Skip to main content
Toxicology Reports logoLink to Toxicology Reports
. 2017 May 27;4:245–259. doi: 10.1016/j.toxrep.2017.05.006

Neurotoxic effect of active ingredients in sunscreen products, a contemporary review

Joanna A Ruszkiewicz a,, Adi Pinkas a, Beatriz Ferrer a, Tanara V Peres a, Aristides Tsatsakis b, Michael Aschner a
PMCID: PMC5615097  PMID: 28959646

Abstract

Sunscreen application is the main strategy used to prevent the maladies inflicted by ultraviolet (UV) radiation. Despite the continuously increasing frequency of sunscreen use worldwide, the prevalence of certain sun exposure-related pathologies, mainly malignant melanoma, is also on the rise. In the past century, a variety of protective agents against UV exposure have been developed. Physical filters scatter and reflect UV rays and chemical filters absorb those rays. Alongside the evidence for increasing levels of these agents in the environment, which leads to indirect exposure of wildlife and humans, recent studies suggest a toxicological nature for some of these agents. Reviews on the role of these agents in developmental and endocrine impairments (both pathology and related mechanisms) are based on both animal and human studies, yet information regarding the potential neurotoxicity of these agents is scant. In this review, data regarding the neurotoxicity of several organic filters: octyl methoxycinnamate, benzophenone-3 and −4, 4-methylbenzylidene camphor, 3-benzylidene camphor and octocrylene, and two allowed inorganic filters: zinc oxide and titanium dioxide, is presented and discussed. Taken together, this review advocates revisiting the current safety and regulation of specific sunscreens and investing in alternative UV protection technologies.

Keywords: Neurotoxicity, Sunscreen, Zinc oxide, Titanium dioxide, Octyl methoxycinnamate, Benzophenone-3, 4-Methylbenzylidene camphor, Octocrylene

1. Introduction

Sunscreen application is the main strategy used to prevent the maladies inflicted by the sun since the 1930s. Unfortunately, although global use of sunscreen is continuously on the rise, so is the prevalence of malignant melanoma − a cancer type which is mainly caused by sun exposure [1], [2], [3], [4]. There are several types of electromagnetic radiation emitted by the sun. One type − ultraviolet (UV) radiation − is composed of three wavelengths: UVA rays, which range at 320–400 nm and are not absorbed by the ozone layer, UVB rays, which range 290–320 nm and are partially absorbed by the ozone layer, and UVC rays, which are stopped by the ozone layer. The detrimental effects of exposure to UVA and UVB rays, which can cross the epidermis, have been reviewed and it was concluded that such exposure leads to reactive oxygen species (ROS) generation, DNA/protein/lipid damage, activation of various signal transduction pathways, compromised skin defense systems, altered growth, differentiation, senescence and tissue degradation, to name a few [5], [6], [7]. Two kinds of UV filters are currently being used in sunscreens for minimization of these adverse effects: organic (chemical) filters, e.g. octyl methoxycinnamate (OMC), benzophenone-3 (BP-3) or octocrylene (Table 1), which absorb light in the UV range, and inorganic (physical) filters, zinc oxide (ZnO) and titanium dioxide (TiO2), which scatter and reflect UV rays. Sunscreens are usually comprised of more than one of these UV filters: organic, inorganic or a mixture of both types, which gives broad-spectrum of protection. Beyond its debatable efficiency, questions regarding the main ingredients of different sunscreens are being raised in recent years, mainly about the prevalence of these ingredients in the environment and about their potential toxicity.

Table 1.

Organic UV filters.

International nomenclature of cosmetic ingredients (INCI) United States adopted name (USAN) Other names
UVB filters
 4-methylbenzylidene camphor* Enzacamene
 Homosalate Homosalate
 Isoamyl-p-methoxycinnamate Amiloxate
 Octyl dimethyl PABA Padimate O OD-PABA
 Octyl methoxycinnamate Octinoxate 2-ethylhexyl 4-methoxy cinnamate
 Octyl salicylate Octisalate 2-ethylhexyl salicylate
 p-aminobenzoic acid p-aminobenzoic acid 4-aminobenzoic acid, PABA
 Triethanolamine Trolamine salicylate



UVA filters
 Disodium phenyl dibenzimidazole tetrasulfonate Bisdisulizole disodium
 Butyl methoxydibenzoylmethane Avobenzone
 Menthyl anthranilate Meradimate
 Terephthalylidene dicamphor sulfonic acid Ecamsule Mexoryl SX



UVB-UVA filters
 Benzophenone-3 Oxybenzone 2-hydroxy-4-methoxybenzophenone
 Benzophenone-4 Sulisobenzone
 Benzophenone-8 Dioxybenzone
 3-Benzylidene camphora Mexoryl SD
 Bis-ethylhexyloxyphenol methoxyphenyl triazinea Bemotrizinol Tinosorb S
 Cinoxate Cinoxate
 Drometrizole trisiloxanea Mexoryl XL
 Methylene bis-benzotriazolyl Tetramethylbutylphenola Bisoctrizole Tinosorb M
 Octocrylene Octocrylene 2-ethylhexyl 2-cyano-3,3-diphenylacrylate
 Phenylbenzimidazole sulfonic acid Ensulizole
a

Not approved by the Food and Drug Administration, used in other parts of the world.

1.1. Human exposure and detrimental effects

Many factors might influence human exposure to UV filters: geographic location, season, lifestyle, gender or occupation, which means it can be highly individualized. For instance, a study in Australia showed 56% of people apply sunscreens at least 5 days per week, and 27% of people use it less frequently − 2 or fewer days per week [8] and a study in Denmark showed 65% of the sunbathers used one or more sunscreens [9].

Dermal exposure is the most relevant entry route of chemicals related to sunscreen use, however considering a common human behavior related to sunscreen application, e.g. eating and drinking with sunscreen applied on hands and lips, gastrointestinal or pulmonary exposure should also be considered [10], [11], [12]. The typically recommended mode of application (2 mg/cm2) [13] implies a single dose of sunscreen product may be as large as 40 g, assuming application on the total body surface (2 m2 for an average adult male), which for an average adult male weighting 78 kg and a typical concentration of about 10% of active ingredient in a commercial product, means maximum exposure around 50 mg/kg body weight (bw) [14]. Simple calculation suggests that with a maximum skin penetration up to 5% for some organic filters [15], the total amount of compound absorbed from a single application might be up to 200 mg, or 2.56 mg/kg bw, assuming an average bw of 78 kg for adult males. However, with application frequently thinner than recommended, partial body cover and different properties of compounds, these doses are usually much lower. For instance, a study on Australian population showed that the median daily amount of sunscreen applied was 1.5 g/day (range, 0–7.4 g/day) and the median quantity of sunscreen applied was 0.79 mg/cm2 [8], whereas sunbather in Denmark applied on average 0.5 mg/cm2 [9], in both cases it was less than half the amount needed to achieve the labeled sun protection factor.

Levels of UV filters found in human samples are usually low. In one epidemiological study, 2517 urine samples from United States (US) general population were analyzed for the presence of benzophenone-3 (BP-3), as part of the 2003–2004 National Health and Nutrition Examination Survey [16]; BP-3 was detected in 97% of the samples, with mean concentration of 22.9 ng/ml and 95th percentile concentration of 1040 ng/ml. In another study, investigating correlation between couples’ presence of urinary benzophenone-type UV filters and sex ratio of their offspring, the mean concentrations of these compounds ranged from 0.05 ng/ml to 8.65 ng/ml, with BP-3 as the most predominant among the study population (samples collected between 2005 and 2009 in Michigan and Texas) [17]. Interestingly, about nine times higher than previously reported levels of BP-3 (up to 13000 ng/ml, average around 200 ng/ml) were found in urine samples collected in 2007–2009 from Californian females, which is probably a result of specific demographics [18].

The experimental studies confirm substantial absorption and distribution of organic filters, whereas inorganic filters seem to penetrate the human skin in a minimal degree. When adults applied a sunscreen formulation containing 10% of BP-3, 4-methylbenzylidene camphor (4-MBC) and octyl methoxycinnamate (OMC) on a daily basis (2 mg/cm2) for a week, the mean urine concentrations for these ingredients were 60, 5, 5 ng/ml for females and 140, 7, 8 ng/ml for males, respectively [19]. At the same time, maximum plasma concentrations for these ingredients, reached 3–4 h after application, were 200, 20, 10 ng/ml for females and 300, 20, 20 ng/ml for males, respectively. Similar findings were reported following a 4-day exposure to these ingredients, which were detectable in the plasma of human males and females merely 2 h following application [20]. More data on human skin penetration and distribution of various UV filters, both organic and inorganic, can be found in recent reviews [21], [22], [15].

Of importance, some UV filters were also found in human milk samples. In a cohort study between 2004 and 2006, 54 human milk samples were analyzed; UV filters were detectable in 46 samples and levels were positively correlated with the reported usage of UV filter products [23]. Concentrations of ethylhexyl methoxy cinnamate (EHMC), octocrylene (OC), 4-MBC, homosalate (HMS) and BP-3 ranged 2.10–134.95 ng/g lipid, with EHMC and OC being most prevalent (42 and 36 positive samples, respectively) and an average of 7 positive samples for the other three [23]. In other study, levels of BP-3 in maternal urinary samples taken in gestational weeks 6–30 were positively correlated with the overall weight and head circumference of the baby [24]. These reports rise concerns about potential prenatal exposure and developmental toxicity of UV filters.

Besides intentional sunscreen application, additional routes might intensify human contact, namely occupational and environmental exposure. Workplace contact may be a source of substantial exposure to sunscreens, especially inorganic filters − nanoparticles (NPs) of ZnO and TiO2, which are frequently manufactured and stored as nanopowder. One study reported the presence of ZnO NPs in the work environment in an industrial scale plant in Japan. Electron microscopy analysis revealed the presence of a large number of submicron and micro-sized aggregated ZnO structures (concentrations not showed) [25]. Occupational exposures to TiO2 NPs have been reported more frequently and was summarized in a recent review, demonstrating that the respirable TiO2 concentration in the workers’ breathing zone might reach 150 μg/m3 [26]. Experimental studies suggest that with insufficient protection, inhalation of nanoparticles aerosol might result in pulmonary and systemic alterations. A single 10–30 min inhalation of a high dose (20–42 mg/m3) of ZnO NPs aerosol increased levels of the inflammatory cytokines interleukin (IL)-6, IL-8, and tumor necrosis factor α (TNF-α) in bronchoalveolar lavage fluid within 3 h after exposure in humans Kuschner et al., 1997. However, chronic, low-concentration exposure is more likely in workplaces, its effects are poorly known. Occupational exposure to sunscreen NPs was discussed more extensively in [12].

Environmental exposure is another way UV filters reach humans. Swimming in or drinking contaminated water might increase the contact, and thus absorption (through dermal and oral route) of these compounds. Recent data reviews indicate that the highest UV filter concentrations were found in rivers, reaching 0.3 mg/l for the benzophenone derivatives (e.g. BP-3), whereas ng to μg/l range were detected in lake and sea water. Moreover, lower levels (few ng/l) of organic UV filters were found in tap and groundwater [27], [28], [29], [31]. Organic UV filters accumulate in wastewater treatment plants (WWTPs) up to mg/l concentrations, and since conventional WWTPs are not able to remove them, they are consequently released into rivers, lakes and oceans Ramos et al., 2016. Swimming pools are sinks for UV filters and its chlorine byproducts, at the μg/l range, or higher. Analysis by Sharifan et al. suggested that small, urban swimming pools might contain significantly higher, than in natural waters, levels of UV filters: 2.85, 1.9, 1.78 and 0.95 g/l, respectively of EHMC, OC, 4-MBC and BP-3, which question their safety for using them people, especially children [30].

Due to the widespread application of these compounds in many daily-use products and growing awareness of the risk associated with the sun exposure, the market of UV filters increases every year. Thus, increasing usage, persistent input and accumulation in environment is becoming an issue of great concern because of threat to human health, but also to the environment. UV filters were found to be ubiquitous in many aquatic systems and aquatic biota. Occurrence and impact (including toxicity) of UV filters on environment have been reviewed extensively elsewhere [27], [28], [29], [31]. Aquatic organisms are frequently studied and UV filters were found at ng/g range in many of them, especially in fish and mussels, but also crustacean, mammals and aquatic birds [28]. In a study of the presence of several UV filters in Swiss lakes and rivers (which receive input from waste water treatment plants and recreational activity), and the fish which live inside them, water concentrations of BP-3, 4-MBC, EHMC and OC ranged 2–35 ng/l, while lower limit of detection (LOD) in fish for those compounds was 3–60 ng/g, with concentrations reaching as high as 166 ng/g for 4-MBC [11]. Later study showed even higher levels of BP-3, 4-MBC and EHMC (range 6–68 ng/l) in Swiss river, moreover substantial amount of EHMC was found in fish (up to 337 ng/g) and in cormorants (up to 701 ng/g), suggesting food-chain accumulation [10]. Many ecotoxicological studies addressed the potential damage of sunscreens and their components and in vitro experiments suggested that UV filters might be toxic for some aquatic microorganisms. UV filters were detected in nearshore waters around Majorca Island at variable concentrations: 53.6–577.5 ng/l for BP-3, 51.4–113.4 ng/l for 4-MBC, 6.9–37.6 μg/l for Ti, 1.0–3.3 μg/l for Zn, and various popular sunscreen formulations were shown to affect negatively the growth of local phytoplankton Chaetoceros gracilis, however at concentrations much higher than those detected in natural waters (EC50: 45–218 mg/l after 72 h treatment) Tovar-Sanchez et al., 2013. This is a typical observation. For instance, EC50 values of selected organic filters (e.g. BP-3, BP-4, EHMC, 4-MBC) in standardized toxicity assays on three aquatic species, Daphnia magna, Raphidocelis subcapitata and Vibrio fischeri, were in the mg/l range for all the species, which suggest minimal risk for these organism in their natural ecosystems [32]. However, like many researchers suggest, toxic effects of chronic, low-dose exposures cannot be ruled out and require further investigations [32]. Moreover, with increased usage and lack of efficient removal, environmental contamination will probably increase in the future. Recent report on coral bleaching showed that environmental contamination with BP-3 already poses a hazard to coral reef. The levels of BP-3 detected in coral reefs in the U.S. Virgin Islands (75–1400 μg/l) and Hawaii (0.8–19.2 μg/l) might lead to death of several local coral species with LC50: 8–340 μg/l and LC20: 0.062–8 μg/l (4 h exposure) [33].

Therefore, while sunscreens have been effective in protecting against a variety of UV-related pathologies, such as sunburns, actinic keratoses, squamous cell carcinomas and melanomas [34], growing popularity and thus, possibility for exposure questions their safety in environment and human health. Available data imply, that sunscreen compounds might block vitamin D synthesis or act as endocrine disruptor and lead to developmental toxicity. The effects of sunscreen on cutaneous synthesis of vitamin D induced by sunlight have been a subject of debate for recent years, however the newest analysis suggests, that normal usage of sunscreen by adults do not decrease cutaneous synthesis of vitamin D [35]. The endocrine disruptive and developmental toxicity of many organic UV filters in experimental models is well established, these filters seem to be associated with altered estrogen, androgen and progesterone activity, reproductive and developmental toxicity and impaired functioning of the thyroid, liver or kidneys, reviewed elsewhere [36], [37], [1], [38], [29]. Since many of UV filters were shown to cross the blood-brain barrier (BBB), the risk for neurotoxicity also occurs. In this review, the potential neurotoxicological effects of exposure to sunscreen have been discussed, as literature regarding the neurotoxicity of both organic and inorganic UV filters is presented.

2. Organic filters

Organic or chemical filters are the most popular and widely used in sunscreens and other cosmetic products. Data from 2003 indicate that over 80% sunscreen products contained OMC, 60% contained BP-3, and 20% contained octocrylene (OC) or HMS, whereas inorganic filters were present in around 20% of products [39]. Organic filters can be classified by the type of ultraviolet (UV) radiation they absorb, namely UVB, UVA or UVB-UVA filters (Table 1). As mentioned previously, the main route of human exposure is dermal absorption, however other routes and environmental exposure should be also considered. The last is particularly true for organic filters, which, due to their high lipophilicity could bioaccumulate in aquatic organism and reach humans through the food chain. Thus, they also are emergent as an environmental pollutant [40]. Chemical UV filters are easily absorbed by the skin and reach the systemic circulation, and accumulate in various tissues, as adipose tissue, liver and the brain [41], [42], [43], [44]. Their lipophilicity permits them to readily cross the BBB, nonetheless, the effect of organic UV filters in the central nervous system (CNS) has been yet to fully addressed. However, there is a wide range of in vitro and in vivo studies of the toxic effects of UV filters as endocrine disruptors. And since it is known that other chemicals classified as endocrine disruptors can impair neuronal transmission, synaptic plasticity and produce neurotoxic effects [45], chemical filters might potentially produce similar effect. The documented neurotoxic effects of organic UV filters have been described below and summarized in Table 2.

Table 2.

Neurotoxic effects of organic UV filters.

