376
Views
5
CrossRef citations to date
0
Altmetric
REVIEW

Neurotoxicity of Titanium Dioxide Nanoparticles: A Comprehensive Review

, , , , , , , , & ORCID Icon show all
Pages 7183-7204 | Received 02 Oct 2023, Accepted 25 Nov 2023, Published online: 04 Dec 2023

Abstract

The increasing use of titanium dioxide nanoparticles (TiO2 NPs) across various fields has led to a growing concern regarding their environmental contamination and inevitable human exposure. Consequently, significant research efforts have been directed toward understanding the effects of TiO2 NPs on both humans and the environment. Notably, TiO2 NPs exposure has been associated with multiple impairments of the nervous system. This review aims to provide an overview of the documented neurotoxic effects of TiO2 NPs in different species and in vitro models. Following exposure, TiO2 NPs can reach the brain, although the specific mechanism and quantity of particles that cross the blood-brain barrier (BBB) remain unclear. Exposure to TiO2 NPs has been shown to induce oxidative stress, promote neuroinflammation, disrupt brain biochemistry, and ultimately impair neuronal function and structure. Subsequent neuronal damage may contribute to various behavioral disorders and play a significant role in the onset and progression of neurodevelopmental or neurodegenerative diseases. Moreover, the neurotoxic potential of TiO2 NPs can be influenced by various factors, including exposure characteristics and the physicochemical properties of the TiO2 NPs. However, a systematic comparison of the neurotoxic effects of TiO2 NPs with different characteristics under various exposure conditions is still lacking. Additionally, our understanding of the underlying neurotoxic mechanisms exerted by TiO2 NPs remains incomplete and fragmented. Given these knowledge gaps, it is imperative to further investigate the neurotoxic hazards and risks associated with exposure to TiO2 NPs.

Graphical Abstract

Introduction

Nanomaterials (NMs) are materials defined as having at least one dimension ranging from 1 to 100 nanometers (nm). Titanium dioxide nanoparticles (TiO2 NPs) rank among the top five NMs used in consumer products, such as food additives, toys, cosmetics, electronic products, and pharmaceuticals ().Citation1, Consequently, the presence of TiO2 NPs in air, water, soil, and other environmental media has gradually increased due to their widespread use.Citation3 This growing application and contamination have made human and animal exposure to TiO2 NPs unavoidable. Apart from skin exposure, inhalation, and oral exposure, other routes of exposure to TiO2 NPs include intraperitoneal injection, subcutaneous injection, and intramuscular injection.Citation4 Importantly, regardless of the route of exposure, TiO2 NPs can ultimately enter the systemic circulation and translocate to various tissues and organs ().Citation4 As the accumulation of TiO2 NPs in the body increases, the associated health hazards become more severe.Citation5

Figure 1 Application of TiO2 NPs.

Figure 1 Application of TiO2 NPs.

Figure 2 TiO2 NPs can enter the human body through different ways and accumulate in the body, posing a threat to human health.

Figure 2 TiO2 NPs can enter the human body through different ways and accumulate in the body, posing a threat to human health.

Before the rise of nanotechnology, TiO2 was widely used in the form of fine particles (FPs), which were considered as poor soluble and low toxicity particles.Citation6 However, some studies have contradicted this view, such as lung tumors in rats exposed to high levels of TiO2 FPs for two years.Citation7 Furthermore, TiO2 has been classified as a Group 2B carcinogen (possibly carcinogenic to humans) by the International Agency for Research on Cancer.Citation8 Although the specific carcinogenicity of TiO2 FPs is still debated, there is no doubt that TiO2 FPs pose a health risk.Citation9 Compared to TiO2 FPs, TiO2 NPs present stronger catalytic activity and bioactivity due to their nanoscale dimensions.Citation9 Consequently, the toxicity of TiO2 NPs cannot be solely inferred from the known toxicology of TiO2 FPs, nor can it be determined using conventional methods.Citation10 In recent years, extensive exploration of the effects of TiO2 NPs exposure on human health also reflects the high concern about the safety of TiO2 NPs. In addition to considering all relevant exposure scenarios and biological intermediate steps, understanding the final toxic outcome is critical for human health risk assessment.Citation11 Results from another child cohort study in China indicated that Ti can cross the placental barrier (PB) to harm fetuses that are extremely sensitive to environmental threats.Citation12 Several epidemiological studies have confirmed that Ti exposure increases the risk of adverse birth outcomes, including neural tube defects, preterm birth, fetal distress, and low birth weight.Citation13–15 Moreover, TiO2 NP exposure can have detrimental effects on the health of the population beyond the fetus. Emerging epidemiological evidence suggests that higher levels of urinary or blood Ti are associated with an increased risk of various adverse health effects, including diabetes and cardiopulmonary disorders ().Citation16–19 In recent years, laboratory studies on the toxicity of TiO2 NPs have surpassed epidemiological studies. Common animal models such as mice, rats, zebrafish, and Drosophila have been used to study TiO2 NPs. In vivo, studies have shown that TiO2 NPs exposure may be linked to lung inflammation, pneumoconiosis, cardiovascular disease, reproductive toxicity, retinal impairments, etc.Citation20–24 In vitro, studies have also supported these toxic effects of TiO2 NPs.Citation25–28 Given that nanoparticles can enter the brain, concerns regarding their neurotoxic effects, including those of TiO2 NPs, have gained significant attention.Citation29

