2005) and is ubiquitous in the
body, being present in all organs, tissues and fluids
An initial concern over the use of metal oxide particles (Rostan et al. 2002). Zinc-binding proteins form the
in sunscreens was that ZnO and TiO2 are semi- largest class of proteins in multicellular animals
conductors. UV absorption by semi-conductors can (Nyborg and Peersen 2004), and constitute a large
excite electrons from the valence band to the con- proportion of the proteins hypothesized to be involved
duction band, leaving a ‘hole’ in the former. Most in the regulation of gene expression (Tupler et al.
electrons return to the valence band, releasing excess 2001). Consequently, physiological levels of zinc are
energy as heat, but ~ 10% do not return and can critical for normal gene expression (Nyborg and
generate free radicals on the surface of the metal oxide Peersen 2004) and intracellular free zinc is generally
in the presence of water (Wolf et al. 2001). The found only in picomolar concentrations (Peck and
inherent photocatalytic activity of ZnO increases Ray 1971; Simons 1991), with its cellular distribution
with decreasing particle size (Park and Kang 1997; tightly regulated (Maret 1995; St. Croix et al. 2005).
Nanotoxicology Downloaded from informahealthcare.com by University of Toronto on 02/19/13

Casey et al. 2006), and also can be influenced by Within the physiological range, zinc is an antioxidant
particle morphology and the method of preparation (Rostan et al. 2002; Prasad et al. 2004) but, should
(Wang et al. 2007). Coating or doping are techniques intracellular levels shift too far in either direction
widely used to reduce the semiconductor activity of outside this range, zinc can become harmful to the
metal oxide particles for sunscreens. Consistent with cell (Krones et al. 2005; St. Croix et al. 2005;
this, microsized ZnO particles, both coated and Wiseman et al. 2006). Disruptions to cellular zinc
uncoated, were reported to be both photo-stable homeostasis have been linked in in vitro systems to
and non-photocatalytic (Mitchnick et al. 1999), and oxidative stress, mitochondrial dysfunction, and loss
an absence of photo-genotoxicity in studies with of cellular viability (Maret 1995; Sensi et al. 1999;
Chinese hamster ovary cells has also been reported Dineley et al. 2003), and excess zinc can also act as a
For personal use only.

(Dufour et al. 2006). neurotoxin (Frederickson et al. 2005). Zinc-ion


Zinc oxide particles extracted from commercially- release from ZnO nanoparticles is more rapid than
available sunscreens have been shown to be photo- the release from ZnO microparticles (Yang and Xie
catalytically active in vitro, but were not investigated 2006). The potential for disruption of intracellular
for biological impact in vivo (Rampaul et al. 2007). levels of zinc from daily applications of sunscreens
Similarly, TiO2 and ZnO nanoparticles present in a containing ZnO nanoparticles over long terms should
number of commercially-available sunscreens were be evaluated, given we know that zinc released from
recently identified as initiators of the accelerated ZnO can undergo percutaneous absorption.
weathering of surface coatings on roofs manufactured
by an Australian company (Barker and Branch 2008).
The incidental contact with the roofs during instal- Potential for translocation
lation by workers who used these sunscreens trans-
ferred nanoparticles in sufficient amounts to Early studies showed that 65Zn, from 65ZnO particles
photocatalytically initiate the free-radical driven deg- in tape applied to the skin of rats, showed a rapid
radation of the roof coating. In this case, biological accumulation in pancreas, kidney, liver, serum, hair
impact was not assessed. In one study, however, ZnO and bone, and was excreted in urine and faeces
particles isolated from commercially-available sun- (Hallmans 1977b; Hallmans et al. 1978). However,
screen were shown to induce DNA damage in vitro intact ZnO nanoparticles have not yet been shown to
after UV irradiation, producing hydroxyl radicals penetrate through healthy human skin and the poten-
that attacked the DNA sugar-phosphate chain tial translocation of ZnO nanoparticles arising from
(Hidaka et al. 2006). In contrast, microsized ZnO dermal absorption has not been investigated.
particles in sunscreen were found to protect UVB-
irradiated murine keratinocyte cells from cytotoxic
stress, DNA damage, disintegration of the cell mem- Is more research required?
brane and oxidative stress, provided the sunscreen
was applied thickly enough to screen at least 90% of At this point it is difficult to conclude whether dermal
UVB radiation; otherwise an increase of intracellular exposure to ZnO nanoparticles in sunscreen poses
ROS was detected (Hayashi et al. 2001). a hazard from internal exposure to free ZnO
Analysis of ZnO for sunscreens 33

nanoparticles. It is probable that the nanoparticles do differential response to nanoparticles should also be
not penetrate through the stratum corneum to living considered. Pregnant BALB/c mice that received
tissue in healthy, intact skin after a limited number of controlled doses of intra-nasally administered suspen-
exposures (Table Ic). However, it is not clear whether sions of TiO2 nanoparticles or diesel-exhaust particles
this situation might alter over a lifetime’s use of during pregnancy showed an elevated inflammatory
sunscreen, when the influences of repeated exposure, response to both compared to non-pregnant controls.
aging, skin health, waxing, shaving, and penetration Additionally, the observed responses correlated with
enhancers such as UV radiation or skin hydration are microarray analyses revealing altered patterns of gene
taken into consideration. Specifically, research should expression for a number of pathways involved in the
be directed at two areas: (i) To determine if zinc inflammatory response and immune regulation, cell
detected in vivo from the application of ZnO particles proliferation and metabolism. Furthermore, the
to skin is in the form of dissolved Zn2+ cations or solid in utero exposure potentiated offspring to diminished
nanoparticles; and (ii) to determine the effects on lung function (Fedulov et al. 2008). These data
health from long-term exposure to sunscreens con- suggest that pregnancy may alter the response to
taining nanoparticles of ZnO, and particularly the nanoparticles even if they are considered inert in
effects of free-radical generation on the surface of the non-pregnant state, and that this may impact
the skin or possibly in hair follicles. foetal development.
Nanotoxicology Downloaded from informahealthcare.com by University of Toronto on 02/19/13

Children may be more susceptible to adverse effects


from exposure to potentially hazardous materials due
Transplacental and childhood exposure to a variety of factors, including potentially immature
detoxification systems, as well as differences in behav-
There are critical periods in utero and during child- ior and metabolism (Selevan et al. 2000). Children
hood when exposure to certain compounds can have a greater ratio of surface area to volume than
affect reproductive development and/or capacity adults – approximately 2.7–1.3 times higher in new-
(Makri et al. 2004; Pryor et al. 2000). While the borns and children, respectively (Selevan et al. 2000).
degree of exposure is critical, the timing of exposure Consequently, although lower mass amounts of
may also have profound impacts. sunscreen may be applied, their effective exposures
For personal use only.