Compound Exposure model Experimental design Effect Reference
Octyl methoxycinnamate Wistar rats Oral (gavage) administration during gestation and lactation
500–1000 mg/kg/day
Decreased motor activity in female offspring, increased spatial learning in male offspring. [46]
Sprague-Dawley rats, female Oral (gavage) administration for
5 days
10–1000 mg/kg/day
Non-estrogenic interference within the rodent HPT axis; no changes in pre-proTRH mRNA in mediobasal-hypothalamus. [47]
Wistar rats In vitro incubation of hypothalamus isolated from adult rats, 60 min
0.263 μM
Decreased hypothalamic release of GnRH. Increased GABA release and decreased Glu production in males.
Decreased Asp and Glu production in females.
[48]
Wistar rats In vitro incubation of hypothalamus isolated from immature rats, 60 min
0.263 μM
Decreased hypothalamic release of LHRH. Increased GABA release in males, decreased Asp and Glu levels in females. [49]
SH-SY5Y neuroblastoma cell line 72 h
10−8–10−4 M
Decreased cell viability and increased caspase-3 activity. [50]
Benzophenone-3 Danio rerio Waterborne
14 days for adult
120 h for embryos
10–600 μg/l
Anti-androgenic activity: decreased expression of esr1, ar and cyp19b expression in the brain of males. [51]
Sprague-Dawley rats Dermal application
30 days
5 mg/kg/day
No changes in behavioral tests (locomotor and motor coordination). [42]
Rat primary cortical astrocytes and neurons 1–7 days
1–10 μg/ml
Decreased cell viability of neurons but not of astrocytes. [42]
SH-SY5Y neuroblastoma cell line 72 h
10−8–10−4 M
Decreased cell viability and increased caspase-3 activity. [50]
Benzophenone-4 Danio rerio Waterborne
14 days
30–3000 μg/l
Upregulated estrogenic-related genes: vtg1, vtg3, cyp19b in the brain of males. [52]
4-methylbenzyli-dine camphor Long Evans rats Oral (in diet) administration during mating, pregnancy, lactation, until adulthood of offspring
7, 24, 47 mg/kg/day
Impaired female proceptive and receptive sexual behavior. Altered expression of oestrogen- related gens in a sex- and region −dependent manner. [53], [54], [55]
Wistar rats Subcutaneous administration during pregnancy
20–500 mg/kg/day
Altered hypothalamic release of Glu and Asp in male offspring. Inhibited testicular axis in male offspring during the pre-pubertal stage and stimulated during peri-pubertal stage. [56]
Danio rerio Embryos exposed in medium
68 h
1–50 μM
Inhibited AChE activity, impaired early muscular and neuronal development. [57]
Neuro-2a mouse neuroblastoma cell line 45 min
0.1–100 μM
Inhibited AChE activity. [57]
SH-SY5Y neuroblastoma cell line 72 h
10−8–10−4 M
Decreased cell viability and increased caspase-3 activity. [50]
3-benzylidene camphor Long Evans rats Oral (in food) administration during mating, pregnancy, lactation, until adulthood of offspring
0.24–7 mg/kg/day
Impaired proceptive and receptive sexual behavior and disturbed estrous cycles of female offspring. Altered expression of oestrogen- related gens in a sex- and region-dependent manner. [55]
Octocrylene Danio rerio Waterborne
14 days
22–383 μg/l
Impaired expression of genes related with development and metabolism in the brain. [58]

Abbreviations: AChE: acetylcholine esterase; ar: androgen receptor; Asp: aspartate; cyp19b: cytochrome P450 aromatase b; esr1: estrogen receptor; GABA: gamma amino butyric acid; Glu: glutamate; GnRH: gonadotrophin-releasing hormone; HPT: hypothalamo-pituitary-thyroid; pre-proTRH: pre-pro-thyrotrophin-releasing hormone; vtg1, vitellogin 1; vtg3: vitellogin 3.

2.1. Octyl methoxycinnamate

Octyl methoxycinnamate (OMC) is a UVB filter also known as octinoxate and 2-ethylhexyl 4-methoxy cinnamate. This compound is approved as a cosmetic ingredient in US and in European Union (EU) in concentrations of 7.5–10% [1].

Dermal penetration of OMC has been measured in vitro, with values ranged from 0.2% to 4.5% of the applied dose, depending on the experimental conditions, however systemic absorption seems to be much lower. In humans, when a cream containing 10% OMC was applied to the entire body (40 g), OMC was absorbed through the skin and is detectable in blood (maximum concentrations 10 ng/ml in females and 20 ng/ml in males) and in urine (5 ng/ml in females and 8 ng/ml in males). Taking the highest detectable concentration (20 ng/ml) and assuming 4.7 l of blood, the systemic absorption represents only 0.002% of the applied dose [19].

Several studies indicated that OMC acts as an endocrine disruptor due to the ability to interfere with endocrine system at different levels [47], [59], [60]. In vitro and in vivo studies in rodents have shown that OMC have estrogen activity [61], [62]. In humans OMC exposure has minor, but statistically significant effects on the levels of testosterone and estradiol [19]. Moreover, some studies suggested that OMC can interact with the hypothalamo-pituitary-thyroid (HPT) axis [63]. Ovariectomized rats exposed to 57.5 mg/20 g body weight of OMC applied via food presented a decrease in thyroxine (T4) levels without changes in triiodothyronine (T3) or thyroid-stimulating hormone (TSH) levels [60].

OMC has also a non-estrogenic endocrine disrupting activity in the HPT axis absent altering the expression of pre-pro-thyrotrophin-releasing hormone (pre-proTRH) in the mediobasal hypothalamus, but affecting the axis in other points, when was administrated orally (10–1000 mg/kg/day) for 5 consecutive days [47]. Experiments with rats showed that OMC (0.263 μM) decreases the hypothalamic release of gonadotrophin-releasing hormone (GnRH) [48] and luteinizing hormone-releasing hormone (LHRH) [49] in vitro. Furthermore, in vitro experiments in hypothalamic cells from male and female adult rats showed that the same dose of OMC inhibited the release of neurotransmitters aspartate (Asp) and glutamate (Glu), but not gamma-aminobutyric acid (GABA) in females, whereas in males decreased Glu and increased GABA release [48]. Similar results were found in hypothalamus isolated from immature rats (pre-pubertal and peri-pubertal males and females) [49]. These results indicate that OMC disrupts the normal neuroendocrine mechanism in a sex-dependent manner. Moreover, a study of offspring of dams treated with OMC (500–1000 mg/kg/day) showed sex-dependent behavioral changes, namely decreased motor activity in females, but not in males, and improved spatial learning in males, suggesting that OMC can affect neuronal development, however the doses used in these experiments were extremely high, not relevant to possible human exposure [46]. Corroborating these observations, recent studies in neuroblastoma cell line (SH-SY5Y) demonstrated that exposure to high concentrations of OMC (0.01–100 μM) decreased cell viability and increased apoptosis, however effective concentrations were not observed in vivo [50].

2.2. Benzophenone-3

Benzophenone-3 (BP-3, oxybenzone) is a common organic filter used in sunscreens and other personal care products (nail polish, lotions, lipsticks) in a maximum allowed concentration of 6% in US. It is used as broad-spectrum UV filter due to absorption of both UVB and short UVA rays [1].

BP-3 applied topically in human can cross the skin by direct penetration through the intercellular laminae of the stratum corneum (SC) or by passive diffusion by high-concentration gradient and then reach the blood [64]. When 25 volunteers applied a commercially available sunscreen containing 4% BP-3 for 5 days, their urine samples showed that approximately 4% of BP-3 is absorbed into the system [65]. BP-3 was detected in more than 80% of urine samples of healthy Danish children and adolescents (median concentration 0.92 ng/ml) [66]. Repeated (4 days) topical applications (2 mg/cm2 of sunscreen formulation) of BP-3 resulted in urine levels up to 81 ng/ml and plasma levels up to 238 ng/ml [20]. Moreover, another concern relates to the fact that the simultaneous application of some insect repellents components such as N, N-diethyl-m-toulamide (DEET) and BP-3 can enhance skin penetration of each other when jointly applied [42]. Once BP-3 is in the systemic circulation, it is transported to different organs. BP-3 is a highly lipophilic, and in rats it has been detected in liver [41], [42], [44], [43] and in brain (15.5–34.1 ng/g) [42]. High concentrations of BP-3 were also detected in adipose tissue after topical administration [41].

High, not environmentally relevant concentration of BP-3 (up to 1000 μg/l) were shown to disrupt the neuro-endocrine system in fish [67], [68]. BP-3 (waterbone exposure 10–600 μg/l, where the lowest concentration represents the worst-case, environmentally relevant concentration) impaired the sexual behavior of Danio rerio zebrafish adult males by decreasing the expression of androgenic genes: estrogen receptor 1 (esr1), androgen receptor (ar) and cytochrome P450 aromatase B (cyp19b) in the brain at concentration 84 μg/l [51]. Whereas topical administration of BP-3, at dosage which mimics possible human exposure (5 mg/kg/day for 30 days) in male and female Sprague-Dawley rats did not affect locomotor activity and behavioral test, nor did it produce neurological deficits [42]. Moreover, no effect on rat primary cortical astrocyte cultures were detected when cells were incubated with low, physiological concentrations (0.1–10 μg/ml) of BP-3 for up to 7 days [42]. However, studies in rat primary cortical neuronal cultures [42] and SH-SY5Y neuroblastoma cell line [50] showed decreased cell viability after BP-3 treatment at moderate concentrations (e.g. 1–10 μg/ml).

2.3. Benzophenone-4

Benzophenone-4 (BP-4, sulisobenzone) is frequently used as UV absorber at concentration up to 10% [1].

BP-4 was found in human placenta (0.25–5.41 ng/g), suggesting efficient skin penetration and accumulation, which may lead to exposure of human embryos and fetuses [69]. BP-4, like BP-3 is a benzophenone derivative, yet its potency as an estrogenic disruptor has been not well defined.

In zebrafish, adult males exposed to high concentrations (3000 μg/l) of BP-4 for 14 days displayed estrogenic activity by up-regulation of estrogenic-related genes: vitellogin 1 (vtg1), vitellogin 3 (vtg3) and the cyp19b in the brain, however lower dosages did not induce changes. In contrast, in the liver, some of these genes (vtg1, vtg3) were down-regulated [52]. No other effects in the nervous system were reported.

2.4. 4-methylbenzylidene camphor

4-methylbenzylidene camphor (4-MBC) or enzacamene is an organic camphor derivative used as a UVB filter in sunscreen and other cosmetic products. Although the compound is not approved by the Food and Drug Administration (FDA), other countries allow it usage at maximum concentration of 4% [1].

4-MBC is a high lipophilic component which can be absorbed through the skin and was found in human tissues, including placenta [70]. Repeated (4 days) topical applications (2 mg/cm2 of sunscreen formulation) of 4-MBC resulted in urine levels up to 4 ng/ml and plasma levels up to 18 ng/ml [20]. When orally administrated to rats, 4-MBC reaches the liver where is metabolized to 3-(4-carboxybenzylidene) camphor and 3-(4-carboxybenzylidene) hydroxycamphor [71]. 4-MBC exhibits a toxic activity as estrogenic endocrine disruptor [62], [59], [68]. Moreover, in vivo studies suggest that 4-MBC affected the thyroid axis [63].

Several studies described the effects of 4-MBC on developing neuroendocrine system. Rats exposed to 4-MBC (7–47 mg/kg) in diet before mating, during pregnancy and lactation, and in the offspring until adulthood, showed a region- and sex-dependent alteration in the oestrogenic genes in the brain [55], [54], [53]. For instance, the expression of progesterone receptor (PR) was decreased in the ventromedial hypothalamic area of 4-MBC-treated females, but not in males [55]. In addition, females showed impaired proceptive and a non-receptive sexual behavior after 4-MBC exposure [55]. Female sexual behavior was significantly impaired at the lowest doses studied 7 mg/kg/day, which resulted in rat milk concentration of 208.6 ng/g lipid, which is over 10 times higher than value (19 ng/g lipid) found in human milk [55]. Subcutaneous administration of high dosages (up to 500 mg/kg/day) during pregnancy and lactation altered the hypothalamic secretion of excitatory amino acids Glu and Asp in male offspring. These neurotransmitters play a role as stimulators of gonadal axis, thus the observed changes are consistent with alterations in sexual development of male offspring, affecting pre-pubertal stage, but stimulating the peri-pubertal stage [56]. In addition, 4-MBC has been reported to have an acetylcholinesterase (AChE) inhibitory activity. Zebrafish embryos exposed to 15 μM 4-MBC for 3 days showed abnormal axial curvature and exhibited impaired motility. 4-MBC also impaired muscle development and axon pathfinding [57]; however, the dose used in the study was significantly higher than those detected in environmental aquatic media. Inhibition of AChE activity was also observed in mammalian Neuro-2a cells exposed to 10 and 100 μM for 45 min, indicating a possible mechanism for the 4-MBC-induced muscular and neuronal defects [57]. 4-MBC (up to 100 μM) has been shown recently to decrease cell viability and induce apoptosis in neuroblastoma cell line (SH-SY5Y), suggesting possible neurotoxic effects, however again, effective concentrations were not observed in vivo [50].

2.5. 3-benzylidene camphor

3-benzylidene camphor (3-BC) is a lipophilic compound closely related to 4-MBC. It is used in sunscreen products in EU, at a maximal concentration of 2% [1].

After topical application to rats for 65 days (60–540 mg/kg/day) 3-BC was detected in all analyzed tissues, including the brain (concentration 0.13–1.2 μg/g), suggesting that similar disposition and distribution may occur in humans [72]. Though not detectable in urine of Danish children [66], the compound was found in human placenta [70].

Analogous to 4-MBC, 3-BC has also been described as an estrogenic disruptor [59], [73]. Moreover, it has been reported that 3-BC can affect the CNS. Rats pre- and postnatally treated with 3-BC (0.24–7 mg/kg/day) showed region- and sex-specific response in expression of genes involved in sexual behavior: PR, estrogen receptors (ERa, ERb), and steroid receptor coactivator-1 (SRC-1) in the brain [55].

2.6. Octocrylene

Octocrylene (OC) is an ester belonging to the cinnamates family and is present in sunscreen and daily care cosmetic products at a maximal concentration of 10%. It can absorb UVB and high energy components of UVA radiation [74]. To date there are few studies on its accumulation and toxicity, especially in aquatic organism [75], [76], [77].

Zebrafish embryos and adult male exposed to environmentally relevant concentrations of OC in water (22–925 μg/l) absorbed and accumulated this compound. Moreover, the microarray analysis from adult zebrafish male exposed to OC (383 μg/l) showed major impairment in the expression of 628 genes in the brain regulating mainly developmental processes and 136 genes in the liver, responsible mainly for metabolism [58].

3. Inorganic filters

Inorganic (physical) ingredients used in modern sunscreens include metal oxide particles, typically titanium dioxide (TiO2) and zinc oxide (ZnO), which occurs typically at 5–10% concentration (maximum allowed is 25%). While chemical filters still dominate in sunscreen products, the usage of physical compounds is constantly growing. One of the reasons is that they have a higher spectrum of protection − TiO2 is very effective in absorbing UVB, while ZnO absorbs mainly the UVA range, and the combination of both particles provides a broad UV protection. Other advantages of physical filters are lack of skin sensitization and limited skin penetration [78]. However, these mineral filters, when in normal pigment size range (200–400 nm for ZnO, 150–300 nm for TiO2) have poor particle dispersion, which makes them difficult to apply; they also reflect and scatter light, which result in undesirable visible white film on the skin. With nanotechnology, these materials can be reduced to nanoparticles (NPs) (<100 nm) which are easier to apply and are transparent on the skin [12]. Nevertheless, with micronization some properties are changed − they may be more bioreactive and easier penetrate the skin and other tissues, leading to concerns about their safety use. Moreover, part of the absorbed UV radiation can generate free radicals on the surface of metal oxides in the presence of water and this photocatalytic activity increases with decreasing NPs size. NP-induced cyto- and genotoxicity has been associated with increased photocatalytic activity, leading to increased production of free radicals [79]. Despite increased awareness of nanomaterials toxicity, the nanoneurotoxicity is a relatively new field with numerous data gaps awaiting improvements. One of the main reasons for this is the lack of reliable methods for NPs detection and quantification. Only estimates and predictions about NPs concentration in natural environments are available, and they suggest that TiO2 might be present in the range 0.7–24.5 μg/l, whereas ZnO might reach higher levels, up to 76 μg/l [80]. Analogously, NPs accumulation and physiological concentrations are difficult to assess; thus, most studies report changes in Zn and Ti ion levels only. This also raises questions regarding the relevance of predominantly high-dose exposures used in toxicological studies. To date, most studies attesting to neurotoxic effect of NPs have been carried out in acutely high concentration exposure scenarios, and their relevance to “real-life” exposure scenarios needs to be further assessed.

3.1. Zinc oxide

Zinc oxide nanoparticles (ZnO NPs) are used not only in sunscreens, but also in pigments (UV-absorbers, paintings) and electronic equipment (thin film transistors, semi-conductors, liquid crystal displays, light-emitting diodes) due to their exceptional optoelectronic, piezoelectric, ferromagnetic and optical properties. Moreover, their antiseptic activity makes them potentially useful in treatment of bacteria-related infections or diseases [81]. As their commercial utilization has increased, wider application raises the potential risk of human exposure [82].