The entry of TiO2 NPs into the brain mainly occurs through the blood-brain barrier (BBB), via absorption-mediated transversion or intranasal pathways.Citation30,Citation31 However, the mechanisms by which nano-titanium dioxide penetrates and targets different brain regions remain unknown. The degree of TiO2 NPs accumulation in each brain region closely correlates with the extent of neurotoxic effects. Common neurotoxic effects include behavior deficits, nervous system dysfunction, and structural changes induced by oxidative stress, autophagy, inflammation, or the activation of specific signaling pathways.Citation32 Although emerging studies support the role of TiO2 NPs exposure as an environmental risk factor for human health, conscientious and systematic investigations are scarce into the extent of TiO2 NPs translocation to different brain regions and the resulting damage to the neuronal system in relation to particle dose and particle size. The lack of information on the neurotoxicity of TiO2 NPs also complicates risk assessment following exposure. Therefore, this paper will mainly focus on current studies concerning the neurotoxicology of TiO2 NPs, while also reviewing the molecular mechanisms underlying their neurotoxic effects to mitigate potential damage resulting from exposure.

Evidence from Epidemiological and Human Exposure Studies

In earlier years, population exposure to TiO2 NPs was primarily investigated among occupational populations. Welding fumes, industrial waste combustion, and mineral mining can all result in environmental contamination by TiO2 NPs, thereby increasing the exposure risk for workers.Citation33 Exposure to fumes from metal-inert gas soldering has been found to increase the risk of Parkinson’s disease (PD).Citation34 Although these fumes mainly consist of zinc, copper, and iron, Andujar et al discovered an excessive accumulation of Ti in the lung tissue sections of welders in 2014.Citation35 Industrial waste, pesticides, and automobile exhaust are common sources of environmental pollutants associated with neurotoxic effects.Citation36 Among various environmental pollutants, NPs can easily penetrate the BBB and induce neurotoxicity by activating innate immune responses in astrocytes, microglia, and neurons.Citation36 TiO2 NPs are a major component among environmental pollutants, with up to 760 tons of TiO2 NPs being released into the soil through sewage and sludge each year.Citation37,Citation38 Currently, there is no direct evidence of neurotoxic effects caused by TiO2 NPs exposure in mineral miners, but a previous study suggested a significantly increased inflammatory response in mineral miners exposed to TiO2 NPs.Citation39 It is well known that the occurrence of inflammatory reactions in other organs is closely related to nervous system damage.Citation40 With the increasing application of TiO2 NPs, concerns have also arisen regarding the neurotoxic effects of non-occupational populations exposed to TiO2 NPs. A recent cohort study demonstrated that high levels of urinary Ti during pregnancy were significantly associated with impaired language development, suggesting that TiO2 NPs might act as developmental neurotoxicants.Citation41 Furthermore, elevated levels of Ti in maternal hair were also significantly associated with an increased risk of neural tube defects.Citation42 However, epidemiological studies on the neurotoxic effects caused by TiO2 NPs are still limited. Currently, laboratory studies are the main basis for evaluating the neurotoxicity of TiO2 NPs.

Literature Search

To review the neurotoxic potential of TiO2 NPs, a comprehensive literature search was conducted using the “Pubmed” database, covering articles from 1991 to September 29, 2023. The search utilized combinations of the following keywords: Titanium dioxide nanoparticles exposure; E171 exposure; Titanium dioxide nanoparticles neuron; Titanium dioxide nanoparticles brain; Titanium dioxide nanoparticles behavior; and Titanium dioxide nanoparticles neurotoxicology. Our search strategy involved an initial screening of all titles and abstracts, followed by a full-text review of the pertinent review articles. One hundred forty-seven papers were selected, including one hundred twenty-one research papers. Citations within twenty-six reviews were also screened for additional studies not identified in the electronic search; however, no additional research papers were found through these references. In the end, a total of one hundred and twenty-one research papers were selected, all of which included neurotoxicity endpoints in their experimental designs.