Low molecular weight, lipid-soluble, non-ionized may be greater, particularly if multiple applications
substances are most able to diffuse through the pla- are administered over the course of each day.
centa or into mammary glands from maternal circu- Currently there are no studies on the long-term
lation (Makri et al. 2004). Solid, orally-ingested effects of in utero or childhood exposure to ZnO
microparticles have been shown to rapidly translocate nanoparticles in sunscreens, and therefore this is an
into the milk of lactating women as well as transpla- area of research that should be addressed.
centally (Volkheimer 1974) but studies conducted
with nanoparticles are few in number. In one exper-
iment, foetal lethality and developmental abnormal-
Ecological impact of product use and disposal
ities in mice were associated with maternal
interperitoneal injection of carbon(60) fullerenes, Hazard identification and potential for exposure
suggesting transplacental transfer of the nanoparticles
via maternal blood flow (Tsuchiya et al. 1996). This In Australia, the intentional disposal of ZnO nano-
contrasts with another study where, although mouse particles is covered by existing regulations for disposal
embryonic cells were shown to internalize polysty- of inorganic zinc salts, which are known to have
rene-based nanoparticles in vitro, there were no asso- biocidal activity (Bruschi and Thomas 2006). How-
ciated developmental abnormalities or toxicities ever, the normal use of sunscreens and other per-
in vivo (Bosman et al. 2005). Therefore, it is possible sonal-care products leads to uncontrolled release of
that developmental effects associated with certain their ingredients to the natural environment (for
nanoparticles may not be representative of all nano- example, by swimming or wet-weather run-off) and
particles (Hagens et al. 2007) and initially they should to water-treatment and sewage systems (for example
be assessed on a case-by-case basis. from showering) (Daughton and Ternes 1999). The
If nanoparticles can translocate through the pla- release to sea-water of four commonly-used chemical
centa or into mammary glands, the possible disrup- UV absorbers in sunscreens has been observed to
tions to the developing foetus or neonate and their indirectly cause rapid and complete bleaching of
impact on adult-onset pathologies bear serious con- hard corals, even at extremely low concentrations,
sideration. The possibility that pregnant females com- by affecting symbiotic algae that nourish reef-building
pared to non-pregnant females may also exhibit a coral species; the chemicals induce a lytic viral cycle in
34 M.J. Osmond & M.J. McCall

symbiotic zooxanthellae with latent infections, leading When assessing the potential ecotoxicity of ZnO
to infection of neighboring coral communities nanoparticles, the dynamics of the particles in solu-
(Danovaro et al. 2008). Similarly, the bioaccumula- tion should be fully characterized, as toxicity may be
tion of up to 2 mg/kg lipid comprised of six different ascribed erroneously to the nanoparticle itself when in
chemical sunscreen components was identified in fish fact it may be due to Zn2+ released from the nano-
found in several German lakes used for recreational particle. ZnO is insoluble in water (Perl 1993) but
swimming during summer periods, even though appears to be at least partially soluble in some bio-
the concentration in the water was much lower logical fluids. For example, Franklin et al. (2007)
(Daughton and Ternes 1999). This suggests that showed that ZnO nanoparticles rapidly aggregated
although sunscreen ingredients may enter ecosystems and then dissolved in a neutral freshwater algal
only at very low levels and may not necessarily elicit an medium. Nano-sized ZnO was shown to be no
acute response, a continual influx may exert more more toxic than bulk ZnO and dissolved ZnCl2 to
subtle, long-term effects (Daughton and Ternes 1999). the freshwater algae Pseudokirchneriella subcapitata,
It is likely that there will be environmental exposure and the toxicity exhibited by all three could be attrib-
to ZnO nanoparticles from human use of sunscreens uted to released Zn2+ (Franklin et al. 2007). This
but the fate and toxicity of nanoparticles in the envi- result was confirmed in another laboratory using the
ronment is expected to be influenced by the degree of same organism (Aruoja et al. 2009). The importance
Nanotoxicology Downloaded from informahealthcare.com by University of Toronto on 02/19/13

aggregation, stability, transport, sedimentation, of characterizing particle solubility was also demon-
adsorption of environmental molecules and bio- strated by Brunner et al. (2006), who showed that the
uptake, as well as potential dissolution. The specific partially-soluble nanoparticles ZnO and Fe2O3 were
behavior of ZnO nanoparticles washed from more cytotoxic to rodent and human cell cultures
sunscreens in the relevant environments has not yet than the insoluble particles Ca3(PO4)2 and TiO2
been addressed. (Brunner et al. 2006). Consistent with this, suspen-
sions of bulk and nanoparticulate ZnO, as well as
dissolved ZnSO4, were toxic to bacteria and crusta-
Potential effects of environmental exposure ceans, and this toxicity was shown to be largely due to
solubilized zinc rather than the ZnO particles
For personal use only.

A small number of studies have been published within themselves (Heinlaan et al. 2008).
the last few years addressing the impact of ZnO In contrast, a recent study demonstrated that a
nanoparticles (not in sunscreen) on the environment solution containing ~ 180 nm aggregates of ZnO
(Table Id). ZnO has been shown to possess nanoparticles, filtered through 100 nm acrodisc
antimicrobial activity (Sawai 2003; Reddy et al. syringe filters to exclude the nanoparticles, exhibited
2007) that increases with decreasing particle size toxicity to zebrafish, again indicative of released Zn2+-
and concomitant increase in specific surface area mediated toxicity. However, the filtered solution con-
(Yamamoto 2001; Jones et al. 2008), and which taining only the dissolved zinc ions was less toxic than
appears to arise from the generation of ROS from the original nanoparticle suspension suggesting that
the particle surfaces (Yamamoto 2001; Yamamoto intact nanoparticles may still enhance overall toxicity
et al. 2004). TEM micrographs of bacteria incubated (Zhu et al. 2008). A follow-up study also suggested
with nano-sized ZnO have also shown nanoparticles that the toxicity of ZnO nanoparticles to developing
located inside and outside of the cell, with associated zebrafish could be attributed to a combination of
membrane damage (Brayner et al. 2006; Huang et al. intact ZnO nanoparticles and released Zn2+. Inter-
2008). However the antimicrobial effect of ZnO estingly, this study also reported that when the assay
diminished markedly when bacteria were exposed was conducted in the presence of sediment, the tox-
under dark conditions, suggesting that the anti- icity of the nanoparticles was largely mitigated
bacterial effect might be enhanced by visible light (Zhu et al. 2009). Similarly, although dissolved
(Jones et al. 2008). This is consistent with photoca- Zn2+ ions were shown to contribute to some toxicity
talytically-activated ROS toxicity. In contrast, against certain bacterial strains, toxicity was largely
Adams et al. reported that the toxicity of ZnO to attributed to exposure to ZnO particles, with nano-
bacteria was largely size- and light-independent particles exerting a more toxic effect than bulk par-
(Adams et al. 2006a, 2006b). Moreover, incubation ticles (Jiang et al. 2009). The impact of chemical
with 3–10 mM nanosized ZnO caused 100% growth stability of metallic nanoparticles on cellular toxicity
inhibition of Escherichia coli, while lower concentra- has recently been reviewed (Auffan et al. 2009).
tions between 1 and 1.5 mM supported only a slight Zinc oxide nanoparticles may exhibit some toxicity
increase in colony growth (Brayner et al. 2006), to plants, based on limited studies. Exposure to
suggesting a dose-dependent effect. suspensions of ZnO nanoparticles inhibited seed
Analysis of ZnO for sunscreens 35

germination of corn and virtually terminated the root effects arising from the long-term use of some other
growth of radish, rape, ryegrass, lettuce, corn and active sunscreen ingredients (Schlumpf et al. 2001).
cucumber (Lin and Xing 2007) with a lower IC50 Nonetheless, there is clearly a dearth of research into
than previously reported for some other plant the specific risks to human health and the environ-
species using non-nano ZnO (Paschke et al. 2006; ment associated with ZnO nanoparticles destined for
Paschke et al. 2000). Importantly, the phytotoxicity of use in sunscreens. Recently, the Environmental
the filtered ZnO nanoparticle solution containing Working Group released a statement in favor of using
Zn2+ was shown to be insignificant, suggesting that sunscreens containing ZnO and TiO2 nanoparticles
the observed phytotoxicity was due to the exposure to on the basis of current evidence, although it was noted
intact nanoparticles rather than released Zn2+ (Lin that the impact of compromised skin had not been
and Xing 2007). completely addressed by existing studies and that
there were “significant unaddressed safety concerns
for workers handling nano materials” (EWG 2009).
Is more research required? The report also noted that insufficient environmental
assessments are available for regulators to adequately
The paucity of information in Table Id suggests a lack
control potential hazard associated with mainstream
of public research targeted specifically to determine
use of these materials.
Nanotoxicology Downloaded from informahealthcare.com by University of Toronto on 02/19/13