3.1.1. ZnO NPs absorption and transport across the BBB

Several in vitro and in vivo studies evaluated the fate and toxicity of ZnO NPs from different exposures: dermal, gastrointestinal or pulmonary. Dermal absorption is a major route of ZnO NPs exposure from sunscreen application. Most studies demonstrated that ZnO NPs did not penetrate into deeper layers of the skin (SC) [83], [84], [22], [85], [86]. However, some data indicated that ZnO NPs penetrated the skin to a limited extent. A small increase of zinc ions (Zn2+) in the blood and urine was observed in humans exposed to ZnO NPs-containing sunscreen products for five constitutive days via healthy skin [87]. Human skin in vitro was shown to absorb 0.34% of ZnO NP after 72 h [88]. In general, penetration ability of NPs increases when the skin barrier is damaged, pursuant to sunburn, skin disease or physical damage. ZnO NPs were found to better penetrate tape-stripped, lesioned or wounded, rather than healthy human skin [85], [89]. Moderate skin sunburn increased the penetration of ZnO NPs in pigs, however transdermal absorption was not detected [90]. In vitro studies reported similar findings, only a limited number of ZnO NPs were found on the outer surface of the SC, and no particles were observed in the deeper SC layers [83], [84]. Generally, the risk of ZnO NPs exposure from dermal absorption is rather low, however, considering a common human behavior related to sunscreen application, e.g. eating and drinking with sunscreen applied on hands and lips, gastrointestinal or pulmonary exposure should also be considered, moreover, as mentioned previously, the occupational exposure might be of high concern for some people [12].

Inhalation might be specifically associated with increased brain exposure, since the olfactory nerves can directly transport particles into the brain. In fact, Kao et al. observed the translocation of ZnO NPs into the brain following nasal administration (6 h airborne exposure) in a Sprague Dawley rats [91]. In healthy human adults inhaling 500 μg/m3 of ZnO NPs for 2 h, the results were below the threshold for acute systemic effects on the respiratory, hematologic, and cardiovascular endpoints [92]. Other studies have shown that various NPs can enter the brain across the BBB [81], [93]; however, a limited number of studies address this issue for ZnO NPs. The BBB was found to be intact in rats after repeated oral administrations of ZnO NPs for 28 days (500 mg/kg) [94], however the presence of ZnO NPs in the rat brain was observed after oral administration for 21 days (500 mg/kg) [95]. Moreover, Yeh et al. (2012) showed increased 65Zn accumulation in the mouse brain up to 10 days after single-dose (120 g) intravenous injection of small (10 nm) 65ZnO NPs [96]. In adult mice, neuronal NPs localization was observed for several days after single oral (gavage) administration of 3 mg of fluorescent ZnO NP. Decreased fluorescent signal over time is consistent with biodegradation or elimination of NPs from the brain [97]. Additional studies are needed to investigate the brain penetration capacity of ZnO NPs.

Other reviews discuss absorption, distribution, metabolism and excretion of ZnO NPs in humans and experimental models more extensively [12], [79], [82]. To date, data available indicate that ZnO NPs can be absorbed via different routes and distributed to a range of organs, including the brain and placenta. Distribution depends on the size of ZnO NPs, the dose, time and route of exposure. The fate of ZnO NPs remains unclear; most data suggest that ZnO NPs decompose in medium or in cells and release Zn2+ which are responsible for toxic effects. However, this issue, together with the risk of long-term exposure and absorption via healthy vs. damaged skin remain to be established.

3.1.2. Neurotoxic effects in vivo

Although increasing number of studies aimed to investigate the potential toxicity of ZnO NPs in different cell types and animal systems [98], [82], [12], little is known about their neurotoxic effects (Table 3), especially in vivo. ZnO NPs exposure was shown to induce neurobehavioral changes in experimental animals. Impaired learning and memory abilities, and hippocampal pathological changes were demonstrated in old (18 months) mice following ZnO NPs exposure (intraperitoneally, i.p., 5.6 mg/kg, three times per week for four weeks) [104]. The spatial learning and memory ability was attenuated in ZnO NPs-treated (i.p. 4 mg/kg, biweekly for 8 weeks) Wistar rats. The exposed animals exhibited prolonged escape latency in the Morris water maze (MWM), and enhanced long-term potentiation (LTP), but not sufficient depotentiation in the dentate gyrus (DG) region of the hippocampus [99]. ZnO NPs administered i.p. for several days ameliorated the behavioral and cognitive impairment in young Swiss male mice with depressive-like behaviors, suggesting that they may affect neuronal synaptic plasticity [105]. Subcutaneous administration of ZnO NPs in pregnant ICR mice at gestation day (GD) 5, 8, 11, 14 and 17 (100 μg/day) affected dopamine (DA), 5-hydroxytriptamine (5-HT) and their metabolites’ levels in a 6-week old offspring [106]. This observation questions the safety of ZnO NPs exposure during pregnancy, potential transfer through placenta and the effect on developing brain. In contrast, single intravenous injection of ZnO NPs (25 mg/kg) did not affect locomotor activity, exploratory behavior, spatial working memory or neurotransmitter: norepinephrine (NE), epinephrine (EPI), DA, and 5-HT levels in adult male Wistar rats 14 days after injection, despite the plasma and brain Zn2+ levels increased in treated group [100]. Sub-acute ZnO NPs treatment (25 mg/kg, 10 days) resulted in minimal effect on emotional behavior (e.g. unaffected anxious index), but showed alteration in trace elements homeostasis in rat brain homogenates: decreased levels of iron (Fe2+) and calcium (Ca2+), while Zn2+, sodium (Na+) and potassium (K+) concentrations remained unchanged [101].

Table 3.

Neurotoxic effects of ZnO NPs.

Compound Exposure model Experimental design Effect Reference
ZnO NPs Wistar rats Intraperitoneal injection biweekly, 8 weeks
4 mg/kg
Attenuated spatial cognition capability, enhanced long-term potentiation. [99]
Wistar rats Intravenous injection single dose
25 mg/kg
Increased brain Zn concentrations; no changes in neurotransmitter levels, locomotor activity, exploratory behavior or spatial working memory. [100]
Wistar rats, male Intraperitoneal injection, 10 days
25 mg/kg/day
Decreased iron and calcium, but not Zn, sodium and potassium levels in rat brain homogenates; unchanged emotional behavior. [101]
Wistar rats, male Oral (gavage)
7 days
600 mg/kg
Elevated TNF-α, IL-1β, IL-6, CRP, MDA, decreased GSH and SOD levels, CAT, and GPx activity. [102]
Sprague-Dawley rats Oral
13 weeks
134.2, 268.4, 536.8 mg/kg/day
Increased Zn levels in the brain of male rats. [103]
C57BL/6J mice, male Intraperitoneal injection 3 times per week, 4 weeks
5.6 mg/kg
Impaired learning and memory abilities, suppression of cAMP/CREB signaling pathway. [104]
Swiss albino mice, male Intraperitoneal injection every other day, 8 times
5.6 mg/kg
Improved behavioral and cognitive impairment in mice with depressive-like behaviors. [105]
Swiss albino mice, male Oral
21 days
500 mg/kg
Elevated ROS levels, altered antioxidant system, increased DA and NE levels, presence of ZnO NPs in neurons. [95]
ICR mice, pregnant female Subcutaneous at GD 5, 8, 11, 14, 17
100 μg/day
Changed DA, 5-HT and their metabolites levels in a 6-week old offspring. [106]
Cyprinus carpio Waterborne
1–14 days
0.5, 5, 50 mg/l
Changed CAT, SOD, GPx activity, GSH levels and lipid peroxidation. [107]
Prochilodus lineatus Waterborne
5, 30 days
7, 70, 700 μg/l
Increased protein oxidative damage, decreased AChE activity. [108]
Apis mellifera carnica Oral (food)
10 days
0.8 mg Zn/ml
Decreased brain weight and increased brain AChE and GST activity. [109]
Isolated rat neurons 1 mg/ml Increased the opening number of sodium channels, delayed rectifier potassium channels, enhanced excitability of neurons. [110]
Rat primary neurons 24 h
1–100 μg/ml
Concntration-dependent cytotoxicity, disrupted cell membranes, DNA damage. [111]
Mouse neural stem cells 24 h
3–24 ppm
Concentration-dependent decrease in cell viability; apoptosis, necrosis, release of zinc ions. [112]
RCS96 rat Schwann cells 6–48 h
4–400 μg/ml
Concentration- and time-dependent decrease in cell viability; apoptosis and necrosis, G2/M phase cell cycle arrest, release of Zn ions. [113]
Human olfactory
neurosphere-derived cells
2–24 h
10–80 μg/ml
Decreased cell viability, activation of numerous pathways associated with stress, inflammation and apoptosis. [114]
RCG-5 rat retinal ganglion cells 4–72 h
2.5–10 μg/ml
Concentration- and time-dependent decrease in cell proliferation; cell cycle arrest, ROS generation, increased caspase-12, decreased bcl-2 and caspase-9. [115]
RCG-5 rat retinal ganglion cells 6–72 h
2.5–10 μg/ml
Decreased mitochondrial membrane potential, increased ROS production, increased caspase-12. [116]
RCG-5 rat retinal ganglion cells 4–72 h
2.5–10 μg/ml
Decreased expression and activity of the plasma membrane calcium ATPase, disrupted intracellular calcium homeostasis, increased ROS production. [117]
PC12 rat pheochromocytoma and SH-SY5Y human neuroblastoma 24 h
10–10000 μM
Decreased cell viability, mitochondrial impairment, internalization of ZnO NPs in membrane-bound vesicles. [91]
SH-SY5Y human neuroblastoma 6, 12, 24 h
5–30 mg/ml
Concentration- and time-dependent decrease in cell viability; apoptosis via the PI3 K/Akt/caspase-3/7 pathway and necrosis by LOX-mediated ROS production. [118]
SH-SY5Y human neuroblastoma 3–48 h
10–80 μg/ml
Concentration- and time-dependent decrease of cell viability, apoptosis and cell cycle alterations, genotoxicity: micronuclei, H2AX phosphorylation, DNA damage. [119]
U87 human brain tumor 24 h
1–200 μg/ml
Concentration-dependent cytotoxicity e.g. increased formation of micronuclei. [120]
Rat primary astrocytes 6, 12, 24 h
4, 8, 12 μg/ml
Reduced cell viability, increased LDH release, stimulated ROS generation, caspase-3 activation, decreased MMP, phosphorylated JNK, ERK, p38 MAPK. [121]
C6 glia cells 3, 6, 24 h
5–80 μg/ml
Time- and concentration-dependent cytotoxicity, apoptosis and increased ROS production. [122]
A172, U87, LNZ308, LN18, LN229 glioma cell lines and normal human astrocytes 24 h
1, 5, 10 mmol/l
Cytotoxicity and ROS generation in glioma lines, but not in normal human astrocytes. [123]
N9 mouse microglial cell line 5–60 min; 1–24 h
1–100 μg/ml
Increased intracellular calcium and ROS levels, decreased intracellular ATP level, upregulated apoptosis markers. [124]
BV-2 mice microglia cell line 2–24 h
10 μg/ml
Increased cytotoxicity; activated PINK1/parkin-mediated mitophagy. [125]

Abbreviations: 5-HT: 5-hydroxytriptamine; Akt: protein kinase B; cAMP: cyclic adenosine monophosphate; CAT: catalase; CREB: cAMP response element binding protein; CRP: c-reactive protein; DA: dopamine; ERK: extracellular signal-related kinase; GSH: glutathione; GPx: glutathione peroxidase; GST: glutathione-S-transferase; H2AX: H2A histone family member X; IL-1β: interleukin-1β; IL-6: interleukin-6; JNK: c-Jun N-terminal kinase; LDH: lactate dehydrogenase; LOX: lipoxygenase; MDA: malondialdehyde; MMP: mitochondrial membrane potential; NE: norepinephrine; NPs: nanoparticles; p38 MAPK: p38 mitogen-activated protein kinase; PINK1: PTEN-induced putative kinase 1; PI3 K: phosphoinositide 3-kinase; ROS: reactive oxygen species; SOD: superoxide dismutase; TNF-α: tumor necrosis factor α; Zn: zinc; ZnO: zinc oxide.

Disrupted ion homeostasis is an important pathomechanisms of neurotoxicity, and ZnO NPs might affect it. Long-term (13 weeks) oral ZnO NPs administration (134.2, 268.4, 536.8 mg/kg/day), resulted in detection of slightly, but significantly higher Zn2+ levels in the brain of male rats (but not in female) [103]. In isolated rat hippocampal CA3 pyramidal neurons the ZnO NPs solution (1 mg/ml) was shown to enhance the current amplitudes of INa and IK by increasing the opening number of sodium channels, delaying rectifier potassium channels, and enhancing the excitability of neurons, leading to intracellular Na+ accumulation and K+ efflux. These might disturb the ionic homeostasis and the physiological functions of neurons [110].

Oxidative stress and disrupted antioxidant system is another effect observed in brains of ZnO NPs-treated animals. Oral ZnO NPs (500 mg/kg) administration for 21 consecutive days resulted in elevated ROS levels and altered antioxidants: glutathione (GSH) levels, superoxide dismutase (SOD), glutathione peroxidase (GPx), and glutathione S-transferase activity (GST), in both the brain and liver of male Swiss albino mice [95]. Combined with increased DA and NE levels in the cerebral cortex, these results suggest a neurotoxic potential for ZnO NPs [95]. Changes in CAT, SOD, GPx activity, GSH levels and lipid peroxidation was also observed in the brain and other organs of juvenile carp (Cyprinus carpio) exposed to waterborne ZnO NPs (0.5, 5, 50 mg/l) for 1, 3, 7, 10 and 14 days [107]. Exposure to environmentally relevant concentrations of ZnO NPs (7, 70, 700 μg/l) for 5 and 30 days led to increased protein oxidative damage in the brain and gills, but not in the liver, and decreased AChE activity in the brain and muscle of Prochilodus lineatus juvenile fish [108]. Honey bees (Apis mellifera carnica) exposed to ZnO NPs (0.8 mg Zn/ml) in food for 10 days showed decreased brain weight and increased brain AChE and GST activity [109]. Week-long oral administration of ZnO NPs (600 mg/kg) to male Wistar rats resulted in decreased brain CAT, GPx, and GR activities, decreased GSH and SOD levels, but elevated malondialdehyde (MDA) level and inflammatory markers: TNF-α, IL-1β, IL-6, C-reactive protein (CRP). The neurotoxic effects were partially reversed by the antioxidant and anti-inflammatory compound, hesperidin [102]. The pro-oxidant and pro-inflammatory effect of ZnO NPs was also observed in the serum and the brain of mice injected with ZnO NPs (i.p., 5.6 mg/kg) three times per week for four weeks [104]. In this study the suppression of cAMP/CREB signaling pathway was also identified: the contents of hippocampal cyclic adenosine monophosphate (cAMP), cAMP response element binding protein (CREB), phosphorylated CREB and synapsin I, were decreased in ZnO NPs-treated mice in an age-dependent manner [104].

3.1.3. Neurotoxic effects in vitro

The neurotoxic effect of ZnO NPs in vitro has been also evaluated, demonstrating oxidative stress- and apoptosis-related cytotoxicity. Deng et al. [112] have demonstrated that ZnO NPs impaired viability of neural stem cells (NSCs) in a concentration-, but not size-dependent manner. Twenty-four-hour exposure of concentrations higher than 12 ppm induced apoptosis and necrosis in the NSCs. Authors suggested that observed changes might result from the Zn2+ dissolved in solution or intracellularly, rather than from NPs, since ZnO NPs were not detectable in apoptotic cells, and similar cytotoxicity was observed after treatment with ZnCl2 [112]. In primary rat astrocytes ZnO NPs exposure (4, 8, 12 μg/ml for 6–24 h) was found to reduce cell viability, increase lactate dehydrogenase (LDH) release, stimulate ROS generation, and elicit caspase-3 activation in a concentration- and time-dependent manner [121]. Apoptosis was shown by nuclear condensation and poly(ADP-ribose) polymerase-1 (PARP) cleavage. ZnO NPs stimulated the phosphorylation of c-Jun N-terminal kinase (JNK), extracellular signal-related kinase (ERK), and p38 mitogen-activated protein kinase (p38 MAPK). A decrease in mitochondrial membrane potential (MMP) and increase in the expression of Bax/Bcl-2 ratio was also observed, suggesting mitochondria-mediated apoptosis [121]. ZnO NPs (1–100 μg/ml, 24 h) induced concentration-dependent cytotoxicity, disrupted cell membranes and DNA damage in rat primary neuronal cells, human fibroblasts and A549 cells, but not in HepG2 cells and human skin keratinocytes [111]. A time- and concentration-dependent cytotoxicity characterized by apoptosis and increased ROS production was also observed in ZnO NPs-treated (5–80 μg/ml) C6 glial cells [122]. ZnO NPs exposure (1–100 μg/ml) resulted in increased intracellular Ca2+ and ROS levels, decreased intracellular ATP level and upregulated apoptosis markers in mouse microglial cell line [124]. ZnO NPs (1, 5, 10 mmol/l, 24 h) evoked cytotoxicity in the human glioma cell lines (A172, U87, LNZ308, LN18, LN229), but not in normal human astrocytes. Cytotoxicity observed in the glioma cells was related to increased ROS generation, and N-acetyl-l-cysteine (NAC) treatment decreased the cytotoxic effect of the ZnO NPs in these cells [123]. ZnO NP-induced cytotoxicity was also observed in microglia (BV-2 cells) exposed to 10 μg/ml ZnO NPs for 2–24 h [125]. ZnO NPs induced parkin protein translocation from the cytoplasm to the mitochondria, implying the involvement of mitophagy in ZnO NPs-induced toxicity [125].