For experimental research, we identified in vitro or in vivo studies involving the administration of TiO2 NPs. The researched organism groups mainly consisted of rodents, zebrafish, and cells. Fifty-five studies were conducted on rodents, twenty on zebrafish, and two on Caenorhabditis elegans (C. elegans), while one each on Pheretima hawayana, Tegillarca granosa, and Drosophila melanogaster. Additionally, forty-one studies used in vitro cells, including various animals and human-derived neuronal cells ().

Figure 3 Study selection flow diagram. The flow chart illustrates the number of citations and resources that underwent screening, exclusion, and/or inclusion in the review.

Figure 3 Study selection flow diagram. The flow chart illustrates the number of citations and resources that underwent screening, exclusion, and/or inclusion in the review.

Neurotoxic Effects of TiO2 NPs in Rodents

The results of the literature search demonstrate that rodents were the most commonly used species in neurotoxicity studies of TiO2 NPs, forming the basis of 55 articles. Among these, 23 articles focused on rat models, with the Wistar rat being the most commonly employed. The exposed TiO2 NPs generally had a particle size of less than 20 nm, and the exposure doses ranged below 200 mg/kg (). Intratracheal instillation, intragastric administration, intravenous injection, and intraperitoneal injection are common methods of rat exposure to TiO2 NPs. Pregnancy and lactation have been identified as critical periods of neurodevelopment and have been selected as exposure windows to toxicants in many neurotoxicological studies, including the neurotoxicity of TiO2 NPs.Citation43–45 Exposure to TiO2 NPs (10 nm, 100 mg/kg) during pregnancy or lactation has been found to impair memory and learning in Wistar offspring rats by reducing hippocampal cell proliferation.Citation44,Citation46 Prenatal exposure to TiO2 NPs (5 nm, 1 μg/μL) via subcutaneous injection has been shown to enhance depressive-like behavior in adult Sprague-Dawley (SD) rats.Citation47 Additionally, Engler-Chiurazzi et al conducted locomotion, learning, and anxiety tests on male adult SD rats and observed significant cognitive impairments in offspring resulting from inhalation exposure to TiO2 NPs (170.9 ± 6.4 nm, 10.4 ± 0.4 mg/m3) during pregnancy.Citation48 In earlier studies, the roles of age and sex in early-life exposure at different developmental stages were often overlooked. However, a recent study demonstrated that female pups were more susceptible to adverse outcomes after early exposure to oral TiO2 NPs (postnatal day, PND 2–5 or PND 7–10) compared to male pups, while male pups exhibited more severe motor deficits following exposure to nano-titanium dioxide during late lactation (PND 17–20).Citation45 The behavioral impairments resulting from TiO2 NPs exposure during pregnancy and lactation involve multiple mechanisms, including oxidative damage, changes in neurotransmitter concentrations, and metabolic disturbances.Citation45,Citation47 Furthermore, exposure to TiO2 NPs during pregnancy has been associated with neuronal apoptosis, decreased neurogenesis, altered expression of brain-derived neurotrophic factor (BDNF), and impaired synaptic plasticity.Citation49–51

Table 1 Overview of Literature Investigating Neurotoxic Effects of TiO2 NPs in Rats

Adolescence is a transitional period of physical and behavioral development between childhood and adulthood, and it represents a particularly vulnerable neurodevelopmental phase.Citation67,Citation68 In a study by Cui et al exposure to TiO2 NPs (5 nm, 20 mg/kg) during adolescence via intravenous injection induced anxiety-like behavior, cognitive impairment, neuroinflammation, and oxidative damage in the hippocampus.Citation63 Similar results were observed in another study using adult Wistar rats, where subacute exposure to 20 mg/kg TiO2 NPs increased the anxious index.Citation61 Furthermore, motor functional damage and spatial cognitive impairments were observed in adult Wistar rats exposed to TiO2 NPs.Citation54,Citation57,Citation60 Certain regions of the mammalian brain continue to exhibit neurogenesis throughout adulthood, such as the subventricular zone of the lateral ventricles and the subgranular zone of the hippocampal dentate gyrus.Citation69 Adult neurogenesis is a multiple-step process, and abnormalities at any stage can impair neurogenesis and brain function, leading to cognitive impairment and neurodegenerative diseases.Citation70 Exposure to TiO2 NPs in adulthood also induces various neurotoxic effects, including mitochondrial dysfunction, oxidative stress, cell apoptosis, alterations in neuronal architecture, neuroinflammation, decreased neurogenesis, and electrophysiological alterations.Citation52–54,Citation56,Citation58,Citation59,Citation64