the impact of ZnO nanoparticles from sunscreens in


In summary, future research efforts should be
the environment, although powdered ZnO has
expended to obtain information on and an under-
received some attention. Studies so far have tended
standing of: (i) Inhalation exposure for workers and
to focus on isolated organisms, rather than holistic
other personnel during the manufacturing phase; (ii)
studies to address the effects on symbiotic organisms
the impact of less than healthy skin and long-term use
or potential bioaccumulation progressing up the food
on the skin-penetrability of ZnO nanoparticles in
chain. An analysis of the impact of ZnO nanoparticles
sunscreen as well as their potential translocation;
on an ecosystem might be appropriate, where possi-
(iii) the potential for free radical activity from ZnO
ble, in order to detect potentially subtle, synergistic
nanoparticles lodged in hair follicles; (iv) the transport
effects that might not otherwise be revealed under
and fate of ZnO nanoparticles after discharge into the
For personal use only.

isolated experimental conditions. In addition, studies


environment arising from manufacturing and con-
are needed on the state of ZnO particles released to
sumer use; and (v) their ultimate effects on ecosys-
the environment from swimmers using sunscreen, as
tems. In order to facilitate comparison between
these particles are likely to be coated with ingredients
different studies, research should be conducted
from the sunscreen formulation, and may behave
together with a thorough characterization of the phys-
differently from pristine ZnO particles in both model
ical and chemical properties of the ZnO nanoparticles
and natural environments. Indeed, studies on the
studied, as several metrics in addition to size may
movement and fate of ZnO nanoparticles released
influence their behavior. Furthermore, the potential
to the environment should precede any comprehen-
risk for any detrimental effects on foetal development
sive toxicity studies so that the aggregation state, and
arising from maternal use of sunscreens containing
altered surface coatings, may be identified; and then
ZnO nanoparticles should be ascertained.
relevant forms of the nanoparticles may be tested in
ecotoxicity studies to obtain useful data.
Acknowledgements
Conclusions
The authors gratefully acknowledge Brendan Carney
ZnO nanoparticles show the broadest UV protection (Department of Infrastructure, Transport, Regional
of all the active ingredients currently available in Development & Local Government Australia) for
commercial sunscreens (Pinnell et al. 2000), and it providing advice regarding the possible OH&S issues
is important to bear this in mind when considering the surrounding the transport of ZnO nanoparticles in
potential risks associated with their use. Their incor- Australia. We would also like to thank Glenn McLeod
poration in sunscreens provides broad protection for preparing the diagrams used in this manuscript,
against the known carcinogenic, immunosuppressant and Phil Casey (CSIRO) for his critical reading,
and photo-ageing effects of UVA and UVB. There- suggestions and comments.
fore, avoidance of sunscreens containing ZnO nano-
particles on the basis of unknown or perceived hazards Declaration of interest: Work on this manuscript
would appear to be counterproductive, particularly was supported by the Nanosafety Theme in the CSIRO
when considered against the potential for adverse Future Manufacturing Flagship, which receives its
36 M.J. Osmond & M.J. McCall

funding from the Australian Government. The authors Escherichia coli bacteria in ultrafine ZnO nanoparticles colloidal
medium. Nano Lett 6:866–870.
report no conflicts of interest. The authors alone are
Breternitz M, Flach M, Prassler J, Elsner P, Fluhr JW. 2007. Acute
responsible for the content and writing of the paper. barrier disruption by adhesive tapes is influenced by pressure,
time and anatomical location integrity and cohesion assessed by
sequential tape stripping; a randomized, controlled study. Br J
References
Dermatol 156:231–240.
Adams L, Lyon D, McIntosh A, Alvarez P. 2006a. Comparative Brouwer DH, Gijsbers JH, Lurvink MW. 2004. Personal exposure
toxicity of nano-scale TiO2, SiO2 and ZnO water suspensions. to ultrafine particles in the workplace: Exploring sampling
Wat Sci Tech 54:327–334. techniques and strategies. Ann Occup Hyg 48:439–453.
Adams LK, Lyon DY, Alvarez PJJ. 2006b. Comparative eco- Brown JS, Zeman KL, Bennett WD. 2002. Ultrafine particle
toxicity of nanoscale TiO2, SiO2, and ZnO water suspensions. deposition and clearance in the healthy and obstructed lung.
Water Res 40:3527–3532. Am J Respir Crit Care Med 166:1240–1247.
Agren MS. 1991. Influence of two vehicles for zinc oxide on zinc Brown MB, Martin GP, Jones SA, Akomeah FK. 2006. Dermal
absorption through intact skin and wounds. Acta Derm Venereol and transdermal drug delivery systems: Current and future
71:153–156. prospects. Drug Deliv 13:175–187.
Aruoja V, Dubourguier HC, Kasemets K, Kahru A. 2009. Toxicity Bruls WA, Slaper JC, van der Leun JC, Berrins L. 1984. Trans-
of nanoparticles of CuO, ZnO and TiO2 to microalgae mission of human epidermis and stratum corneum as a function
Pseudokirchneriella subcapitata. Sci Total Environ 407: of thickness in the ultraviolet and visible wavelengths.
1461–1468. Photochem Photobiol 40:485–494.
Nanotoxicology Downloaded from informahealthcare.com by University of Toronto on 02/19/13

Auffan M, Rose J, Wiesner MR, Bottero JY. 2009. Chemical Brunner TJ, Wick P, Manser P, Spohn P, Grass RN,
stability of metallic nanoparticles: A parameter controlling Limbach LK, Bruinink A, Stark WJ. 2006. In vitro cytotoxicity
their potential cellular toxicity in vitro. Environ Pollut of oxide nanoparticles: Comparison to asbestos, silica, and the
157:1127–1133. effect of particle solubility. Environ Sci Technol 40:4374–4381.
Baggs RB, Ferin J, Oberdörster G. 1997. Regression of pulmonary Bruschi S, Thomas S. 2006. A review of the potential occupa-
lesions produced by inhaled titanium dioxide in rats. Vet Pathol tional health & safety implications of nanotechnology. In:
34:592–597. Council ASaC, editor: accessed from http://www.safe-
Barker P, Branch A. 2008. The interaction of modern sunscreen workaustralia.gov.au/NR/rdonlyres/8FBDE049-ADB4-417D-
formulations with surface coatings. Prog Org Coat 62: 8771-B9DA1D6BC7E6/0/ASCCReviewOHSImplications-
313–320. Nanotechnology2006.pdf..
Baroli B, Ennas MG, Loffredo F, Isola M, Pinna R, Lopez- Casey PS, Rossouw CJ, Boskovic S, Lawrence KA, Turney TW.
Quintela MA. 2007. Penetration of metallic nanoparticles in 2006. Incorporation of dopants into the lattice of ZnO
For personal use only.