The neurotoxicity of ZnO with four different hierarchical architecture: monodispersed spherical NPs (35 nm), hollow ZnO microspheres (2.7 mm), and larger, prism- and flower-like structures, was evaluated in RSC96 rat Schwann cells [113]. Cells were treated with ZnO at doses 4, 8, 40, 80, 400 μg/ml for 6, 12, 24, and 48 h. ZnO NPs and microspheres displayed significant cytotoxic effects on Schwann cells in concentration- and time-dependent manners, whereas no or low cytotoxic effect was observed when the cells were treated with the prism-like and flower-like ZnO. Cell apoptosis and G2/M cell cycle arrest were observed when RSC96 Schwann cells were exposed to ZnO nanoparticles and microspheres at a dose of 80 μg/ml for 12 h. The time-dependent increase of Zn2+ concentration in the culture media suggests that the cytotoxic effects were associated with the decomposition of ZnO hierarchical architecture and the subsequent release of Zn2+, and not exclusively to the nanoparticulated fraction [113].

Neurotoxicity of ZnO NPs was examined in rat pheochromocytoma PC12 and human neuroblastoma SH-SY5Y cells, showing significant cell loss after 24 h treatment at concentration of 0.1 mM in PC12 cells and 1 mM in SH-SY5Y cells [91]. Moreover, when the PC12 cells were treated with 1 mM (81.4 μg/ml) ZnO NPs for 10 min, the endocytosis of ZnO NPs was observed and increased cellular Zn2+ levels indicated that ZnO NPs may be converted to Zn2+ in endosomes, and then be mobilized into the cytoplasm, leading to Zn2+ dyshomeostasis [91]. The cytotoxic and genotoxic effects of ZnO NPs in SHSY5Y cells under different exposure conditions were also investigated by Valdiglesias et al. [119]. Despite the results showed that ZnO NPs (10–80 μg/ml) do not enter the neuronal cells, their presence in the medium induced decrease in cell viability, apoptosis, cell cycle alterations, and genotoxicity, including micronuclei production, H2AX (H2A histone family, member X) phosphorylation and DNA damage (primary and oxidative) in a concentration- and time-dependent manner. Unlike in previously described studies, free Zn2+ released from the ZnO NPs was not responsible for the viability decrease [119]. Exposure of SH-SY5Y cells to ZnO NPs (10, 15, 20, 25, 30 mg/ml) resulted in neurotoxicity, as confirmed by LDH activity assay, mitochondria toxicity test (MTT) and Muse™ cell viability assay. Allopurinol, NAC and α-tocopherol protected from ZnO NP-induced cytotoxicity. Electron microscopy revealed typical necrotic characteristics, such as swelling or loss of cell organelles and rupture of the cytosolic or nuclear membrane at 12 h and 24 h after ZnO NPs exposure. Apoptotic changes (annexin V and caspase-3/7 activities) were evident at 12 h and 24 h, but not 6 h after exposure to 15 mg/ml ZnO NPs. PI3 kinase (PI3 K) and p-Akt/Akt (protein kinase B) activities induced by ZnO NPs were significantly decreased by esculetin (antioxidant) or LY294002 (PI3 K inhibitor). Esculetin reduced the production of ROS and the depletion of antioxidant enzymes induced by ZnO NPs. ZnO NPs induced apoptosis via the PI3 K/Akt/caspase-3/7 pathway and necrosis by lipoxygenase (LOX)-mediated ROS production [118]. Treatment with ZnO NPs (1–200 μg/ml) induced cytotoxicity (e.g. increased formation of micronuclei) in the human brain tumor U87 cells in a concentration-dependent manner, but did not affect normal human HEK cells [120]. Different types of ZnO NPs (coated <200 nm and uncoated <30 nm) (10–80 μg/ml, incubated for 2, 6 or 24 h) induced cytotoxicity in human olfactory neurosphere-derived (hONS) cells via mechanisms associated with cell stress, inflammation and apoptosis [114]. Changes in cytokines IL-6 and IL-8 secretion, increase in caspase-3/7 activity, and phosphorylation of key proteins involved in signaling pathways: MAPK/ERK (pMEK, pERK, pJNK, p-cJUN, p-p38), Akt (pAkt, pBAD) and NF-κB (pNF-κB, pI-κB) has been demonstrated. Microarray RNA analysis revealed that short-term (2 h) exposure to ZnO NPs activated pathways involved in cellular stress responses (e.g. upregulation of Nrf2-mediated oxidative stress response pathway), whereas longer (6 h) exposure affected pathways more related to cell injury and repair. Of note, the cellular response was dependent on NPs surface coatings [114]. ZnO NPs (2.5–10 μg/ml) induced cytotoxicity in rat retinal ganglion cells (RGC-5) and inhibited cell proliferation in a time- and concentration-dependent manner. Moreover, ZnO NPs treatment led to cell cycle arrest of S and G2/M phases, ROS production and increased level of caspase-12 and decreased levels of bcl-2 and caspase-9 [115]. Further, the same group showed that ZnO NPs decreased the MMP in RGC-5 cells [116], ZnO NPs (2.5–10 μg/ml) have been shown to decrease the expression and activity of the plasma membrane calcium ATPase, increase intracellular Ca2+ level and disrupt the intracellular calcium homeostasis which might trigger mitochondrial dysfunction, ROS production and cell death [117].

3.2. Titanium dioxide

Titanium dioxide (TiO2) is widely used as a white pigment in paint, ink, plastic, and paper and as food additive, while the nanosized TiO2 is also used for its photocatalytic activity in self-cleaning materials and for its UV absorption capacity in sunscreen. Moreover, TiO2 is included in the list of inactive ingredients by the FDA, considering it safe to be used in dental paste, oral capsules, suspensions, tablets, dermal preparations and non-parenteral medicines. TiO2 particles are believed to possess low toxicity and thus are widely used in biomedical applications for their excellent biocompatibility. The range of light that is scattered as well as other properties of TiO2 depend on the particle size. It naturally exists in three crystal structures: anatase (tetragonal), rutile (tetragonal), and brookite (orthorhombic). Anatase and rutile TiO2 both have a tetragonal structure, while the TiO6 octahedron of anatase TiO2 is distorted to be larger than that of the rutile phase [126]. When the size of TiO2 is diminished to nanoscale (diameter <100 nm), the bioactivity and physiochemical properties of nano-sized TiO2 are significantly different from the properties of their bulk analogue [127], [128]. Nanoparticles of TiO2 (TiO2 NPs) are allowed as sunscreen additives in concentrations of up to 25% [1]. The increased use of nanosized materials has led to an increased burden of TiO2 NPs in aquatic environments. It is, however, unclear how high levels might occur in environment and if they are harmful to organisms [129]. Analogous to ZnO NPs, the increased demand for products containing TiO2 is met by increased occupational exposure. Apart from the NIOSH 2011 current intelligence bulletin, to date, no occupational or environmental exposure limits for TiO2 NPs have been set by any other regulatory agency. The number of workers currently exposed to TiO2 dust is not available.

Often when a product is so attractive to industry, the understanding about its risk assessments is insufficient and lags behind their rapid advancement and widespread applications [130]. In the case of TiO2 NPs, it is not yet clear how they are transported into or out of the brain, how they accumulate or what kind of behavioral or cognitive dysfunction they may cause, however the evidence summarized in this (Table 4) and other review articles [130], [128], [79], [149], [150], [151], [152], [153], [154] may indicate that their toxic potential remains to be fully elucidated

Table 4.

Neurotoxic effects of TiO2 NPs.

Compound Exposure model Experimental design Effect Reference
TiO2 NPs Mice Intratracheal instillations once per week for 4 weeks
13.2 mg/kg
Inflammatory cell aggregation and neuron necrosis. Ti level in the brain 3 days after a single instillation was upregulated by 100%. [131]
Wistar rats, male Intratracheal
0.1, 1.0, 10.0 mg/kg
Ti accumulation in the brain and dose-dependent injury. TiO2 NPs with diameter of 200 nm did not cause significant alterations in the brain. [132]
BBB model based on rat primary endothelial cells (BECs) and astrocytes Acute exposure: 24 h, 0–500 μg/ml
Chronic exposure: 5 days, 0–100 μg/ml
Reduced expression of P-gp, claudin 5, caveolin-1, and caveolin-2 associated with BBB integrity. [133]
Fisher F344 rats, male Intravenous
single dose
1 mg/kg
Upregulation of tight junction proteins, modulation of P-gp mRNA expression and persistent brain inflammation markers: IL-1β, IP-10, GFAP and CXCL1. No Ti accumulation in the brain after 24 [134]
Mice Intranasal
90 days
2.5, 5.0, 10 mg/kg
Ti accumulation in the brain. Oxidative stress, high levels of lipid, protein, and DNA peroxidation, overproliferation of glial cells, tissue necrosis, hippocampal cell apoptosis. Microarray showed significant alterations of 249 genes expression. [135]
Mice, female Intranasal instillation
every other day for 2, 10, 20, 30 days
500 μg
Ti accumulation in hippocampus after 30 days of rutile exposure.
Irregular arrangement and loss of neurons, morphological changes and oxidative damage in hippocampus. Increased TNF-α and IL-1β levels.
[136]
Mice, female Intranasal instillation
every other day for 2, 10, 20, 30 days
500 μg
Imbalance of monoaminergic neurotransmitters, increased NE and 5-HT, while levels of DA, DOPAC, HVA and 5-HIAA were decreased. [137]
Wistar rats, male Intragastrical
60 days
50, 100, 200 mg/kg
Downregulated AChE activity.
Increased plasmatic and brain IL-6.
Increased GFAP expression.
[138]
Zebrafish embryos 96 hpf
0.1, 1, 10 μg/ml
Hatching time was decreased, with increase in malformation rate. Accumulation in brain with ROS and cell death in hypothalamus. Alterations in behavior and PD-related genes. [139]
Caenorhabditis elegans 24 h
7.7, 38.5 μg/ml
GC–MS-based metabolomics perturbations mainly occurred in TCA cycle, glyoxalate, tricarboxylate, inositol phosphate, Gly, Ser, Thr, Gln, and Glu metabolism. [140]
Caenorhabditis elegans 96 h under dark or light conditions
1–100 mg/l
Light exposure induced the production of ROS and increased toxicity from a median effect concentration of more than 100 mg/l to 53 mg/l. [141]
D384 glial cell line and SH-SY5Y human neuroblastoma 24 h
15, 31 μg/ml,
Concentration- and time-dependent alterations of mitochondrial function, cell membrane damage, inhibition of cell proliferation. Effects dependent on TiO2 size. Neuronal cells were more sensitive than glial cells. [142]
U373 human glial cells and C6 rat glial cells 24–96 h
2.5–40 μg/cm2
DNA fragmentation assessed in U373 cells, but not in C6 cells. Morphological changes associated with depolymerization of F-actin, apoptotic cell death. [143]
U373 human glial cells and C6 rat glial cells 2–24 h
20 μg/cm2
Increased expression of antioxidant enzymes: GPx, CAT, SOD2, lipid peroxidation and mitochondrial depolarization. [144]
PC12 rat pheochromocytoma 6–48 h
1–100 μg/ml for
Apoptosis prevented by a ROS scavenger, N-MPG. [145]
Co-culture of PC12 cells with primary rat microglia 24–48 h
0.25–0.5 mg/ml
Supernatant from TiO2 NPs treated microglia caused significant cytotoxicity in PC12 cells. [146]
PC12 cell line 24 h
1–125 μg/ml
Decreased cell viability, mitochondrial impairment and decreased DA levels. [139]
BV2 microgial cells 6, 18 h
2.5–120 ppm
Release of ROS, mitochondrial hyperpolarization [147]
BV2 microgial cells, N27 neurons, primary cultures of rat striatum 2, 6, 24, 48 h
2.5–120 ppm
Microglia generated ROS damages neurons in complex primary cultures. No cytotoxicity in isolated N27 neurons [148]

Abbreviations: 5-HIAA: 5-hydroxyindole; 5-HT: 5-hydroxytriptamine; AchE: acetylcholine estarese; BBB: blood-brain barrier; CAT: catalase; CXCL1: chemokine C-X-C motif ligand 1; DA: dopamine; DOPAC: 3,4-dihydrophenylacetic acid; GC–MS: gas chromatography mass spectrometry; GFAP: glial fibrillary acidic protein; Gly: glycine; Gln: glutamine: Glu: glutamate; GPx: glutathione peroxidase; HVA: homovanillic acid; hpf: hours post fertilization; IL-1β: interleukin-1β; IL-6: interleukin-6; IP-10: interferon gamma-induced protein 10; NE: norepinephrine; N-MPG: N-(2-mercaptopropionyl)glycine; NPs: nanoparticles; PD: Parkinson’s disease; P-gp: P-glycoprotein; ROS: reactive oxygen species; Ser: serine; SOD2: superoxide dismutase 2; TCA: tricarboxylic acid cycle; TNF-α: tumor necrosis factor α; Thr: threonine; Ti: titanium; TiO2: titanium dioxide.

3.2.1. TiO2 absorption and transport across the BBB

Dermal absorption is the most relevant entry route of chemicals related to sunscreen use. Several studies have analyzed TiO2 penetrance into intact or damaged skin using different models. On the whole, studies demonstrated that TiO2 NPs cannot permeate intact and damaged skin and can be found only in the stratum corneum and epidermis, without reaching the brain or peripheral organs [155], [156], [157], [158]. Furthermore, low cytotoxicity observed in human HaCaT keratinocytes, suggests a low toxic potential of these nano-compounds at the skin level. These results can be explained by the great stability and low ionizing capacity of these particles and are in accordance with several studies in the literature [159], [160], [161]. However, studies simulating real-world scenarios on sunburned skin, with UV exposure in long-term chronic exposure conditions need to be conducted to assure the safety of TiO2 in sunscreen.

Long-term intake of TiO2 NPs at low doses was assayed in rats. Animals received 1 or 2 mg/kg TiO2 suspension per day for 5 consecutive days. On the sixth day their gut tissue was analyzed for TiO2 content and possible adverse effects. A sex-specific effect on villus cells proliferation was observed in male rats, indicating a potential role for the endocrine system in this process. Oxidative stress in intestinal cells was transient and decreased after 24 h [162].

NPs have the ability to cross the BBB. While this may be desirable for drug-delivery systems [163], it can also pose a risk of unwanted accumulation of potentially harmful chemicals in the brain. In an in vivo study by Li et al. (2010), mice were chronically exposed to TiO2 NPs of 3 nm diameter (4 mg/kg) via intratracheal instillations. After 4 weeks, inflammatory cell aggregation and neuron necrosis were present. The amount of Ti in the brain was measured by inductively coupled plasma mass spectrometry (ICP-MS) 3 days after a single instillation of 4 mg/kg TiO2 and found to be upregulated by 100% (120 ng/g Ti in controls, compared to 240 ng/g in treated animals) [131].

3.1.2. TiO2 neurotoxic effects in vivo

The diameter of TiO2 NPs seems to be important for its carriage. Rats were treated with TiO2 NPs (0.1, 1, 10 mg/kg) suspension of different diameters (10, 20, and 200 nm) through intratracheal treatment. Seventy-two hours later, TiO2 NPs with diameters of 10 and 20 nm were both transported into the brain, inducing dose-dependent alteration in pro-inflammatory markers (TNF-α, IL-1β and IL-10). However, TiO2 NPs with diameter of 200 nm did not cause significant alterations in the brain [132]. In a BBB model based on rat primary endothelial cells (BECs) and astrocytes, TiO2 NPs (acute exposure for 24 h with 0–500 μg/ml or chronic exposure for 5 days with 0–100 μg/ml) could not only pass through the BBB but also disrupt its integrity by reducing the expression of P-glycoprotein (P-gp), claudin 5, caveolin-1, and caveolin-2, which are associated with the BBB integrity [133].

The effects of TiO2 NPs on the brain may not occur by a direct interaction between the chemical and the BBB. [134] described the in vivo uptake and clearance of TiO2 NPs by BECs and demonstrated a Ti burden in the liver, spleen and lungs up to a year after intravenous (i.v.) administration of TiO2 NPs (1 mg/kg) to rats, with a very low clearance rate. At this dose, the authors did not observe Ti accumulation in the brain, however upregulation of tight junction proteins, modulation of P-gp mRNA expression and persistent brain inflammation markers such as IL-1β, IP-10 (interferon gamma-induced protein 10), GFAP (glial fibrillary acidic protein) and CXCL1 (chemokine C-X-C motif ligand 1) were observed. The authors suggested that TiO2 NPs can exert an indirect effect on the CNS that seems dependent on circulating biomarkers potentially released by organs accumulating Ti [134].

Brain levels of 0.05–0.15 μg/ml were detected after intranasal administration of 2.5–10 mg/kg TiO2 NPs for 90 consecutive days in association with oxidative stress, high levels of lipid, protein, and DNA peroxidation, overproliferation of glial cells, tissue necrosis, hippocampal cell apoptosis in mice. Microarray showed significant alterations of 249 genes expression involved in oxidative stress, apoptosis, memory and learning, brain development, lipid metabolism, DNA repair, signal transduction, immune response and response to stimulus in the brain-injured mice. Some of these genes may be potential biomarkers of brain toxicity caused by TiO2 NPs exposure [135].