human full-thickness skin. Soc Invest Dermatol 127:1701–1712. nanoparticles to control photoactivity. Superlattic Microst
Bauermann LP, Bill J, Aldinger F. 2006. Bio-friendly synthesis of 39:97–106.
ZnO nanoparticles in aqueous solution at near-neutral pH and Cayrol C, Sarraute J, Tarroux R, Redoules D, Charveron M,
low temperature. J Phys Chem B 110:5182–5185. Gall Y. 1999. A mineral sunscreen affords genomic protection
Beckett W, Chalupa D, Pauly-Brown A, Speers D, Stewart J, against ultraviolet (UV)B and UVA radiation: In vitro and in situ
Frampton M, Utell M, Huang L, Cox C, Zareba W, et al. assays. Br J Dermatol 141:250–258.
2005. Comparing inhaled ultrafine versus fine zinc oxide parti- Cerimele D, Celleno L, Serri F. 1990. Physiological changes in
cles in healthy adults. Am J Respir Crit Care Med 171: ageing skin. Br J Dermatol 122:13–20.
1129–1135. Champion JA, Mitragotri S. 2006. Role of target geometry in
Benson HA. 2005. Transdermal drug delivery: Penetration phagocytosis. PNAS 103:4930–4934.
enhancement techniques. Curr Drug Deliv 2:2–33. Choi EH, Man MQ, Xu P, Xin S, Liu Z, Crumrine DA, Jiang YJ,
Bermudez E, Mangum JB, Asgharian B, Wong BA, Reverdy EE, Fluhr JW, Feingold KR, Elias PM, et al. 2007. Stratum corneum
Janszen DB, Hext PM, Warheit DB, Everitt JI. 2002. Long-term acidification is impaired in moderately aged human and murine
pulmonary responses of three laboratory rodent species to sub- skin. J Invest Dermatol 127:2847–2856.
chronic inhalation of pigmentary titanium dioxide particles. Churg A, Stevens B, Wright JL. 1998. Comparison of the uptake of
Toxicol Sci 70:86–97. fine and ultrafine TiO2 in a tracheal explant system. Am J Physiol
Bermudez E, Mangum JB, Wong BA, Asgharian B, Hext PM, 274:L81–86.
Warheit DB, Everitt JI. 2004. Pulmonary responses of mice, rats, Cross SE, Innes B, Roberts MS, Tsuzuki T, Robertson TA,
and hamsters to subchronic inhalation of ultrafine titanium McCormick P. 2007. Human skin penetration of sunscreen
dioxide particles. Toxicol Sci 77:347–357. nanoparticles: In-vitro assessment of a novel micronized zinc
Borm PJ, Kreyling W. 2004. Toxicological hazards of inhaled oxide formulation. Skin Pharmacol Physiol 20:148–154.
nanoparticles – potential implications for drug delivery. J Danovaro R, Bongiorni L, Corinaldesi C, Giovannelli D,
Nanosci Nanotechnol 4:521–531. Damiani E, Astolfi P, Greci L, Pusceddu A. 2008. Sunscreens
Borm PJA, Schins RPF, Albrecht C. 2004. Inhaled particles and cause coral bleaching by promoting viral infections. Environ
lung cancer, Part B: Paradigms and risk assessment. Int J Cancer Health Perspect 116:441–447.
110:3–14. Darr D, Fridovich I. 1994. Free radicals in cutaneous biology.
Bosman SJ, Nieto SP, Patton WC, Jacobson JD, Corselli JU, J Invest Dermatol 102:671–675.
Chan PJ. 2005. Development of mammalian embryos exposed Daughton CG, Ternes TA. 1999. Pharmaceuticals and personal
to mixed-size nanoparticles. Clin Exp Obstet Gynecol care products in the environment: Agents of subtle change?
32:222–224. Environ Health Perspect 107:907–938.
Bouwstra JA, Ponec M. 2006. The skin barrier in healthy and de Jong WH, Hagens WI, Krystek P, Burger MC, Sips AJ,
diseased state. Biochim Biophys Acta 1758:2080–2095. Geertsma RE. 2008. Particle size-dependent organ distribution
Brayner R, Ferrari-lliou R, Brivois N, Djediat S, Benedetti MF, of gold nanoparticles after intravenous administration.
Fievet F. 2006. Toxicological impact studies based on Biomaterials 29:1912–1919.
Analysis of ZnO for sunscreens 37
Derry JE, McLean WM, Freeman JB. 1983. A study of the Frederick JE, Snell HE. 1988. Ultraviolet radiation levels during the
percutaneous absorption from topically applied zinc oxide Antarctic spring. Science 241:438–440.
ointment. J Parenter Enteral Nutr 7:131–135. Frederickson CP, Koh JY, Bush AI. 2005. The neurobiology of zinc
Diffey BL. 2002. Human exposure to solar ultraviolet radiation. J in health and disease. Nat Rev Neurosci 6:449–462.
Cosmet Dermatol 1:124–130. Fubini B. 1997. Surface reactivity in the pathogenic response to
Dineley KE, Votyakova TV, Reynolds IJ. 2003. Zinc inhibition of particulates. Environ Health Perspect 105:1013–1020.
cellular energy production: Implications for mitochondria and Gamer AO, Leibold E, van Ravenzwaay B. 2006. The in vitro
neurodegeneration. J Neurochem 85:563–570. absorption of microfine zinc oxide and titanium dioxide through
Donaldson K, Li XY, MacNee W. 1998. Ultrafine (nanometre) porcine skin. Toxicol in Vitro 20:301–307.
particle mediated lung injury. J. Aerosol Sci 29:553–560. Geiser M, Casaulta M, Kupferschmid B, Schulz H, Semmler-
Donaldson K, Stone V, Gilmour PS, Brown DM, MacNee W. Behnke M, Kreyling W. 2008. The role of macrophages in
2000. Ultrafine particles: Mechanisms of lung injury. Roy Soc the clearance of inhaled ultrafine titanium dioxie particles.
London Phil Tr A 358:2741–2749. Am J Respir Cell Mol Biol 38:371–376.
Duffy GM, Pillai SC, McCormack DE. 2007. A novel processing Gelis C, Mavon A, Delverdier M, Paillous N, Vicendo P. 2002.
route for the production of nanoparticulate zinc oxide using an Modifications of in vitro skin penetration under solar irradiation:
isophthalate precursor. Smart Mater Struct 16:1379–1381. Evaluation of flow-through diffusion cells. Photochem Photobiol
Dufour EK, Kumaravel T, Nohynek GJ, Kirkland D, Toutain H. 75:598–604.
2006. Clastogenicity, photo-clastogenicity or pseudo-photo- Ghadially R, Brown BE, Sequeira-Marin SM, Feingold KR,
clastogenicity: Genotoxic effects of zinc oxide in the dark, in Elias PM. 1995. The aged epidermal permeability barrier.
pre-irradiated or simultaneously irradiated Chinese hamster J Clin Invest 95:2281–2290.
Nanotoxicology Downloaded from informahealthcare.com by University of Toronto on 02/19/13

ovary cells. Mut Res 607:215–224. Gojova A, Guo B, Kota RS, Rutledge JC, Kennedy IM, Barakat AI.
Dussert A-S, Gooris E, Hemmerle J. 1997. Characterization of 2007. Induction of inflammation in vascular endothelial cells by
the mineral content of a physical sunscreen emulsion and its metal oxide nanoparticles: Effect of particle composition.
distribution onto human stratum corneum. Int J Cos Sci 19: Environ Health Perspect 115:403–409.
119–129. Goldstein AM, Abramovits W. 2003. Ceramides and the stratum
Elder A, Gelein R, Silva V, Feikert T, Opanashuk L, Carter J, corneum: Structure, function, and new methods to promote
Potter R, Maynard A, Ito Y, Finkelstein J, et al. 2006. Trans- repair. Int J Dermatol 42:256–259.
location of inhaled ultrafine manganese oxide particles to the Gopalan R, Osman I, Amani A, Matas Md, Anderson D. 2009. The
central nervous system. Environ Health Perspect 114: effect of zinc oxide and titanium dioxide nanoparticles in the
1172–1178. comet assay with UVA photoactivation of human sperm and
Elias PM. 1983. Epidermal lipids, barrier function, and desqua- lymphocytes. Nanotox 3:33–39.
mation. J Invest Dermatol 80:44S–49S. Gordon T, Chen LC, Fine JM, Schlesinger RB, Su WY,
For personal use only.