Female mice were intranasally instilled with 500 μg of two types of well-characterized TiO2 NPs (i.e. 80 nm, rutile or 155 nm, anatase) every other day for 2, 10, 20 or 30 days. High Ti accumulation (ranging from 0.13 to 0.3 μg/ml) was more pronounced in hippocampus after 30 days of rutile exposure, compared to other brain regions (cerebellum, olfactory bulb or cortex). Histological analysis revealed irregular arrangement and loss of neurons, morphological changes and oxidative damage in the hippocampus. Increased TNF-α and IL-1β levels were also observed [136]. Translocated TiO2 NPs (500 μg) caused imbalance of monoaminergic neurotransmitters, with significantly increased NE and 5-HT levels, while levels of DA, 3,4-dihydrophenylacetic acid (DOPAC), homovanillic (HVA), and 5-hydroxyindole acetic acid (5-HIAA) were decreased [137].

Acetylcholinesterase activity was evaluated in plasma and brain of rats after 60 days intragastric treatment with anatase TiO2 NPs (50, 100, 200 mg/kg). Plasmatic AChE activity was decreased with the increasing TiO2 NPs doses. The higher doses of TiO2 NPS caused a significant decrease in the AChE activity in the brain. These effects were accompanied by IL-6 increase in the brain and plasma and increased levels of GFAP in cerebral cortex, suggesting neuroinflammation [138]. Cognitive function may have also been compromised in this model, but behavioral experiments are lacking. Studies that describe the specific proteins that carry TiO2 to and/or from the brain are lacking.

In zebrafish larvae exposed to environmentally relevant concentrations (1–10 μg/ml) of TiO2 NPs induced Parkinson's disease (PD)-like symptoms, with locomotor alteration, reduced DA, Lewy bodies formation and alterations in mRNA levels of pink1, parkin and α-syn, that were significantly increased in a dose-dependent manner. The authors observed TiO2 accumulation in brain and oxidative stress, with cell death in hypothalamus. To further investigate TiO2 effects on DAergic cells, the authors exposed PC12 cells to 1–125 μg/ ml TiO2 NPs for 24 h. Cell viability was decreased at the higher dose and similarly to zebrafish, DA levels were decreased. This study suggests a role for TiO2 exposure in the development of PD [139].

Caenorhabditis elegans (C. elegans) is an excellent biological model organism for environmental risk assessment. Gas chromatography mass spectrometry (GC–MS)-based metabolomics approach was used to understand the toxicity of sub-lethal concentrations (7.7 and 38.5 μg/ml) of TiO2 NPs (<25 nm). Most of the significant perturbations occurred in organic acids (citric, lactic, fumaric, succinic and malic acids) and amino acids. Differential marker metabolites identified from the metabolomic analysis suggested that the disturbances, mainly occurred in metabolism of: glyoxalate, inositol phosphate, tricarboxylate, glycine (Gly), serine (Ser) threonine (Thr) glutamine (Gln) and Glu [140]. Toxicity of bulk-scale (∼160 nm) and nanoscale (21 nm) TiO2 was tested under dark and light conditions. Light exposure induced the production of ROS by nanoscale TiO2 and increased toxicity of the nanomaterial from a median effect concentration of more than 100 mg/l to 53 mg/l. The observation that light increased the toxicity of the highly photoactive nanoscale TiO2 suggests that ROS play a role in the photoactivated toxicity of the nanomaterial. No evidence of intracellular oxidative stress was found. Because TiO2 accumulated in worm intestines, as observed by microscopy, the authors suggested that ROS were formed extracellularly in the apical surface of the worms’ intestinal cells [141].

3.2.3. TiO2 neurotoxic effects in vitro

In vitro human cell models may represent a valid instrument to investigate TiO2 NPs effects on CNS and to determine their underlying mechanistic processes, providing information about doses of exposure. [142] demonstrated concentration- and time-dependent alterations of the mitochondrial function on D384 (glial cell line) and SH-SY5Y (neuronal cell line) cells starting at the dose of 31 and 15 μg/ml TiO2 (15–69 nm in diameter, anatase isoform), respectively, after 24 h exposure. Neuronal cells were more sensitive than glial cells. These effects were more pronounced in cells exposed to NPs compared to TiO2 bulk, where with the latter effects appeared only at the highest doses (125 and 250 μg/ml) after 24 and 48 h, similarly in both cerebral cell lines. Cell membrane damage was present in both cell lines starting at 125 μg/ml after 24 h exposure and also dependent on TiO2 size. TiO2 NPs were potent inhibitors of cell proliferation in human CNS cells after prolonged exposure (up to 10 days) at doses ranging from 0.1 to 1.5 μg/ml [142].

TiO2 NPs induced apoptosis in both human (U373) and rat (C6) glial cells at 96 h of treatment, evidenced by active caspase-3 starting at 5 μg/cm2. At this concentration, DNA fragmentation assessed with the TUNEL assay was observed in U373 cells, but not in C6 cells. Morphological changes associated with depolymerization of F-actin were found, accompanied by apoptotic cell death [143]. In a similar protocol of exposure, TiO2 NPs induced oxidative stress in U373 and C6 glial cells, mediating changes in the cellular redox state, which was correlated with increase in antioxidant enzyme expression (GPx, catalase and SOD2) and lipoperoxidation. Mitochondrial depolarization was also observed. These effects occurred within 24 h exposure to 20 μg/cm2 TiO2 NPs [144]. Oxidative stress was also present in rat PC12 cells exposed to TiO2 NPs 50 μg/ml for 24 h (P25 type, 21 nm in average size) and N-(2-mercaptopropionyl)-glycine (N-MPG), a kind of ROS scavenger, prevented apoptosis in this model [145], indicating that oxidative stress is an important factor in TiO2 NPs-induced neurotoxicity.

P25 (an uncoated photo-active, largely anatase form of nanosize TiO2, not used in sunscreen) stimulates ROS in BV2 microglia at 2.5–120 ppm, starting at 5 min exposure [147] and was later found to be nontoxic to isolated N27 neurons. However, P25 rapidly damages neurons at low concentrations (5 ppm, 6 h) in complex brain cultures of striatal cells, suggesting that microglial generated ROS damages neurons [148]. Ability of activated microglia to induce death of target cells was studied by Xue et al. (2012) in co-culture with PC12 cells. Supernatant from TiO2 NPs-treated (0.25–0.5 mg/ml) microglia caused significant cytotoxicity in PC12 cells. The authors suggested that TiO2 NPs stimulated microglia produced inflammatory factors, which caused PC12 cells cytotoxicity [146].

Recent studies report endoplasmic reticulum stress (ER stress) as a common response to NPs related toxicity. The ER stress also known as unfolded protein response (UPR) refers to an important cellular self-protection mechanism, which can be activated to counteract the cell situation of stress (overloading proteins or direct ER damage). ER stress was observed in human epidermal keratinocytes (HEKn) and human umbilical vein endothelial cells (HUVEC) exposed to up to 20 μg/cm2 for 16–24 h [164], [165]. Analogous to neural cells, oxidative stress was also observed in different cell types, demonstrating that TiO2 can affect a wide range of tissues. For example, the lung is a primary target of NPs exposure, especially in occupational settings. In the case of TiO2 inhalation, nanoscale particles may deposit in the lung interstitium and cause inflammation [166]. Several excellent reports are available on TiO2 effects on peripheral tissues, such as skin [167], kidney [168], liver [169], lung [170] and vascular endothelial cells [171].

4. Conclusions and future perspectives

Although some UV-related pathologies could be prevented by applying sunscreen, the efficiency and safety of these products is questionable. As the use of sunscreen is continuously increasing worldwide, so do the levels of environmental accumulation and human, and wildlife exposure. Whether concentration resulted from daily use and/or environmental contact possesses a realistic hazard to humans and other organisms is still unknown. Numerous studies raised concerns about the association between exposure to substances commonly found in sunscreens and endocrine and developmental impairments. In this review, the potential neurotoxicity of such substances is presented and the question of cost-benefit is raised regarding large scale use of sunscreen in its current composition. Although most studies reviewed in this paper reported adverse neurotoxic effects of UV filters at concentrations substantially higher that those observed in environment and human tissues, these studies should not be disregarded, as they afford potential pathomechanisms which might occur in other conditions or sensitive populations. It is noteworthy, that gene x environment interactions vis-à-vis toxicity of sunscreen components has yet to be studied. Unfortunately, the effects of repeated, long-term and low-dose exposures to single compounds and mixtures of various UV filters is also poorly studied. More studies are needed to evaluate the realistic hazard of contemporary sunscreens. Furthermore, it is also timely and meritorious to advance studies on alternative, safer and more efficient UV filters.

Conflicts of interest

The authors declare no conflict of interest.

Acknowledgment

This work has been supported by the National Institutes of Health [grant numbers NIEHS R01ES07331, NIEHS R01ES10563 and NIEHS R01ES020852].

Contributor Information

Joanna A. Ruszkiewicz, Email: joruszkiewicz@gmail.com, joanna.ruszkiewicz@einstein.yu.edu.

Adi Pinkas, Email: adi.pinkas@gmail.com.

Beatriz Ferrer, Email: beatriz.ferrervillahoz@einstein.yu.edu.

Tanara V. Peres, Email: Tanara.Peres-Vieira@einstein.yu.edu.

Aristides Tsatsakis, Email: aristsatsakis@gmail.com.

Michael Aschner, Email: michael.aschner@einstein.yu.edu.

References

  • 1.Krause M., Klit A., Blomberg Jensen M., Søeborg T., Frederiksen H., Schlumpf M., Lichtensteiger W., Skakkebaek N.E., Drzewiecki K.T. Sunscreens: are they beneficial for health? An overview of endocrine disrupting properties of UV-filters. Int. J. Androl. 2012;35:424–436. doi: 10.1111/j.1365-2605.2012.01280.x. [DOI] [PubMed] [Google Scholar]
  • 2.Thompson S.C., Jolley D., Marks R. Reduction of solar keratoses by regular sunscreen use. N. Engl. J. Med. 1993;329:1147–1151. doi: 10.1056/NEJM199310143291602. [DOI] [PubMed] [Google Scholar]
  • 3.Green A., Williams G., Neale R., Hart V., Leslie D., Parsons P., Marks G.C., Gaffney P., Battistutta D., Frost C., Lang C., Russell A. Daily sunscreen application and betacarotene supplementation in prevention of basal-cell and squamous-cell carcinomas of the skin: a randomised controlled trial. Lancet. 1999;354:723–729. doi: 10.1016/S0140-6736(98)12168-2. [DOI] [PubMed] [Google Scholar]
  • 4.Dupuy A., Dunant A., Grob J.J. Randomized controlled trial testing the impact of high-protection sunscreens on sun-exposure behavior. Arch. Dermatol. 2005;141:950–956. doi: 10.1001/archderm.141.8.950. [DOI] [PubMed] [Google Scholar]
  • 5.Debacq-Chainiaux F., Leduc C., Verbeke A., Toussaint O. UV, stress and aging. Dermatoendocrinology. 2012;4:236–240. doi: 10.4161/derm.23652. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Fisher G.J., Kang S., Varani J., Bata-Csorgo Z., Wan Y., Datta S., Voorhees J.J. Mechanisms of photoaging and chronological skin aging. Arch. Dermatol. 2002;138:1462–1470. doi: 10.1001/archderm.138.11.1462. [DOI] [PubMed] [Google Scholar]
  • 7.Wlaschek M., Tantcheva-Poór I., Naderi L., MAW, Schneider L.A., Razi-Wolf Z., Schüller J., Scharffetter-Kochanek K. Solar UV irradiation and dermal photoaging. J. Photochem. Photobiol. B. 2001;63:41–51. doi: 10.1016/s1011-1344(01)00201-9. [DOI] [PubMed] [Google Scholar]
  • 8.Neale R., Williams G., Green A. Application patterns among participants randomized to daily sunscreen use in a skin cancer prevention trial. Arch. Dermatol. 2002;138:1319–1325. doi: 10.1001/archderm.138.10.1319. [DOI] [PubMed] [Google Scholar]
  • 9.Wulf H.C., Stender I.M., Lock-Andersen J. Sunscreens used at the beach do not protect against erythema: a new definition of SPF is proposed. Photodermatol. Photoimmunol. Photomed. 1997;13:129–132. doi: 10.1111/j.1600-0781.1997.tb00215.x. [DOI] [PubMed] [Google Scholar]
  • 10.Fent K., Zenker A., Rapp M. Widespread occurrence of estrogenic UV-filters in aquatic ecosystems in Switzerland. Environ. Pollut. 2010;158:1817–1824. doi: 10.1016/j.envpol.2009.11.005. [DOI] [PubMed] [Google Scholar]
  • 11.Balmer M.E., Buser H.R., Müller M.D., Poiger T. Occurrence of some organic UV filters in wastewater, in surface waters, and in fish from Swiss Lakes. Environ. Sci. Technol. 2005;39:953–962. doi: 10.1021/es040055r. [DOI] [PubMed] [Google Scholar]
  • 12.Osmond M.J., Mccall M.J. Zinc oxide nanoparticles in modern sunscreens: an analysis of potential exposure and hazard. Nanotoxicology. 2010;4:15–41. doi: 10.3109/17435390903502028. [DOI] [PubMed] [Google Scholar]
  • 13.Petersen B., Wulf H.C. Application of sunscreen–theory and reality. Photodermatol. Photoimmunol. Photomed. 2014;30:96–101. doi: 10.1111/phpp.12099. [DOI] [PubMed] [Google Scholar]
  • 14.EPA . U.S. Environmental Protection Agency; Washington, DC: 2015. Dermal exposure factors. T.U.S.E.P. Agency (Ed.), Exposure Factors Handbook; p. 6. [Google Scholar]
  • 15.Gonzalez H. Percutaneous absorption with emphasis on sunscreens. Photochem. Photobiol. Sci. 2010;9:482–488. doi: 10.1039/b9pp00149b. [DOI] [PubMed] [Google Scholar]
  • 16.Calafat A.M., Wong L.Y., YE X., Reidy J.A., Needham L.L. Concentrations of the sunscreen agent benzophenone-3 in residents of the United States: national Health and Nutrition Examination Survey 2003–2004. Environ. Health Perspect. 2008;116:893–897. doi: 10.1289/ehp.11269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Bae J., Kim S., Kannan K., Buck Louis G.M. Couples' urinary concentrations of benzophenone-type ultraviolet filters and the secondary sex ratio. Sci. Total Environ. 2016;543:28–36. doi: 10.1016/j.scitotenv.2015.11.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Philippat C., Bennett D., Calafat A.M., Picciotto I.H. Exposure to select phthalates and phenols through use of personal care products among Californian adults and their children. Environ. Res. 2015;140:369–376. doi: 10.1016/j.envres.2015.04.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Janjua N.R., Mogensen B., Andersson A.M., Petersen J.H., Henriksen M., Skakkebaek N.E., Wulf H.C. Systemic absorption of the sunscreens benzophenone-3, octyl-methoxycinnamate, and 3-(4-methyl-benzylidene) camphor after whole-body topical application and reproductive hormone levels in humans. J. Invest. Dermatol. 2004;123:57–61. doi: 10.1111/j.0022-202X.2004.22725.x. [DOI] [PubMed] [Google Scholar]
  • 20.Janjua N.R., Kongshoj B., Andersson A.M., Wulf H.C. Sunscreens in human plasma and urine after repeated whole-body topical application. J. Eur. Acad. Dermatol. Venereol. 2008;22:456–461. doi: 10.1111/j.1468-3083.2007.02492.x. [DOI] [PubMed] [Google Scholar]
  • 21.Rodriguez J., Maibach H.I. Percutaneous penetration and pharmacodynamics: wash-in and wash-off of sunscreen and insect repellent. J. Dermatolog. Treat. 2016;27:11–18. doi: 10.3109/09546634.2015.1050350. [DOI] [PubMed] [Google Scholar]
  • 22.Gulson B., Mccall M.J., Bowman D.M., Pinheiro T. A review of critical factors for assessing the dermal absorption of metal oxide nanoparticles from sunscreens applied to humans, and a research strategy to address current deficiencies. Arch. Toxicol. 2015;89:1909–1930. doi: 10.1007/s00204-015-1564-z. [DOI] [PubMed] [Google Scholar]
  • 23.Schlumpf M., Kypke K., Wittassek M., Angerer J., Mascher H., Mascher D., Vökt C., Birchler M., Lichtensteiger W. Exposure patterns of UV filters, fragrances, parabens, phthalates, organochlor pesticides, PBDEs, and PCBs in human milk: correlation of UV filters with use of cosmetics. Chemosphere. 2010;81:1171–1183. doi: 10.1016/j.chemosphere.2010.09.079. [DOI] [PubMed] [Google Scholar]
  • 24.Philippat C., Mortamais M., Chevrier C., Petit C., Calafat A.M., YEX, Silva M.J., Brambilla C., Pin I., Charles M.A., Cordier S., Slama R. Exposure to phthalates and phenols during pregnancy and offspring size at birth. Environ. Health Perspect. 2012;120:464–470. doi: 10.1289/ehp.1103634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Ogura I., Sakurai H., Gamo M. Onsite aerosol measurements for various engineered nanomaterials at industrial manufacturing plants. J. Phys.: Conf. Ser. 2011;304:012004. [Google Scholar]
  • 26.Debia M., Bakhiyi B., Ostiguy C., Verbeek J.H., Brouwer D.H., Murashov V. A systematic review of reported exposure to engineered nanomaterials. Ann. Occup. Hyg. 2016;60:9160935. doi: 10.1093/annhyg/mew041. [DOI] [PubMed] [Google Scholar]
  • 27.Silvia Díaz-Cruz M., Llorca M., Barceló D., Barceló D. Organic UV filters and their photodegradates: metabolites and disinfection by-products in the aquatic environment. TrAC Trends Anal. Chem. 2008;27:873–887. [Google Scholar]
  • 28.Ramos S., Homem V., Alves A., Santos L. Advances in analytical methods and occurrence of organic UV-filters in the environment–A review. Sci. Total Environ. 2015;526:278–311. doi: 10.1016/j.scitotenv.2015.04.055. [DOI] [PubMed] [Google Scholar]
  • 29.Kim S., Choi K. Occurrences, toxicities, and ecological risks of benzophenone-3, a common component of organic sunscreen products: a mini-review. Environ. Int. 2014;70:143–157. doi: 10.1016/j.envint.2014.05.015. [DOI] [PubMed] [Google Scholar]
  • 30.Sharifan H., Klein D., Morse A.N. UV filters interaction in the chlorinated swimming pool, a new challenge for urbanization, a need for community scale investigations. Environ. Res. 2016;148:273–276. doi: 10.1016/j.envres.2016.04.002. [DOI] [PubMed] [Google Scholar]
  • 31.Sanchez-Quiles D., Tovar-Sanchez A. Are sunscreens a new environmental risk associated with coastal tourism? Environ. Int. 2015;83:158–170. doi: 10.1016/j.envint.2015.06.007. [DOI] [PubMed] [Google Scholar]
  • 32.Molins-Delgado D., Gago-Ferrero P., Diaz-Cruz M.S., Barcelo D. Single and joint ecotoxicity data estimation of organic UV filters and nanomaterials toward selected aquatic organisms. Urban groundwater risk assessment. Environ. Res. 2016;145:126–134. doi: 10.1016/j.envres.2015.11.026. [DOI] [PubMed] [Google Scholar]
  • 33.Downs C.A., Kramarsky-Winter E., Segal R., Fauth J., Knutson S., Bronstein O., Ciner F.R., Jeger R., Lichtenfeld Y., Woodley C.M., Pennington P., Cadenas K., Kushmaro A., Loya Y. Toxicopathological effects of the sunscreen UV filter oxybenzone (Benzophenone-3), on coral planulae and cultured primary cells and its environmental contamination in hawaii and the U.S. Virgin Islands. Arch. Environ. Contam. Toxicol. 2016;70:265–288. doi: 10.1007/s00244-015-0227-7. [DOI] [PubMed] [Google Scholar]
  • 34.Young A.R., Claveau J., Rossi A.B. Ultraviolet radiation and the skin: photobiology and sunscreen photoprotection. J. Am. Acad. Dermatol. 2017;76:S100–S109. doi: 10.1016/j.jaad.2016.09.038. [DOI] [PubMed] [Google Scholar]
  • 35.Kannan S., Lim H.W. Photoprotection and vitamin D: a review. Photodermatol. Photoimmunol. Photomed. 2014;30:137–145. doi: 10.1111/phpp.12096. [DOI] [PubMed] [Google Scholar]
  • 36.Wang J., Pan L., Wu S., Lu L., Xu Y., Zhu Y., Guo M., Zhuang S. Recent advances on endocrine disrupting effects of UV filters. Int. J. Environ. Res. Public Health. 2016;13(8):782. doi: 10.3390/ijerph13080782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Maipas S., Nicolopoulou-Stamati P. Sun lotion chemicals as endocrine disruptors. Hormones (Athens) 2015;14:32–46. doi: 10.1007/BF03401379. [DOI] [PubMed] [Google Scholar]
  • 38.Ponzo O.J., Silvia C. Evidence of reproductive disruption associated with neuroendocrine changes induced by UV-B filters, phthalates and nonylphenol during sexual maturation in rats of both gender. Toxicology. 2013;311:41–51. doi: 10.1016/j.tox.2013.05.014. [DOI] [PubMed] [Google Scholar]
  • 39.Nash J., Tanner P., Grosick T., Zimnawoda M. Sunscreen market analysis: the evolution and use of UVA-1 actives. J. Am. Acad. Dermatol. 2004;50:34. [Google Scholar]
  • 40.Tovar-Sánchez A., Sánchez-Quiles D., Basterretxea G., Benedé J.L., Chisvert A., Salvador A., Moreno-Garrido I., Blasco J. Sunscreen products as emerging pollutants to coastal waters. PLoS One. 2013;8:e65451. doi: 10.1371/journal.pone.0065451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Dareer E.L.S.M., Kalin J.R., Tillery K.F., Hill D.L. Disposition of 2-hydroxy-4-methoxybenzophenone in rats dosed orally, intravenously, or topically. J. Toxicol. Environ. Health. 1986;19:491–502. doi: 10.1080/15287398609530947. [DOI] [PubMed] [Google Scholar]
  • 42.Fediuk D.J., Wang T., Raizman J.E., Parkinson F.E., GU X. Tissue deposition of the insect repellent DEET and the sunscreen oxybenzone from repeated topical skin applications in rats. Int. J. Toxicol. 2010;29:594–603. doi: 10.1177/1091581810380147. [DOI] [PubMed] [Google Scholar]
  • 43.Kadry A.M., Okereke C.S., Abdel-Rahman M.S., Friedman M.A., Davis R.A. Pharmacokinetics of benzophenone-3 after oral exposure in male rats. J. Appl. Toxicol. 1995;15:97–102. doi: 10.1002/jat.2550150207. [DOI] [PubMed] [Google Scholar]
  • 44.Okereke C.S., Abdel-Rhaman M.S., Friedman M.A. Disposition of benzophenone-3 after dermal administration in male rats. Toxicol. Lett. 1994;73:113–122. doi: 10.1016/0378-4274(94)90101-5. [DOI] [PubMed] [Google Scholar]
  • 45.Kajta M., Wójtowicz A.K. Impact of endocrine-disrupting chemicals on neural development and the onset of neurological disorders. Pharmacol. Rep. 2013;65:1632–1639. doi: 10.1016/s1734-1140(13)71524-x. [DOI] [PubMed] [Google Scholar]
  • 46.Axelstad M., Boberg J., Hougaard K.S., Christiansen S., Jacobsen P.R., Mandrup K.R., Nellemann C., Lund S.P., Hass U. Effects of pre- and postnatal exposure to the UV-filter octyl methoxycinnamate (OMC) on the reproductive: auditory and neurological development of rat offspring. Toxicol. Appl. Pharmacol. 2011;250:278–290. doi: 10.1016/j.taap.2010.10.031. [DOI] [PubMed] [Google Scholar]
  • 47.Klammer H., Schlecht C., Wuttke W., Schmutzler C., Gotthardt I., Köhrle J., Jarry H. Effects of a 5-day treatment with the UV-filter octyl-methoxycinnamate (OMC) on the function of the hypothalamo-pituitary-thyroid function in rats. Toxicology. 2007;238:192–199. doi: 10.1016/j.tox.2007.06.088. [DOI] [PubMed] [Google Scholar]
  • 48.Carbone S., Szwarcfarb B., Reynoso R., Ponzo O.J., Cardoso N., Ale E., Moguilevsky J.A., Scacchi P. In vitro effect of octyl − methoxycinnamate (OMC) on the release of Gn-RH and amino acid neurotransmitters by hypothalamus of adult rats. Exp. Clin. Endocrinol. Diabetes. 2010;118:298–303. doi: 10.1055/s-0029-1224153. [DOI] [PubMed] [Google Scholar]
  • 49.Szwarcfarb B., Carbone S., Reynoso R., Bollero G., Ponzo O., Moguilevsky J., Scacchi P. Octyl-methoxycinnamate (OMC), an ultraviolet (UV) filter, alters LHRH and amino acid neurotransmitters release from hypothalamus of immature rats. Exp. Clin. Endocrinol. Diabetes. 2008;116:94–98. doi: 10.1055/s-2007-1004589. [DOI] [PubMed] [Google Scholar]
  • 50.Broniowska Ż., Pomierny B., Smaga I., Filip M., Budziszewska B. The effect of UV-filters on the viability of neuroblastoma (SH-SY5Y) cell line. Neurotoxicology. 2016;54:44–52. doi: 10.1016/j.neuro.2016.03.003. [DOI] [PubMed] [Google Scholar]
  • 51.Blüthgen N., Zucchi S., Fent K. Effects of the UV filter benzophenone-3 (oxybenzone) at low concentrations in zebrafish (Danio rerio) Toxicol. Appl. Pharmacol. 2012;263:184–194. doi: 10.1016/j.taap.2012.06.008. [DOI] [PubMed] [Google Scholar]
  • 52.Zucchi S., Blüthgen N., Ieronimo A., Fent K. The UV-absorber benzophenone-4 alters transcripts of genes involved in hormonal pathways in zebrafish (Danio rerio) eleuthero-embryos and adult males. Toxicol. Appl. Pharmacol. 2011;250:137–146. doi: 10.1016/j.taap.2010.10.001. [DOI] [PubMed] [Google Scholar]
  • 53.Maerkel K., Lichtensteiger W., Durrer S., Conscience M., Schlumpf M. Sex- and region-specific alterations of progesterone receptor mRNA levels and estrogen sensitivity in rat brain following developmental exposure to the estrogenic UV filter 4-methylbenzylidene camphor. Environ. Toxicol. Pharmacol. 2005;19:761–765. doi: 10.1016/j.etap.2004.12.055. [DOI] [PubMed] [Google Scholar]
  • 54.Maerkel K., Durrer S., Henseler M., Schlumpf M., Lichtensteiger W. Sexually dimorphic gene regulation in brain as a target for endocrine disrupters: developmental exposure of rats to 4-methylbenzylidene camphor. Toxicol. Appl. Pharmacol. 2007;218:152–165. doi: 10.1016/j.taap.2006.10.026. [DOI] [PubMed] [Google Scholar]
  • 55.Faass O., Schlumpf M., Reolon S., Henseler M., Maerkel K., Durrer S., Lichtensteiger W. Female sexual behavior: estrous cycle and gene expression in sexually dimorphic brain regions after pre- and postnatal exposure to endocrine active UV filters. Neurotoxicology. 2009;30:249–260. doi: 10.1016/j.neuro.2008.12.008. [DOI] [PubMed] [Google Scholar]
  • 56.Carou M.E., Szwarcfarb B., Deguiz M.L., Reynoso R., Carbone S., Moguilevsky J.A., Scacchi P., Ponzo O.J. Impact of 4-methylbenzylidene-camphor (4-MBC) during embryonic and fetal development in the neuroendocrine regulation of testicular axis in prepubertal and peripubertal male rats. Exp. Clin. Endocrinol. Diabetes. 2009;117:449–454. doi: 10.1055/s-0028-1112153. [DOI] [PubMed] [Google Scholar]
  • 57.Li V.W., Tsui M.P., Chen X., Hui M.N., Jin L., Lam R.H., Yu R.M., Murphy M.B., Cheng J., Lam P.K., Cheng S.H. Effects of 4-methylbenzylidene camphor (4-MBC) on neuronal and muscular development in zebrafish (Danio rerio) embryos. Environ. Sci. Pollut. Res. Int. 2016;23:8275–8285. doi: 10.1007/s11356-016-6180-9. [DOI] [PubMed] [Google Scholar]
  • 58.Blüthgen N., Meili N., Chew G., Odermatt A., Fent K. Accumulation and effects of the UV-filter octocrylene in adult and embryonic zebrafish (Danio rerio) Sci. Total Environ. 2014;476–477:207–217. doi: 10.1016/j.scitotenv.2014.01.015. [DOI] [PubMed] [Google Scholar]