Elias PM. 2005. Stratum corneum defensive functions: An Kimmel TA, Amdur MO. 1992. Pulmonary effects of inhaled
integrated view. J Invest Dermatol 125:183–200. zinc oxide in human subjects, guinea pigs, rats, and rabbits. Am
EPA. 1995. Zinc oxide; Toxic chemical release reporting; Ind Hyg Assoc J 53:503–509.
Community right-to know. Federal Register 60:47334–47337. Gulson B, Gomez L, Casey P, Korsch M, Greenoak G, Trotter J,
Environmental Working Group (EWG). 2009. Nanotechnology & McCulloch M, McCall MJ. 2009. Are zinc oxide particles in
sunscreens. Accessed from the website: http://www.ewg.org/ sunscreens absorbed through the skin? 4th International Con-
cosmetics/report/sunscreen09/investigation/Nanotechnology- ference on Nanotechnology – Occupational and Environmental
Sunscreens. Health, Helsinki: page 85 of programme.
Fedulov AV, Leme A, Yang Z, Dahl M, Lim R, Marian TJ, Hachem J-P, Crumrine D, Fluhr J, Brown BE, Feingold KR,
Kobzik L. 2008. Pulmonary exposure to particles during preg- Elias PM. 2003. pH directly regulates epidermal permeability
nancy causes increased neonatal asthma susceptibility. Am J barrier homeostasis, and stratum corneum integrity/cohesion.
Respir Cell Mol Biol 38:57–67. J Invest Dermatol 121:345–353.
Fine JM, Gordon T, Chen LC, Kinney P, Falcone G, Beckett W. Hagens WI, Oomen AG, de Jong WH, Cassee FR, Sips AJ. 2007.
1997. Metal fume fever: Characterization of clinical and plasma What do we (need to) know about the kinetic properties
IL-6 responses in controlled human exposures to zinc oxide of nanoparticles in the body? Regul Toxicol Pharmacol
fume at and below the threshold limit value. J Occup Environ 49:217–229.
Med 39:722–726. Hallmans G. 1977a. Treatment of burns with zinc-tape. A study of
Fine JM, Gordon T, Chen LC, Kinney P, Falcone G, Sparer J, local absorption of zinc in humans. Scand J Plast Reconstr Surg
Beckett WS. 2000. Characterization of clinical tolerance to 11:155–161.
inhaled zinc oxide in naive subjects and sheet metal workers. Hallmans G. 1977b. Zinc resorption from zinc-tape during wound
J Occup Environ Med 42:1085–1091. healing. An experimental study in rats. Scand J Plast Reconstr
Fluhr JW, Behne MJ, Brown BE, Moskowitz DG, Selden C, Surg 11:27–32.
Mao-Qiang M, Mauro TM, Elias PM, Feingold KR. 2004. Hallmans G. 1978. Local absorption of zinc from wounds treated
Stratum corneum acidification in neonatal skin: Secretory with various zinc-compounds. Acta Derm Venereol 58:
phospholipase A2 and the sodium/hydrogen antiporter-1 acidify 251–256.
neonatal rat stratum corneum. J Invest Dermatol 122:320–329. Hallmans G, Nilsson AM, Sjostrom R. 1978. Absorption, retention
Forestier S. 2008. Rationale for sunscreen development. J Am Acad and excretion of 65Zn in rats with wounds treated with 65Zn-
Dermatol 58:S133–1389. labelled zinc tape. Scand J Plast Reconstr Surg 12:111–117.
Franklin NM, Rogers NJ, Apte SC, Batley GE, Gadd GE, Haratake A, Uchinda Y, Schmuth M, Tanno O, Yasuda R,
Casey PS. 2007. Comparative toxicity of nanoparticulate Epstein JH, Elias PM, Holleran WM. 1997. UVB-induced
ZnO, bulk ZnO, and ZnCl2 to a freshwater microalga alterations in permeability barrier function: Roles for epidermal
(Pseudokirchneriella subcapitata): The importance of particle hyperproliferation and thymocyte-mediated response. J Invest
solubility. Environ Sci Technol 41:8484–8490. Dermatol 108:769–775.
38 M.J. Osmond & M.J. McCall
Harvell JD, Maibach HI. 1994. Percutaneous absorption and extrapulmonary organs is size dependent but very low. J Toxicol
inflammation in aged skin: A review. J Am Acad Dermatol Environ Health A 65:1513–1530.
31:1015–1021. Krones CJ, Klosterhalfen B, Butz N, Hoelzl F, Junge K, Stumpf M,
Hayashi S, Takeshita H, Nagao N, Nikaido O, Miwa N. 2001. The Peiper C, Klinge U, Schumpelick V. 2005. Effect of zinc pre-
relationship between UVB screening and cytoprotection by treatment on pulmonary endothelial cells in vitro and pulmonary
microcorpuscular ZnO or ascorbate against DNA photodamage function in a porcine model of endotoxemia. J Surg Res
and membrane injuries in keratinocytes by oxidative stress. 123:251–256.
J Photochem Photobiol B 64:27–35. Kuo T-R, Wu C-L, Hsu C-T, Lo W, Chiang S-J, Lin S-J,
Heinlaan M, Ivask A, Blinova I, Dubourguier HC, Kahru A. 2008. Dong C-Y, Chen C-C. 2009. Chemical enhancer induced
Toxicity of nanosized and bulk ZnO, CuO and TiO2 to bacteria changes in the mechanisms of transdermal delivery of zinc oxide
Vibrio fischeri and crustaceans Daphnia magna and nanoparticles. Biomaterials 30:3002–3008.
Thamnocephalus platyurus. Chemosphere 71:1308–1316. Kuschner WG, D’Alessandro A, Wong H, Blanc PD. 1997. Early
Hermanns-Le T, Uhoda I, Smitz S, Pierard GE. 2004. Skin tensile pulmonary cytokine responses to zinc oxide fume inhalation.
properties revisited during ageing. Where now, what next? Environ Res 75:7–11.
J Cosmet Dermatol 3:35–40. Lademann J, Otberg N, Jacobi U, Hoffman RM, Blume-Peytavi U.
Hext PM, Tomenson JA, Thompson P. 2005. Titanium dioxide: 2005. Follicular penetration and targeting. J Invest Dermatol
Inhalation toxicology and epidemiology. Ann Occup Hyg Symp Proc 10:301–303.
49:461–472. Lademann J, Otberg N, Richter H, Weigmann HJ, Lindemann U,
Hidaka H, Kobayashi H, Koike T, Sato T, Serpone N. 2006. DNA Schaefer H, Sterry W. 2001. Investigation of follicular penetra-
damage photoinduced by cosmetic pigments and sunscreen tion of topically applied substances. Skin Pharmacol Appl Skin
Nanotoxicology Downloaded from informahealthcare.com by University of Toronto on 02/19/13

agents under solar exposure and artificial UV illumination. Physiol 14:17–22.


J Oleo Sci 55:249–261. Lademann J, Weigmann H, Rickmeyer C, Barthelmes H,
Hohr D, Steinfartz Y, Schins RPF, Knaapen AM, Martra G, Schaefer H, Mueller G, Sterry W. 1999. Penetration of titanium
Fubini B, Borm PJA. 2002. The surface area rather than the dioxide microparticles in a sunscreen formulation into the horny
surface coating determines the acute inflammatory response after layer and the follicular orifice. Skin Pharmacol Appl Skin Physiol
instillation of fine and ultrafine TiO2 in the rat. Int J Hyg Environ 12:247–256.
Health 205:239–244. Lam HF, Conner MW, Rogers AE, Fitzgerald S, Amdur MO.
Hostynek JJ. 2003. Factors determining percutaneous metal 1985. Functional and morphologic changes in the lungs of
absorption. Food Chem Toxicol 41:327–345. guinea pigs exposed to freshly generated ultrafine zinc oxide.
Huang Z, Zheng X, Yan D, Yin G, Liao X, Kang Y, Yao Y, Toxicol Appl Pharmacol 78:29–38.
Huang D, Hao B. 2008. Toxicological effect of ZnO Lam HF, Peisch R, Amdur MO. 1982. Changes in lung volumes
nanoparticles based on bacteria. Langmuir 24:4140–4144. and diffusing capacity in guinea pigs exposed to a combination of
For personal use only.