  • 59.Schlumpf M., Schmid P., Durrer S., Conscience M., Maerkel K., Henseler M., Gruetter M., Herzog I., Reolon S., Ceccatelli R., Faass O., Stutz E., Jarry H., Wuttke W., Lichtensteiger W. Endocrine activity and developmental toxicity of cosmetic UV filters-an update. Toxicology. 2004;205:113–122. doi: 10.1016/j.tox.2004.06.043. [DOI] [PubMed] [Google Scholar]
  • 60.Seidlová-Wuttke D., Jarry H., Christoffel J., Rimoldi G., Wuttke W. Comparison of effects of estradiol (E2) with those of octylmethoxycinnamate (OMC) and 4-methylbenzylidene camphor (4MBC)-2 filters of UV light − on several uterine, vaginal and bone parameters. Toxicol. Appl. Pharmacol. 2006;210:246–254. doi: 10.1016/j.taap.2005.05.006. [DOI] [PubMed] [Google Scholar]
  • 61.Klammer H., Schlecht C., Wuttke W., Jarry H. Multi-organic risk assessment of estrogenic properties of octyl-methoxycinnamate in vivo: A 5-day sub-acute pharmacodynamic study with ovariectomized rats. Toxicology. 2005;215:90–96. doi: 10.1016/j.tox.2005.06.026. [DOI] [PubMed] [Google Scholar]
  • 62.Schlumpf M., Cotton B., Conscience M., Haller V., Steinmann B., Lichtensteiger W. In vitro and in vivo estrogenicity of UV screens. Environ. Health Perspect. 2001;109:239–244. doi: 10.1289/ehp.01109239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Schmutzler C., Hamann I., Hofmann P.J., Kovacs G., Stemmler L., Mentrup B., Schomburg L., Ambrugger P., Grüters A., Seidlova-Wuttke D., Jarry H., Wuttke W., Köhrle J. Endocrine active compounds affect thyrotropin and thyroid hormone levels in serum as well as endpoints of thyroid hormone action in liver heart and kidney. Toxicology. 2004;205:95–102. doi: 10.1016/j.tox.2004.06.041. [DOI] [PubMed] [Google Scholar]
  • 64.Sarveiya V., Risk S., Benson H.A. Liquid chromatographic assay for common sunscreen agents: application to in vivo assessment of skin penetration and systemic absorption in human volunteers. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 2004;803:225–231. doi: 10.1016/j.jchromb.2003.12.022. [DOI] [PubMed] [Google Scholar]
  • 65.Gonzalez H., Farbrot A., Larko O., Wennberg A.M. Percutaneous absorption of the sunscreen benzophenone-3 after repeated whole-body applications: with and without ultraviolet irradiation. Br. J. Dermatol. 2006;154:337–340. doi: 10.1111/j.1365-2133.2005.07007.x. [DOI] [PubMed] [Google Scholar]
  • 66.Frederiksen H., Nielsen O., Skakkebaek N.E., Juul A., Andersson A.M. UV filters analyzed by isotope diluted TurboFlow-LC-MS/MS in urine from Danish children and adolescents. Int. J. Hyg. Environ. Health. 2016;S1438–S4639:30112. doi: 10.1016/j.ijheh.2016.08.005. [DOI] [PubMed] [Google Scholar]
  • 67.Coronado M., DE Haro H., Deng X., Rempel M.A., Lavado R., Schlenk D. Estrogenic activity and reproductive effects of the UV-filter oxybenzone (2-hydroxy-4-methoxyphenyl-methanone) in fish. Aquat. Toxicol. 2008;90:182–187. doi: 10.1016/j.aquatox.2008.08.018. [DOI] [PubMed] [Google Scholar]
  • 68.Schreurs R., Lanser P., Seinen W., Van Der Burg B. Estrogenic activity of UV filters determined by an in vitro reporter gene assay and an in vivo transgenic zebrafish assay. Arch. Toxicol. 2002;76:257–261. doi: 10.1007/s00204-002-0348-4. [DOI] [PubMed] [Google Scholar]
  • 69.Valle-Sistac J., Molins-Delgado D., Díaz M., Ibáñez L., Barceló D., Silvia Díaz-Cruz M. Determination of parabens and benzophenone-type UV filters in human placenta. First description of the existence of benzyl paraben and benzophenone-4. Environ. Int. 2016;88:243–249. doi: 10.1016/j.envint.2015.12.034. [DOI] [PubMed] [Google Scholar]
  • 70.Jiménez-Díaz I., Molina-Molina J.M., Zafra-Gómez A., Ballesteros O., Navalón A., Real M., Sáenz J.M., Fernández M.F., Olea N. Simultaneous determination of the UV-filters benzyl salicylate, phenyl salicylate, octyl salicylate, homosalate, 3-(4-methylbenzylidene) camphor and 3-benzylidene camphor in human placental tissue by LC-MS/MS. Assessment of their in vitro endocrine activity. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 2013;936:80–87. doi: 10.1016/j.jchromb.2013.08.006. [DOI] [PubMed] [Google Scholar]
  • 71.Völkel W., Colnot T., Schauer U.M., Broschard T.H., Dekant W. Toxicokinetics and biotransformation of 3-(4-methylbenzylidene)camphor in rats after oral administration. Toxicol. Appl. Pharmacol. 2006;216:331–338. doi: 10.1016/j.taap.2006.05.012. [DOI] [PubMed] [Google Scholar]
  • 72.Søeborg T., Ganderup N.C., Kristensen J.H., Bjerregaard P., Pedersen K.L., Bollen P., Hansen S.H., Halling-Sørensen B. Distribution of the UV filter 3-benzylidene camphor in rat following topical application. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 2006;834:117–121. doi: 10.1016/j.jchromb.2006.02.026. [DOI] [PubMed] [Google Scholar]
  • 73.Schlumpf M., Durrer S., Faass O., Ehnes C., Fuetsch M., Gaille C., Henseler M., Hofkamp L., Maerkel K., Reolon S., Timms B., Tresguerres J.A., Lichtensteiger W. Developmental toxicity of UV filters and environmental exposure: a review. Int. J. Androl. 2008;31:144–151. doi: 10.1111/j.1365-2605.2007.00856.x. [DOI] [PubMed] [Google Scholar]
  • 74.de Groot A.C., Roberts D.W. Contact and photocontact allergy to octocrylene: a review. Contact Dermatitis. 2014;70:193–204. doi: 10.1111/cod.12205. [DOI] [PubMed] [Google Scholar]
  • 75.Park C.B., Jang J., Kim S., Kim Y.J. Single- and mixture toxicity of three organic UV-filters, ethylhexyl methoxycinnamate, octocrylene, and avobenzone on Daphnia magna. Ecotoxicol. Environ. Saf. 2017;137:57–63. doi: 10.1016/j.ecoenv.2016.11.017. [DOI] [PubMed] [Google Scholar]
  • 76.Gao L., Yuan T., Zhou C., Cheng P., Bai Q., Ao J., Wang W., Zhang H. Effects of four commonly used UV filters on the growth, cell viability and oxidative stress responses of the Tetrahymena thermophila. Chemosphere. 2013;93:2507–2513. doi: 10.1016/j.chemosphere.2013.09.041. [DOI] [PubMed] [Google Scholar]
  • 77.Zhang Q.Y., Ma X.Y., Wang X.C., Ngo H.H. Assessment of multiple hormone activities of a UV-filter (octocrylene) in zebrafish (Danio rerio) Chemosphere. 2016;159:433–441. doi: 10.1016/j.chemosphere.2016.06.037. [DOI] [PubMed] [Google Scholar]
  • 78.Antoniou C., Kosmadaki M.G., Stratigos A.J., Katsambas A.D. Sunscreens −what’s important to know. J. Eur. Acad. Dermatol. Venereol. 2008;22:1110–1118. doi: 10.1111/j.1468-3083.2008.02580.x. [DOI] [PubMed] [Google Scholar]
  • 79.Smijs T.G., Pavel S. Titanium dioxide and zinc oxide nanoparticles in sunscreens: focus on their safety and effectiveness. Nanotechnol. Sci. Appl. 2011;4:95–112. doi: 10.2147/NSA.S19419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Pérez S., Farré M.L., Barceló D. Analysis, behavior and ecotoxicity of carbon-based nanomaterials in the aquatic environment. TrAC Trends Anal. Chem. 2009;28:820–832. [Google Scholar]
  • 81.Migliore L., Uboldi C., DI Bucchianico S., Coppede F. Nanomaterials and neurodegeneration. Environ. Mol. Mutagen. 2015;56:149–170. doi: 10.1002/em.21931. [DOI] [PubMed] [Google Scholar]
  • 82.Liu J., Feng X., Wei L., Chen L., Song B., Shao L. The toxicology of ion-shedding zinc oxide nanoparticles. Crit. Rev. Toxicol. 2016;46:348–384. doi: 10.3109/10408444.2015.1137864. [DOI] [PubMed] [Google Scholar]
  • 83.Cross S.E., Innes B., Roberts M.S., Tsuzuki T., Robertson T.A., Mccormick P. Human skin penetration of sunscreen nanoparticles: in-vitro assessment of a novel micronized zinc oxide formulation. Skin Pharmacol. Physiol. 2007;20:148–154. doi: 10.1159/000098701. [DOI] [PubMed] [Google Scholar]
  • 84.Filipe P., Silva J.N., Silva R., Cirne de Castro J.L., Marques Gomes M., Alves L.C., Santus R., Pinheiro T. Stratum corneum is an effective barrier to TiO2 and ZnO nanoparticle percutaneous absorption. Skin Pharmacol. Physiol. 2009;22:266–275. doi: 10.1159/000235554. [DOI] [PubMed] [Google Scholar]
  • 85.Lin L.L., Grice J.E., Butler M.K., Zvyagin A.V., Becker W., Robertson T.A., Soyer H.P., Roberts M.S., Prow T.W. Time-correlated single photon counting for simultaneous monitoring of zinc oxide nanoparticles and NAD(P)H in intact and barrier-disrupted volunteer skin. Pharm. Res. 2011;28:2920–2930. doi: 10.1007/s11095-011-0515-5. [DOI] [PubMed] [Google Scholar]
  • 86.Schilling K., Bradford B., Castelli D., Dufour E., Nash J.F., Pape W., Schulte S., Tooley I., Van Den Bosch J., Schellauf F. Human safety review of “nano” titanium dioxide and zinc oxide. Photochem. Photobiol. Sci. 2010;9:495–509. doi: 10.1039/b9pp00180h. [DOI] [PubMed] [Google Scholar]
  • 87.Gulson B., Mccall M., Korsch M., Gomez L., Casey P., Oytam Y., Taylor A., Mcculloch M., Trotter J., Kinsley L., Greenoak G. Small amounts of zinc from zinc oxide particles in sunscreens applied outdoors are absorbed through human skin. Toxicol. Sci. 2010;118:140–149. doi: 10.1093/toxsci/kfq243. [DOI] [PubMed] [Google Scholar]
  • 88.Pirot F., Millet J., Kalia Y.N., Humbert P. In vitro study of percutaneous absorption, cutaneous bioavailability and bioequivalence of zinc and copper from five topical formulations. Skin Pharmacol. 1996;9:259–269. doi: 10.1159/000211423. [DOI] [PubMed] [Google Scholar]
  • 89.Raphael A.P., Sundh D., Grice J.E., Roberts M.S., Soyer H.P., Prow T.W. Zinc oxide nanoparticle removal from wounded human skin. Nanomedicine (Lond.) 2013;8:1751–1761. doi: 10.2217/nnm.12.196. [DOI] [PubMed] [Google Scholar]
  • 90.Monteiro-Riviere N.A., Wiench K., Landsiedel R., Schulte S., Inman A.O., Riviere J.E. Safety evaluation of sunscreen formulations containing titanium dioxide and zinc oxide nanoparticles in UVB sunburned skin: an in vitro and in vivo study. Toxicol. Sci. 2011;123:264–280. doi: 10.1093/toxsci/kfr148. [DOI] [PubMed] [Google Scholar]
  • 91.Kao Y.Y., Cheng T.J., Yang D.M., Wang C.T., Chiung Y.M., Liu P.S. Demonstration of an olfactory bulb-brain translocation pathway for ZnO nanoparticles in rodent cells in vitro and in vivo. J. Mol. Neurosci. 2012;48:464–471. doi: 10.1007/s12031-012-9756-y. [DOI] [PubMed] [Google Scholar]
  • 92.Beckett W.S., Chalupa D.F., Pauly-Brown A., Speers D.M., Stewart J.C., Frampton M.W., Utell M.J., Huang L.S., Cox C., Zareba W., Oberdörster G. Comparing inhaled ultrafine versus fine zinc oxide particles in healthy adults: a human inhalation study. Am. J. Respir. Crit. Care Med. 2005;171:1129–1135. doi: 10.1164/rccm.200406-837OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Oberdörster G., Elder A., Rinderknecht A. Nanoparticles and the brain: cause for concern? J. Nanosci. Nanotechnol. 2009;9:4996–5007. doi: 10.1166/jnn.2009.gr02. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Shim K.H., Jeong K.H., Bae S.O., Kang M.O., Maeng E.H., Choi C.S., Kim Y.R., Hulme J., Lee E.K., Kim M.K., An S.S.A. Assessment of ZnO and SiO2 nanoparticle permeability through and toxicity to the blood–brain barrier using evans blue and tem. Int. J. Nanomed. 2014;9:225–233. doi: 10.2147/IJN.S58205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Shrivastava R., Raza S., Yadav A., Kushwaha P., Flora S.J.S. Effects of sub acute exposure to TiO2 ZnO and Al2O3 nanoparticles on oxidative stress and histological changes in mouse liver and brain. Drug Chem. Toxicol. 2014;37:336–347. doi: 10.3109/01480545.2013.866134. [DOI] [PubMed] [Google Scholar]
  • 96.Yeh T.K., Chen J.K., Lin C.H., Yang M.H. Kinetics and tissue distribution of neutron-activated zinc oxide nanoparticles and zinc nitrate in mice: effects of size and particulate nature. Nanotechnology. 2012;23 doi: 10.1088/0957-4484/23/8/085102. 085102-085102. [DOI] [PubMed] [Google Scholar]
  • 97.Kielbik P., Kaszewski J., Rosowska J., Wolska E., Witkowski B.S., Gralak M.A., Gajewski Z., Godlewski M., Godlewski M.M. Biodegradation of the ZnO:Eu nanoparticles in the tissues of adult mouse after alimentary application. Nanomedicine. 2017;13:843–852. doi: 10.1016/j.nano.2016.11.002. [DOI] [PubMed] [Google Scholar]
  • 98.Vandebriel R.J., De Jong W.H. A review of mammalian toxicity of ZnO nanoparticles. Nanotechnol. Sci. Appl. 2012;5:61–71. doi: 10.2147/NSA.S23932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Han D., Tian Y., Zhang T., Ren G., Yang Z. Nano-zinc oxide damages spatial cognition capability via over-enhanced long-term potentiation in hippocampus of Wistar rats. Int. J. Nanomed. 2011;6:1453–1461. doi: 10.2147/IJN.S18507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Amara S., Ben-Slama I., Mrad I., Rihane N., Jeljeli M., El-Mir L., Ben-Rhouma K., Rachidi W., Sève M., Abdelmelek H., Sakly M. Acute exposure to zinc oxide nanoparticles does not affect the cognitive capacity and neurotransmitters levels in adult rats. Nanotoxicology. 2014;8:208–215. doi: 10.3109/17435390.2013.879342. [DOI] [PubMed] [Google Scholar]
  • 101.Amara S., Slama I.B., Omri K., Ghoul J.E.L., Mir L.E.L., Rhouma K.B., Abdelmelek H., Sakly M. Effects of nanoparticle zinc oxide on emotional behavior and trace elements homeostasis in rat brain. Toxicol. Ind. Health. 2015;31:1202–1209. doi: 10.1177/0748233713491802. [DOI] [PubMed] [Google Scholar]
  • 102.Ansar S., Abudawood M., Hamed S.S., Aleem M.M. Exposure to zinc oxide nanoparticles induces neurotoxicity and proinflammatory response: amelioration by hesperidin. Biol. Trace Elem. Res. 2017;175:360–366. doi: 10.1007/s12011-016-0770-8. [DOI] [PubMed] [Google Scholar]
  • 103.Cho W.-S., Kang B.-C., Lee J.K., Jeong J., Che J.-H., Seok S.H. Comparative absorption, distribution, and excretion of titanium dioxide and zinc oxide nanoparticles after repeated oral administration. Part. Fibre Toxicol. 2013;10:9. doi: 10.1186/1743-8977-10-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Tian L., Lin B., Wu L., Li K., Liu H., Yan J., Liu X., Xi Z. Neurotoxicity induced by zinc oxide nanoparticles: age-related differences and interaction. Sci. Rep. 2015;5:16117. doi: 10.1038/srep16117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Xie Y., Wang Y., Zhang T., Ren G., Yang Z. Effects of nanoparticle zinc oxide on spatial cognition and synaptic plasticity in mice with depressive-like behaviors. J. Biomed. Sci. 2012;19:14. doi: 10.1186/1423-0127-19-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Okada Y., Tachibana K., Yanagita S., Takeda K. Prenatal exposure to zinc oxide particles alters monoaminergic neurotransmitter levels in the brain of mouse offspring. J. Toxicol. Sci. 2013;38:363–370. doi: 10.2131/jts.38.363. [DOI] [PubMed] [Google Scholar]
  • 107.Hao L., Chen L. Oxidative stress responses in different organs of carp (Cyprinus carpio) with exposure to ZnO nanoparticles. Ecotoxicol. Environ. Saf. 2012;80:103–110. doi: 10.1016/j.ecoenv.2012.02.017. [DOI] [PubMed] [Google Scholar]
  • 108.Miranda R.R., Damaso Da Silveira A.L.R., De Jesus I.P., Grötzner S.R., Voigt C.L., Campos S.X., Garcia J.R.E., Randi M.A.F., Oliviera Ribeiro C.A., Filipak Neto F. Effects of realistic concentrations of TiO2 and ZnO nanoparticles in Prochilodus lineatus juvenile fish. Environ. Sci. Pollut. Res. 2016;23:5179–5188. doi: 10.1007/s11356-015-5732-8. [DOI] [PubMed] [Google Scholar]
  • 109.Milivojević T., Glavan G., BOŽIČ J., Sepčić K., Mesarič T., Drobne D. Neurotoxic potential of ingested ZnO nanomaterials on bees. Chemosphere. 2015;120:547–554. doi: 10.1016/j.chemosphere.2014.07.054. [DOI] [PubMed] [Google Scholar]
  • 110.Zhao J., Xu L., Zhang T., Ren G., Yang Z. Influences of nanoparticle zinc oxide on acutely isolated rat hippocampal CA3 pyramidal neurons. Neurotoxicology. 2009;30:220–230. doi: 10.1016/j.neuro.2008.12.005. [DOI] [PubMed] [Google Scholar]
  • 111.Chiang H.M., Xia Q., Zou X., Wang C., Wang S., Miller B.J., Howard P.C., Yin J.J., Beland F.A., YU H., FU P.P. Nanoscale ZnO induces cytotoxicity and DNA damage in human cell lines and rat primary neuronal cells. J. Nanosci. Nanotechnol. 2012;12:2126–2135. doi: 10.1166/jnn.2012.5758. [DOI] [PubMed] [Google Scholar]
  • 112.Deng X., Luan Q., Chen W., Wang Y., WU M., Zhang H., Jiao Z. Nanosized zinc oxide particles induce neural stem cell apoptosis. Nanotechnology. 2009;20:115101. doi: 10.1088/0957-4484/20/11/115101. [DOI] [PubMed] [Google Scholar]
  • 113.Yin Y., Lin Q., Sun H., Chen D., Wu Q., Chen X., Li S. Cytotoxic effects of ZnO hierarchical architectures on RSC96 Schwann cells. Nanoscale Res. Lett. 2012;7:439. doi: 10.1186/1556-276X-7-439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Osmond-Mcleod M.J., Osmond R.I.W., Oytam Y., Mccall M.J., Feltis B., Mackay-Sim A., Wood S.A., Cook A.L. Surface coatings of ZnO nanoparticles mitigate differentially a host of transcriptional, protein and signalling responses in primary human olfactory cells. Part. Fibre Toxicol. 2013;10:54. doi: 10.1186/1743-8977-10-54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Guo D., BI H., WU Q., Wang D., Cui Y. Zinc oxide nanoparticles induce rat retinal ganglion cell damage through bcl-2, caspase-9 and caspase-12 pathways. J. Nanosci. Nanotechnol. 2013;13:3769–3777. doi: 10.1166/jnn.2013.7169. [DOI] [PubMed] [Google Scholar]
  • 116.Guo D., BI H., Liu B., WU Q., Wang D., Cui Y. Reactive oxygen species-induced cytotoxic effects of zinc oxide nanoparticles in rat retinal ganglion cells. Toxicol. In Vitro. 2013;27:731–738. doi: 10.1016/j.tiv.2012.12.001. [DOI] [PubMed] [Google Scholar]
  • 117.Guo D., BI H., Wang D., WU Q. Zinc oxide nanoparticles decrease the expression and activity of plasma membrane calcium ATPase, disrupt the intracellular calcium homeostasis in rat retinal ganglion cells. Int. J. Biochem. Cell Biol. 2013;45:1849–1859. doi: 10.1016/j.biocel.2013.06.002. [DOI] [PubMed] [Google Scholar]
  • 118.Kim J.-H., Jeong M.S., Kim D.-Y., Her S., Wie M.-B. Zinc oxide nanoparticles induce lipoxygenase-mediated apoptosis and necrosis in human neuroblastoma SH-SY5Y cells. Neurochem. Int. 2015;90:204–214. doi: 10.1016/j.neuint.2015.09.002. [DOI] [PubMed] [Google Scholar]
  • 119.Valdiglesias V., Costa C., Kiliç G., Costa S., Pásaro E., Laffon B., Teixeira J.P. Neuronal cytotoxicity and genotoxicity induced by zinc oxide nanoparticles. Environ. Int. 2013;55:92–100. doi: 10.1016/j.envint.2013.02.013. [DOI] [PubMed] [Google Scholar]
  • 120.Wahab R., Kaushik N.K., Verma A.K., Mishra A., Hwang I.H., Yang Y.B., Shin H.S., Kim Y.S. Fabrication and growth mechanism of ZnO nanostructures and their cytotoxic effect on human brain tumor U87, cervical cancer HeLa, and normal HEK cells. J. Biol. Inorg. Chem. 2011;16:431–442. doi: 10.1007/s00775-010-0740-0. [DOI] [PubMed] [Google Scholar]
  • 121.Wang J., Deng X., Zhang F., Chen D., Ding W. ZnO nanoparticle-induced oxidative stress triggers apoptosis by activating JNK signaling pathway in cultured primary astrocytes. Nanoscale Res. Lett. 2014;9:117. doi: 10.1186/1556-276X-9-117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Sruthi S., Mohanan P.V. Investigation on cellular interactions of astrocytes with zinc oxide nanoparticles using rat C6 cell lines. Colloids Surf. B Biointerfaces. 2015;133:1–11. doi: 10.1016/j.colsurfb.2015.05.041. [DOI] [PubMed] [Google Scholar]
  • 123.Ostrovsky S., Kazimirsky G., Gedanken A., Brodie C. Selective cytotoxic effect of ZnO nanoparticles on glioma cells. Nano Res. 2009;2:882–890. [Google Scholar]
  • 124.Sharma A.K., Singh V., Gera R., Purohit M.P., Ghosh D. Zinc oxide nanoparticle induces microglial death by NADPH-oxidase-independent reactive oxygen species as well as energy depletion. Mol. Neurobiol. 2016:1–14. doi: 10.1007/s12035-016-0133-7. [DOI] [PubMed] [Google Scholar]
  • 125.Wei L., Wang J., Chen A., Liu J., Feng X., Shao L. Involvement of PINK1/parkin-mediated mitophagy in ZnO nanoparticle-induced toxicity in BV-2 cells. Int. J. Nanomed. 2017;12:1891–1903. doi: 10.2147/IJN.S129375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Diebold U. The surface science of titanium dioxide. Surf. Sci. Rep. 2003;48:53–229. [Google Scholar]
  • 127.Chen X., Mao S.S. Titanium dioxide nanomaterials: synthesis, properties, modifications, and applications. Chem. Rev. 2007;107:2891–2959. doi: 10.1021/cr0500535. [DOI] [PubMed] [Google Scholar]
  • 128.Skocaj M., Filipic M., Petkovic J., Novak S. Titanium dioxide in our everyday life; is it safe? Radiol. Oncol. 2011;45:227–247. doi: 10.2478/v10019-011-0037-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Ates M., Demir V., Adiguzel R., Arslan Z. Bioaccumulation, subacute toxicity, and tissue distribution of engineered titanium dioxide nanoparticles in goldfish (Carassius auratus) J. Nanomater. 2013;2013 doi: 10.1155/2013/460518. pii: 460518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Newman M.D., Stotland M., Ellis J.I. The safety of nanosized particles in titanium dioxide- and zinc oxide-based sunscreens. J. Am. Acad. Dermatol. 2009;61:685–692. doi: 10.1016/j.jaad.2009.02.051. [DOI] [PubMed] [Google Scholar]
  • 131.Li Y., Li J., Yin J., Li W., Kang C., Huang Q., Li Q. Systematic influence induced by 3 nm titanium dioxide following intratracheal instillation of mice. J. Nanosci. Nanotechnol. 2010;10:8544–8549. doi: 10.1166/jnn.2010.2690. [DOI] [PubMed] [Google Scholar]
  • 132.Liu Y., Xu Z., Li X. Cytotoxicity of titanium dioxide nanoparticles in rat neuroglia cells. Brain Inj. 2013;27:934–939. doi: 10.3109/02699052.2013.793401. [DOI] [PubMed] [Google Scholar]
  • 133.Brun E., Carriere M., Mabondzo A. In vitro evidence of dysregulation of blood-brain barrier function after acute and repeated/long-term exposure to TiO(2) nanoparticles. Biomaterials. 2012;33:886–896. doi: 10.1016/j.biomaterials.2011.10.025. [DOI] [PubMed] [Google Scholar]
  • 134.Disdier C., Devoy J., Cosnefroy A., Chalansonnet M., Herlin-Boime N., Brun E., Lund A., Mabondzo A. Tissue biodistribution of intravenously administrated titanium dioxide nanoparticles revealed blood-brain barrier clearance and brain inflammation in rat. Part. Fibre Toxicol. 2015;12:27. doi: 10.1186/s12989-015-0102-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Ze Y., Hu R., Wang X., Sang X., Ze X., Li B., Su J., Wang Y., Guan N., Zhao X., Gui S., Zhu L., Cheng Z., Cheng J., Sheng L., Sun Q., Wang L., Hong F. Neurotoxicity and gene-expressed profile in brain-injured mice caused by exposure to titanium dioxide nanoparticles. J. Biomed. Mater. Res. A. 2014;102:470–478. doi: 10.1002/jbm.a.34705. [DOI] [PubMed] [Google Scholar]
  • 136.Wang J., Liu Y., Jiao F., Lao F., LI W., Gu Y., Li Y., Ge C., Zhou G., Li B., Zhao Y., Chai Z., Chen C. Time-dependent translocation and potential impairment on central nervous system by intranasally instilled TiO(2) nanoparticles. Toxicology. 2008;254:82–90. doi: 10.1016/j.tox.2008.09.014. [DOI] [PubMed] [Google Scholar]
  • 137.Wang J.X., Li Y.F., Zhou G.Q., Li B., Jiao F., Chen C.Y., Gao Y.X., Zhao Y.L., Chai Z.F. Influence of intranasal instilled titanium dioxide nanoparticles on monoaminergic neurotransmitters of female mice at different exposure time. Zhonghua Yu Fang Yi Xue Za Zhi. 2007;41:91–95. [PubMed] [Google Scholar]
  • 138.Grissa I., Guezguez S., Ezzi L., Chakroun S., Sallem A., Kerkeni E., Elghoul J., Mir E.L.L., Mehdi M., Cheikh H.B., Haouas Z. The effect of titanium dioxide nanoparticles on neuroinflammation response in rat brain. Environ. Sci. Pollut. Res. Int. 2016;23:20205–20213. doi: 10.1007/s11356-016-7234-8. [DOI] [PubMed] [Google Scholar]
  • 139.Hu Q., Guo F., Zhao F., Fu Z. Effects of titanium dioxide nanoparticles exposure on parkinsonism in zebrafish larvae and PC12. Chemosphere. 2017;173:373–379. doi: 10.1016/j.chemosphere.2017.01.063. [DOI] [PubMed] [Google Scholar]
  • 140.Ratnasekhar C., Sonane M., Satish A., Mudiam M.K. Metabolomics reveals the perturbations in the metabolome of Caenorhabditis elegans exposed to titanium dioxide nanoparticles. Nanotoxicology. 2015;9:994–1004. doi: 10.3109/17435390.2014.993345. [DOI] [PubMed] [Google Scholar]
  • 141.Angelstorf J.S., Ahlf W., Von der kammer F., Heise S. Impact of particle size and light exposure on the effects of TiO2 nanoparticles on Caenorhabditis elegans. Environ. Toxicol. Chem. 2014;33:2288–2296. doi: 10.1002/etc.2674. [DOI] [PubMed] [Google Scholar]
  • 142.Coccini T., Grandi S., Lonati D., Locatelli C., DE Simone U. Comparative cellular toxicity of titanium dioxide nanoparticles on human astrocyte and neuronal cells after acute and prolonged exposure. Neurotoxicology. 2015;48:77–89. doi: 10.1016/j.neuro.2015.03.006. [DOI] [PubMed] [Google Scholar]
  • 143.Marquez-Ramirez S.G., Delgado-Buenrostro N.L., Chirino Y.I., Iglesias G.G., Lopez-Marure R. Titanium dioxide nanoparticles inhibit proliferation and induce morphological changes and apoptosis in glial cells. Toxicology. 2012;302:146–156. doi: 10.1016/j.tox.2012.09.005. [DOI] [PubMed] [Google Scholar]
  • 144.Huerta-Garcia E., Perez-Arizti J.A., Marquez-Ramirez S.G., Delgado-Buenrostro N.L., Chirino Y.I., Iglesias G.G., Lopez-Marure R. Titanium dioxide nanoparticles induce strong oxidative stress and mitochondrial damage in glial cells. Free Radic. Biol. Med. 2014;73:84–94. doi: 10.1016/j.freeradbiomed.2014.04.026. [DOI] [PubMed] [Google Scholar]
  • 145.Liu S., XU L., Zhang T., Ren G., Yang Z. Oxidative stress and apoptosis induced by nanosized titanium dioxide in PC12 cells. Toxicology. 2010;267:172–177. doi: 10.1016/j.tox.2009.11.012. [DOI] [PubMed] [Google Scholar]
  • 146.Xue Y., Wu J., Sun J. Four types of inorganic nanoparticles stimulate the inflammatory reaction in brain microglia and damage neurons in vitro. Toxicol. Lett. 2012;214:91–98. doi: 10.1016/j.toxlet.2012.08.009. [DOI] [PubMed] [Google Scholar]
  • 147.Long T.C., Saleh N., Tilton R.D., Lowry G.V., Veronesi B. Titanium dioxide (P25) produces reactive oxygen species in immortalized brain microglia (BV2): implications for nanoparticle neurotoxicity. Environ. Sci. Technol. 2006;40:4346–4352. doi: 10.1021/es060589n. [DOI] [PubMed] [Google Scholar]
  • 148.Long T.C., Tajuba J., Sama P., Saleh N., Swartz C., Parker J., Hester S., Lowry G.V., Veronesi B. Nanosize titanium dioxide stimulates reactive oxygen species in brain microglia and damages neurons in vitro. Environ. Health Perspect. 2007;115:1631–1637. doi: 10.1289/ehp.10216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Czajka M., Sawicki K., Sikorska K., Popek S., Kruszewski M., Kapka-Skrzypczak L. Toxicity of titanium dioxide nanoparticles in central nervous system. Toxicol. In Vitro. 2015;29:1042–1052. doi: 10.1016/j.tiv.2015.04.004. [DOI] [PubMed] [Google Scholar]
  • 150.Rollerova E., Tulinska J., Liskova A., Kuricova M., Kovriznych J., Mlynarcikova A., Kiss A., Scsukova S. Titanium dioxide nanoparticles: some aspects of toxicity/focus on the development. Endocr. Regul. 2015;49:97–112. doi: 10.4149/endo_2015_02_97. [DOI] [PubMed] [Google Scholar]
  • 151.Song B., Liu J., Feng X., Wei L., Shao L. A review on potential neurotoxicity of titanium dioxide nanoparticles. Nanoscale Res. Lett. 2015;10:1042. doi: 10.1186/s11671-015-1042-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Song B., Zhang Y., Liu J., Feng X., Zhou T., Shao L. Unraveling the neurotoxicity of titanium dioxide nanoparticles: focusing on molecular mechanisms. Beilstein J. Nanotechnol. 2016;7:645–654. doi: 10.3762/bjnano.7.57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Grande F., Tucci P. Titanium dioxide nanoparticles: a risk for human health? Mini Rev. Med. Chem. 2016;16:762–769. doi: 10.2174/1389557516666160321114341. [DOI] [PubMed] [Google Scholar]
  • 154.Zhang X., Li W., Yang Z. Toxicology of nanosized titanium dioxide: an update. Arch. Toxicol. 2015;89:2207–2217. doi: 10.1007/s00204-015-1594-6. [DOI] [PubMed] [Google Scholar]
  • 155.Kertész Z., Szikszai Z., Gontier E., Moretto P., Surlève-Bazeille J., Kiss B., Juhász I., Hunyadi J., Kiss Á. Nuclear microprobe study of TiO2-penetration in the epidermis of human skin xenografts. Nucl. Instrum. Methods Phy.s Res. B. 2005;231:280–285. [Google Scholar]
  • 156.Gamer A.O., Leibold E., Van Ravenzwaay B. The in vitro absorption of microfine zinc oxide and titanium dioxide through porcine skin. Toxicol. In Vitro. 2006;20:301–307. doi: 10.1016/j.tiv.2005.08.008. [DOI] [PubMed] [Google Scholar]
  • 157.Senzui M., Tamura T., Miura K., Ikarashi Y., Watanabe Y., Fujii M. Study on penetration of titanium dioxide (TiO(2)) nanoparticles into intact and damaged skin in vitro. J. Toxicol. Sci. 2010;35:107–113. doi: 10.2131/jts.35.107. [DOI] [PubMed] [Google Scholar]
  • 158.Miquel-Jeanjean C., Crepel F., Raufast V., Payre B., Datas L., Bessou-Touya S., Duplan H. Penetration study of formulated nanosized titanium dioxide in models of damaged and sun-irradiated skins. Photochem. Photobiol. 2012;88:1513–1521. doi: 10.1111/j.1751-1097.2012.01181.x. [DOI] [PubMed] [Google Scholar]
  • 159.Jonaitis T.S., Card J.W., Magnuson B. Concerns regarding nano-sized titanium dioxide dermal penetration and toxicity study. Toxicol. Lett. 2010;192:268–269. doi: 10.1016/j.toxlet.2009.10.007. [DOI] [PubMed] [Google Scholar]
  • 160.Sadrieh N., Wokovich A.M., Gopee N.V., Zheng J., Haines D., Parmiter D., Siitonen P.H., Cozart C.R., Patri A.K., Mcneil S.E., Howard P.C., Doub W.H., Buhse L.F. Lack of significant dermal penetration of titanium dioxide from sunscreen formulations containing nano- and submicron-size TiO2 particles. Toxicol. Sci. 2010;115:156–166. doi: 10.1093/toxsci/kfq041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Crosera M., Prodi A., Mauro M., Pelin M., Florio C., Bellomo F., Adami G., Apostoli P., DE Palma G., Bovenzi M., Campanini M., Filon F.L. Titanium dioxide nanoparticle penetration into the skin and effects on HaCaT cells. Int. J. Environ. Res. Public Health. 2015;12:9282–9297. doi: 10.3390/ijerph120809282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Ammendolia M.G., Iosi F., Maranghi F., Tassinari R., Cubadda F., Aureli F., Raggi A., Superti F., Mantovani A., DE Berardis B. Short-term oral exposure to low doses of nano-sized TiO2 and potential modulatory effects on intestinal cells. Food Chem. Toxicol. 2017;102:63–75. doi: 10.1016/j.fct.2017.01.031. [DOI] [PubMed] [Google Scholar]
  • 163.Dominguez A., Suarez-Merino B., Goni-De-Cerio F. Nanoparticles and blood-brain barrier: the key to central nervous system diseases. J. Nanosci. Nanotechnol. 2014;14:766–779. doi: 10.1166/jnn.2014.9119. [DOI] [PubMed] [Google Scholar]
  • 164.Yu K.N., Chang S.H., Park S.J., Lim J., Lee J., Yoon T.J., Kim J.S., Cho M.H. Titanium dioxide nanoparticles induce endoplasmic reticulum stress-mediated autophagic cell death via mitochondria-associated endoplasmic reticulum membrane disruption in normal lung cells. PLoS One. 2015;10:e0131208. doi: 10.1371/journal.pone.0131208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Simon M., Saez G., Muggiolu G., Lavenas M., LE Trequesser Q., Michelet C., Deves G., Barberet P., Chevet E., Dupuy D., Delville M.H., Seznec H. In situ quantification of diverse titanium dioxide nanoparticles unveils selective endoplasmic reticulum stress-dependent toxicity. Nanotoxicology. 2017;11:134–145. doi: 10.1080/17435390.2017.1278803. [DOI] [PubMed] [Google Scholar]
  • 166.Wang J., Fan Y. Lung injury induced by TiO2 nanoparticles depends on their structural features: size, shape, crystal phases, and surface coating. Int. J. Mol. Sci. 2014;15:22258–22278. doi: 10.3390/ijms151222258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Kim M.S., Stees M., Karuturi B.V.K., Vijayaraghavalu S., Peterson R.E., Madsen G.L., Labhasetwar V. Pro-NP protect against TiO2 nanoparticle-induced phototoxicity in zebrafish model: exploring potential application for skin care. Drug Deliv. Transl. Res. 2017;7:372–382. doi: 10.1007/s13346-017-0374-7. [DOI] [PubMed] [Google Scholar]
  • 168.Fartkhooni F.M., Noori A., Mohammadi A. Effects of titanium dioxide nanoparticles toxicity on the kidney of male rats. Int. J. Life Sci. 2016;10:65–69. [Google Scholar]
  • 169.Alarifi S., Ali D., Al-Doaiss A.A., Ali B.A., Ahmed M., Al-Khedhairy A.A. Histologic and apoptotic changes induced by titanium dioxide nanoparticles in the livers of rats. Int. J. Nanomed. 2013;8:3937–3943. doi: 10.2147/IJN.S47174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Yoshiura Y., Izumi H., Oyabu T., Hashiba M., Kambara T., Mizuguchi Y., Lee B.W., Okada T., Tomonaga T., Myojo T., Yamamoto K., Kitajima S., Horie M., Kuroda E., Morimoto Y. Pulmonary toxicity of well-dispersed titanium dioxide nanoparticles following intratracheal instillation. J. Nanopart. Res. 2015;17:241. doi: 10.1007/s11051-015-3054-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Han S.G., Newsome B., Hennig B. Titanium dioxide nanoparticles increase inflammatory responses in vascular endothelial cells. Toxicology. 2013;306:1–8. doi: 10.1016/j.tox.2013.01.014. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Toxicology Reports are provided here courtesy of Elsevier

RESOURCES