Hussain N, Jaitley V, Florence A. 2001. Recent advances in the sulfur dioxide and submicron zinc oxide mixed in a humidified
understanding of uptake of microparticulates across the gastro- furnace. Toxicol Appl Pharmacol 66:427–433.
intestinal lymphatics. Adv Drug Deliv Rev 50:107–142. Lewtas J. 2007. Air pollution combustion emissions: Characteri-
Jeng HA, Swanson J. 2006. Toxicity of metal oxide nano- zation of causative agents and mechanisms associated with
particles in mammalian cells. J Environ Sci Health A cancer, reproductive, and cardiovascular effects. Mut Res
41:2699–2711. 636:95–133.
Jiang SJ, Chu AW, Lu ZF, Pan MH, Che DF, Zhou X. 2007. Limbach LK, Li Y, Grass RN, Brunner TJ, Hintermann MA,
Ultraviolet B-induced alterations of the skin barrier and epider- Muller M, Gunther D, Stark WJ. 2005. Oxide nanoparticle
mal calcium gradient. Exp Dermatol 16:985–992. uptake in human lung fibroblasts: Effects of particle size,
Jiang W, Mashayekhi H, Xing B. 2009. Bacterial toxicity compar- agglomeration, and diffusion at low concentrations. Environ
ison between nano- and micro-scaled oxide particles. Environ Sci Technol 39:9370–9376.
Pollut 157:1619–1625. Lin D, Xing B. 2007. Phytotoxicity of nanoparticles: Inhibition
Jones N, Ray B, Ranjit KT, Manna AC. 2008. Antibacterial activity of seed germination and root growth. Environ Pollut 150:
of ZnO nanoparticle suspensions on a broad spectrum of micro- 243–250.
organisms. FEMS Microbiol Lett 279:71–76. Lockman PR, Koziara JM, Mumper RJ, Allen DD. 2004. Nano-
Kelleher P, Pacheco K, Newman LS. 2000. Inorganic dust pneu- particle surface charges alter blood-brain barrier integrity and
monias: The metal-related parenchymal disorders. Environ permeability. J Drug Target 12:635–641.
Health Perspect 108:685–696. Lundqvist M, Stigler J, Elia G, Lynch I, Cedervall T, Dawson KA.
Kendall M, Tetley TD, Wigzell E, Hutton B, Nieuwenhuijsen M, 2008. Nanoparticle size and surface properties determine the
Luckham P. 2002. Lung lining liquid modified PM2.5 in favor of protein corona with possible implications for biological impacts.
particle aggregation: A protective mechanism. Am J Physiol PNAS 105:14265–14270.
Lung Cell Mol Physiol 282:L109–114. Lynch I, Dawson KA. 2008. Protein-nanoparticle interactions.
Kleinwechter H, Janzen C, Knipping J, Wiggers H, Roth P. 2002. NanoToday 3:40–47.
Formation and properties of ZnO nano-particles from gas phase Madronich S, McKenzie RL, Bjorn LO, Caldwell MM. 1998.
synthesis processes. J Mater Sci 37:4349–4360. Changes in biologically active ultraviolet radiation reaching
Kreuter J. 2004. Influence of the surface properties on nanoparticle- the earth’s surface. J Photochem Photobiol B 46:5–19.
mediated transport of drugs to the brain. J Nanosci Nanotechnol Maier T, Korting HC. 2005. Sunscreens – which and what for?
4:484–488. Skin Pharmacol Physiol 18:253–262.
Kreyling W, Semmler-Behnke M, Moller W. 2006. Ultrafine Makri A, Goveia M, Balbus J, Parkin R. 2004. Children’s suscep-
particle-lung interactions: Does size matter? J Aerosol Med tibility to chemicals: A review by developmental stage. J Toxicol
19:74–83. Environ Health B Crit Rev 7:417–435.
Kreyling WG, Semmler M, Erbe F, Mayer P, Tanenaka S, Maret W. 1995. Metallothionein/disulfide interactions, oxidative
Schulz H, Oberdörster G, Ziesenis A. 2002. Translocation of stress, and the mobilization of cellular zinc. Neurochem Int
ultrafine insoluble iridium particles from lung epithelium to 27:111–117.
Analysis of ZnO for sunscreens 39
Marrot L, Meunier JR. 2008. Skin DNA photodamage and its Paschke MW, Perry LG, Redente EF. 2006. Zinc toxicity thre-
biological consequences. J Am Acad Dermatol 58:S139–148. sholds for reclamation forb species. Wat Air Soil Pollut
Matts PJ. 2006. Solar ultraviolet radiation: Definitions and termi- 140:317–330.
nology. Dermatol Clin 24:1–8. Paschke MW, Redente EF, Levy DB. 2000. Zinc toxicity thresholds
Maynard AD, Baron PA, Foley M, Shvedova AA, Kisin ER, for important reclamation grass species of the western United
Castranova V. 2004. Exposure to carbon nanotube material: States. Environ Toxicol Chem 19:2751–2751.
Aerosol release during the handling of unrefined single-walled Peck EJ, Ray WJJ. 1971. Metal complexes of phosphoglucomu-
carbon nanotube material. J Toxicol Environ Health A 67:87–107. tase in vivo. Alterations induced by insulin. J Biol Chem
Maynard AD, Kuempel ED. 2005. Airborne nanostructured 246:1160–1167.
particles and occupational health. J Nanopart Res 7:587-614. Perl AS. 1993. Zinc oxide. Am Ceramic Soc Bull 72:122–123.
Mitchnick MA, Fairhurst D, Pinnell SR. 1999. Microfine zinc Pinnell SR, Fairhurst D, Gillies R, Mitchnick MA, Kollias N. 2000.
oxide (z-cote) as a photostable UVA/UVB sunblock agent. Microfine zinc oxide is a superior sunscreen ingredient to
J Am Acad Dermatol 40:85–89. microfine titanium dioxide. Dermatol Surg 26:309–314.
Monteiller C, Tran L, MacNee W, Faux SP, Jones A, Miller B, Pirot F, Millet J, Kalia YN, Humbert P. 1996. In vitro study of
Donaldson K. 2007. The pro-inflammatory effects of low- percutaneous absorption, cutaneous bioavailability and bio-
toxicity low-solubility particles, nanoparticles and fine particles, equivalence of zinc and copper from five topical formulations.
on epithelial cells in vitro: The role of surface area. Occup Skin Pharmacol 9:259–269.
Environ Med 64:609–615. Popov AP, Priezzhev AV, Lademann J, Myllylä R. 2005.
Morrow PE. 1988. Possible mechanisms to explain dust overload- TiO 2 nanoparticles as an effective UV-B radiation skin-
ing of the lungs. Fund Appl Toxicol 10:369–384. protective compound in sunscreens. J Phys D: Appl Phys
Nanotoxicology Downloaded from informahealthcare.com by University of Toronto on 02/19/13

Mortensen LJ, Oberdörster G, Pentland AP, DeLouise LA. 2008. 38:2564–2570.


In vivo skin penetration of quantum dot nanoparticles in the Prasad AS, Bao B, Beck FWJ, Kucuk O, Sarkar FH. 2004.
murine model: The effect of UVR. NanoLetters 8:2779–2787. Antioxidant effect of zinc in humans. Free Radic Biol Med
Nanoderm. 2007. Quality of skin as a barrier to ultra-fine particles. 37:1182–1190.
Available from the website: www.uni-leipzig.de/~nanoderm/ Pryor JL, Hughes C, Foster W, Hales BF, Robaire B. 2000. Critical
Downloads/Nanoderm_Final_Summary.pdf. windows of exposure for children’s health: The reproductive
Nash JF, Tanner PR, Matts PJ. 2006. Ultraviolet A radiation: system in animals and humans. Environ Health Perspect
Testing and labeling for sunscreen products. Dermatol Clin 108:491–503.
24:63–74. Rampaul A, Parkin IP, Cramer LP. 2007. Damaging and protective
Nemmar A, Hoet PH, Vanquickenborne B, Dinsdale D, properties of inorganic components of sunscreens applied to
Thomeer M, Hoylaerts MF, Vanbilloen H, Mortelmans L, cultured human skin cells. J Photochem Photobiol A, Chem
Nemery B. 2002. Passage of inhaled particles into blood 191:138–148.
For personal use only.

circulation in humans. Circulation 105:411–414. Reddy KM, Feris K, Bell J, Wingett DG, Hanley C, Punnoose A.
Nemmar A, Hoylaerts MF, Hoet PHM, Nemery B. 2004. Possible 2007. Selective toxicity of zinc oxide nanoparticles to
mechanisms of the cardiovascular effects of inhaled particles: prokaryotic and eukaryotic systems. Appl Phys Lett 90:
Systemic translocation and prothrombotic effects. Toxicol Lett 213902-1–213902-3.
149:243–253. Renwick LC, Donaldson K, Clouter A. 2001. Impairment of
Nikula KJ, Avila KJ, Griffith WC, Mauderly JL. 1997. Lung tissue alveolar macrophage phagocytosis by ultrafine particles. Toxicol
responses and sites of particle retention differ between rats and Appl Pharmacol 172:119–127.
cynomolgus monkeys exposed chronically to diesel exhaust and Roberts M, Roberts M, Robertson T, Sanchez W, Thorling C,
coal dust. Fundam Appl Toxicol 37:37–53. Zou Y, Zhao X, Becker W, Zvyagin A. 2008. In vitro and in vivo
Nohynek GJ, Lademann J, Ribaud C, Roberts MS. 2007. Grey goo imaging of xenobiotic transport in human skin and in the rat
on the skin? Nanotechnology, cosmetic and sunscreen safety. liver. Journal of Biophotonics 1:478–493.
Crit Rev Toxicol 37:251–277. Rostan EF, DeBuys HV, Madey DL, Pinnell SR. 2002. Evidence
Nyborg JK, Peersen OB. 2004. That zincing feeling: The effects supporting zinc as an important antioxidant for skin. Int J
of EDTA on the behaviours of zinc-binding transcriptional Dermatol 41:606–611.
regulators. Biochem J 381:e3–4. Rothen-Rutischauser B, Schurch S, Haenni B, Kapp N, Gehr P.
Oberdörster G. 2000. Toxicology of ultrafine particles: In vivo 2006. Interaction of fine particles and nanoparticles with red
studies. Roy Soc London Phil Tr A 358:2719–2740. blood cells visualized with advanced microscopic techniques.
Oberdörster G. 2001. Pulmonary effects of inhaled ultrafine par- Environ Sci Technol 40:4353–4359.
ticles. Int Arch Occup Environ Health 74:1–8. Rouse JG, Yang J, Ryman-Rasmussen JP, Barron AR, Monteiro-
Oberdörster G, Ferin J, Lehnert BE. 1994. Correlation between Riviere NA. 2007. Effects of mechanical flexion on the pene-
particle size, in vivo particle persistence, and lung injury. Environ tration of fullerene amino acid-derivatized peptide nanoparticles
Health Perspect 102:173–179. through skin. Nano Lett 7:155–160.
Oberdörster G, Sharp Z, Atudorei V, Elder A, Gelein R, Ryman-Rasmussen JP, Riviere JE, Monteiro-Riviere NA. 2006.
Lunts A, Kreyling W, Cox C. 2002. Extrapulmonary trans- Penetration of intact skin by quantum dots with diverse
location of ultrafine carbon particles following whole-body physicochemical properties. Toxicol Sci 91:159–165.
inhalation exposure of rats. J Toxicol Environ Health A Sakurai H, Yasui H, Yamada Y, Nishimura H, Shigemoto M. 2005.
65:1531–1543. Detection of reactive oxygen species in the skin of live mice and
Otberg N, Richter H, Knuttel A, Schaefer H, Sterry W, Lademann J. rats exposed to UVA light: A research review on chemilumines-
2004. Laser spectroscopic methods for the characterization of cence and trials for UVA protection. Photochem Photobiol Sci
open and closed follicles. Laser Phys Lett 1:46–49. 4:715–720.
Park SB, Kang YC. 1997. Photocatalytic activity of nanometer size Sander CS, Chang H, Hamm F, Elsner P, Thiele JJ. 2004. Role of
ZnO particles prepared by spray pyrolysis. J Aerosol Sci 28: oxidative stress and the antioxidant network in cutaneous
S473–474. carcinogenesis. Int J Dermatol 43:326–335.
40 M.J. Osmond & M.J. McCall
Sato J, Denda M, Chang S, Elias PM, Feingold KR. 2002. van Ravenzwaay B, Leibold E. 2004. The significance of in vitro rat
Abrupt decreases in environmental humidity induce abnor- skin absorption studies to human risk assessment. Toxicol in
malities in permeability barrier homeostasis. J Invest Dermatol Vitro 18:219–225.
119:900–904. Viswanathan R, Gupta RB. 2003. Formation of zinc oxide nano-
Sawai J. 2003. Quantitative evaluation of antibacterial activities of particles in supercritical water. J Supercrit Fluids 27:187–193.
metallic oxide powders (ZnO, MgO and CaO) by conductimet- Vogt A, Combadiere B, Hadam S, Stieler KM, Lademann
ric assay. J Microbiol Meth 54:177–182. J, Schaefer H, Autran B, Sterry W, Blume-Peytavi U. 2006.
Sayes CM, Reed KL, Warheit DB. 2007. Assessing toxicity of fine 40 nm, but not 750 or 1,500 nm, nanoparticles enter epidermal
and nanoparticles: Comparing in vitro measurements to in vivo CD1a+ cells after transcutaneous application on human skin.
pulmonary toxicity profiles. Toxicol Sci 97:163–180. J Invest Dermatol 126:1316–1322.
Scientific Committee on Consumer Products (SCCP). 2006. Basic Vogt A, Mandt N, Lademann J, Schaefer H, Blume-Peytavi U.
criteria for the in vitro assessment of dermal absorption of 2005. Follicular targeting – a promising tool in selective derma-
cosmetic ingredients. Scientific Committee on Consumer totherapy. J Invest Dermatol Symp Proc 10:252–255.
Products SCCP/0970/60: Volkheimer G. 1974. Passage of particles through the wall of the
Schlumpf M, Cotton B, Conscience M, Haller V, Steinmann B, gastrointestinal tract. Environ Health Perspect 9:215–225.
Lichtensteiger W. 2001. In vitro and in vivo estrogenicity of UV Wagner H, Kostka K-H, Lehr C-M, Schaefer UF. 2003. pH
screens. Environ Health Perspect 109:239–244. profiles in human skin: Influence of two in vitro test
Scott RC, Walker M, Dugard PH. 1986. A comparison of the in systems for drug delivery testing. Eur J Pharm Biopharm
vitro permeability properties of human and some laboratory 55:57–65.
animal skins. Int J Cos Sci 8:189–194. Wahie S, Lloyd JJ, Farr PM. 2007. Sunscreen ingredients and
Nanotoxicology Downloaded from informahealthcare.com by University of Toronto on 02/19/13

Selevan SG, Kimmel CA, Mendola P. 2000. Identifying critical labelling: A survey of products available in the UK. Clin Exp
windows of exposure for children’s health. Environ Health Dermatol 32:359–364.
Perspect 108:451–455. Waller JM, Maibach HI. 2005. Age and skin structure and
Semmler M, Seitz J, Erbe F, Mayer P, Heyder J, Oberdörster G, function, a quantitative approach (I): Blood flow, pH, thick-
Kreyling WG. 2004. Long-term clearance kinetics of inhaled ness, and ultrasound echogenicity. Skin Res Technol
ultrafine insoluble iridium particles from the rat lung, including 11:221–235.
transient translocation into secondary organs. Inhal Toxicol Wang B, Feng W, Wang M, Wang T, Gu Y, Zhu M, Ouyang H,
16:453–459. Shi J, Zhang F, Zhao Y, et al. 2008. Acute toxicological impact of
Sensi SL, Yin HZ, Carriedo SG, Rao SS, Weiss JH. 1999. Pref- nano- and submicro-scaled zinc oxide powder on healthy adult
erential Zn2+ influx through Ca2+-permeable AMPA/kainate mice. J Nanopart Res 10:263–276.
channels triggers prolonged mitochondrial superoxide produc- Wang B, Feng WY, Wang TC, Jia G, Wang M, Shi JW, Zhang F,
tion. PNAS 96:2414–2419. Zhao YL, Chai ZF. 2006. Acute toxicity of nano- and micro-
For personal use only.

Simons TJB. 1991. Intracellular free zinc and zinc buffering in scale zinc powder in healthy adult mice. Toxicol Lett
human red blood cells. J Memb Biol 123:63–71. 161:115–123.
St. Croix CM, Leelavanichul K, Watkins SC, Kagan VE, Pitt BR. Wang H, Xie C, Zhang W, Cai S, Yang Z, Gui Y. 2007. Com-
2005. Nitric oxide and zinc homeostasis in acute lung injury. parison of dye degradation efficiency using ZnO powders with
Proc Am Thorac Soc 2:236–242. various size scales. J Haz Mat 141:645–652.
Suzuki T, Chow CW, Downey GP. 2008. Role of innate immune Wesselkamper SC, Chen LC, Gordon T. 2001a. Development of
cells and their products in lung immunopathology. Int J Biochem pulmonary tolerance in mice exposed to zinc oxide fumes.
Cell Biol 40:1348–1361. Toxicol Sci 60:144–151.
Tabata Y, Ikada Y. 1988. Effect of the size and surface charge of Wesselkamper SC, Chen LC, Kleeberger SR, Gordon T. 2001b.
polymer microspheres on their phagocytosis by macrophage. Genetic variability in the development of pulmonary tolerance to
Biomaterials 9:356–362. inhaled pollutants in inbred mice. Am J Physiol Lung Cell Mol
Takenaka S, Karg E, Roth C, Schulz H, Ziesenis A, Heinzmann U, Physiol 281:L1200–1209.
Schramel P, Heyder J. 2001. Pulmonary and systemic distribu- Wiebert P, Sanchez-Crespo A, Falk R, Philipson K, Lundin A,
tion of inhaled ultrafine silver particles in rats. Environ Health Larsson S, Moller W, Kreyling W, Svartengren M. 2006a. No
Perspect 109:547–551. significant translocation of inhaled 35-nm carbon particles to the
Tinkle SS, Antonini JM, Rich BA, Roberts JR, Salmen R, circulation in humans. Inhal Toxicol 18:741–747.
DePree K, Adkins EJ. 2003. Skin as a route of exposure and Wiebert P, Sanchez-Crespo A, Seitz J, Falk R, Philipson K,
sensitization in chronic beryllium disease. Environ Health Kreyling W, Moller W, Sommerer K, Larsson S,
Perspect 111:1202–1208. Svartengren M. 2006b. Negligible clearance of ultrafine particles
Tran CL, Buchanan D, Cullen RT, Searl A, Jones AD, retained in healthy and affected human lungs. Eur Respir J
Donaldson K. 2000. Inhalation of poorly soluble particles. II. 28:286–290.
Influence of particle surface area on inflammation and clearance. Wiseman DA, Wells SM, Wilham J, Hubbard M, Welker JE,
Inhal Toxicol 12:1113–1126. Black SM. 2006. Endothelial response to stress from exogenous
Tsai SJ, Ashter A, Ada E, Mead JL, Barry CF, Ellenbecker MJ. Zn2+ resembles that of NO-mediated nitrosative stress, and is
2008. Airborne nanoparticle release associated with the com- protected by MT-1 overexpression. Am J Physiol Cell Physiol
pounding of nanocomposites using nanoalumina as fillers. Aeros 291:555–568.
Air Qual Res 8:160–177. Wolf R, Wolf D, Morganti P, Ruocco V. 2001. Sunscreens. Clin
Tsuchiya T, Oguri I, Yamakoshi YN, Miyata N. 1996. Novel Dermatol 19:452–459.
harmful effects of [60]fullerene on mouse embryos in vitro Wu J, Liu W, Xue C, Znou S, Lan F, Bi L, Xu H, Yang X,
and in vivo. FEBS Lett 393:139–145. Zeng F-D. 2009. Toxicity and penetration of TiO2 nano-
Tupler R, Perini G, Green MG. 2001. Expressing the human particles in hairless mice and porcine skin after subchronic
genome. Nature 409:832–833. dermal exposure. Toxicol Lett 191:1–8.
Analysis of ZnO for sunscreens 41
Yamamoto O. 2001. Influence of particle size on the antibacterial Zhai H, Maibach HI. 2001. Effects of skin occlusion on percuta-
activity of zinc oxide. Int J Inorgan Mat 3:643–646. neous absorption: An overview. Skin Pharmacol Physiol 14:1–10.
Yamamoto O, Komatsu M, Sawai J, Nakagawa H. 2004. Effect of Zhu X, Wang J, Zhang X, Chang Y, Chen Y. 2009. The impact of
lattice constant of zinc oxide on antibacterial characteristics. ZnO nanoparticle aggregates on the embryonic development of
J Mater Sci Mater Med 15:847–851. zebrafish (Danio rerio). Nanotechnology 20:195103.
Yang H, Liu C, Yang D, Zhang H, Xi Z. 2008. Comparative study Zhu X, Zhu L, Duan Z, Qi R, Li Y, Lang Y. 2008. Comparative
of cytotoxicity, oxidative stress and genotoxicity induced by four toxicity of several metal oxide nanoparticle aqueous suspensions
typical nanomaterials: The role of particle size, shape and to zebrafish (Danio rerio) early developmental stage. J Environ Sci
composition. J Appl Toxicol 29:69–78. Health A Tox Hazard Subst Environ Eng 43:278–284.
Yang Z, Xie C. 2006. Zn2+ release from zinc and zinc oxide Zvyagin A, Zhao X, Gierden A, Sanchez W, Ross J, Roberts M.
particles in simulated uterine solution. Colloids Surf 2008. Imaging of zinc oxide nanoparticle penetration in human
B Biointerfaces 47:140–145. skin in vitro and in vivo. Journal of Biomedical Optics 13:064031.
Nanotoxicology Downloaded from informahealthcare.com by University of Toronto on 02/19/13
For personal use only.

You might also like