To date, slightly more neurotoxicology studies on TiO2 NPs have been conducted using mice as animal models compared to rats, totaling 32 studies. Among them, 21 studies used ICR mice, 7 studies used C57BL/6, 2 studies used Albino mice, while BALB/c mice and Swiss Webster mice were used in 1 study each (). The exposure methods used in most of the neurotoxicological studies of TiO2 NPs using mice as animal models mimic the actual pathways of human exposure to TiO2 NPs. TiO2 NPs can enter the human body via intentional or unintentional inhalation; therefore, intranasal administration was chosen as the exposure method in 10 studies. Intranasal drug administration is a promising method to bypass BBB, resulting in higher bioavailability and greater brain exposure compared to oral administration at the same dose.Citation71 Several studies have shown that intranasal administration of TiO2 NPs in mice resulted in various neurological damages, including hippocampal cell apoptosis, tissue necrosis, oxidative damage, neuroinflammation, imbalance of glutamate metabolism, and altered gene expression.Citation72–79 These structural and functional impairments in the nervous system are closely related to behavioral deficits. Ze et al found that exposure to TiO2 NPs (208–330 nm, 2.5 mg/kg body weight, BW) for 90 consecutive days via intranasal administration damaged the hippocampus structure, decreased long-term potentiation, altered gene expression, and caused spatial recognition deficits.Citation80 Intragastric administration, a method commonly used to assess the effects of oral exposure in the hazard assessment of environmental toxicants, was employed in 13 neurotoxicological studies of TiO2 NPs using mice as animal models.Citation81 Pregnancy exposure to TiO2 NPs (21 nm, 150 mg/kg) via intragastric administration caused the delayed appearance of neurobehavioral impairments in both dams and offspring, which may be related to disruption of the gut-brain axis.Citation82,Citation83 Furthermore, exposure to low levels of TiO2 NPs (6.5 nm, < 5 mg/kg) via intragastric administration during pregnancy and lactation retarded axonal and dendritic outgrowth, impaired cognitive ability, and increased hippocampal neurons apoptosis.Citation84–86 In addition to common impairments in the nervous system associated with TiO2 NPs exposure, such as neuroinflammation, oxidative stress, and altered gene expression, Wang et al found that exposure to TiO2 NPs (10 nm) increased the risk of PD.Citation87

Table 2 Overview of the Literature Investigating Neurotoxic Effects of TiO2 NPs in Mice

Neurotoxic Effects of TiO2 NPs in Zebrafish

Zebrafish (Danio rerio) is commonly used as in vivo model system for studying the toxicity of nanomaterials due to its low cost, rapid growth, and significant homology to humans.Citation105 A total of 20 studies have investigated the neurotoxic effects of TiO2 NPs in zebrafish, with 11 of them examining co-exposure to other compounds (). Among the 11 studies, 8 studies selected the embryonic stage of zebrafish for TiO2 NPs exposure, 2 studies selected adult zebrafish, and one study selected zebrafish larvae. The most commonly used dose of TiO2 NPs in studies involving co-exposure to other compounds was 100 μg/L. So far, TiO2 NPs have been shown to enhance Pb,Citation106,Citation107 decabromodiphenyl oxide (BDE-209),Citation108 cypermethrin,Citation109 triphenyl phosphate,Citation110 bisphenol A,Citation111,Citation112 difenoconazole,Citation113 tetracycline,Citation114 and microcystin-LRCitation115 -induced neurotoxicity. TiO2 NPs mainly enhance the neurotoxicity of these compounds by increasing their bioconcentration and bioavailability in zebrafish. Interestingly, co-exposure with TiO2 NPs did not alter pentachlorophenol-induced neurotoxicity.Citation116

Table 3 Overview of Literature Investigating Neurotoxic Effects of TiO2 NPs in Zebrafish

Exposure to TiO2 NPs alone is also able to induce a variety of neurotoxic effects in zebrafish. The embryonic stage of zebrafish is the most commonly used exposure stage for TiO2 NPs exposure models, which may be attributed to the incomplete development of the BBB during this period.Citation126 TiO2 NPs exposure during the embryonic stage of zebrafish significantly alters motor behavior, social behavior, and spatial recognition memory.Citation117–120 In addition to behavioral impairments, TiO2 NPs exposure causes oxidative stress, promotes neuronal proliferation, decreases motor neuron axon length, alters gene expression, and increases cell apoptosis.Citation120–123 Two study chose the adult stage of zebrafish for TiO2 NPs exposure, and their results suggested that TiO2 NPs exposure caused cognitive deficit, promoted neuroinflammation, and altered biochemical constituents of the brain.Citation124,Citation125

Neurotoxic Effects of TiO2 NPs in Other Animal Models

In vivo, studies investigating the potential neurotoxic effects of TiO2 NPs exposure on animals other than rodents and zebrafish are relatively scarce. To date, only five studies investigated the neurotoxicity of TiO2 NPs exposure in other animal models, namely C. elegans, Tegillarca granosa, Pheretima hawayana, and Drosophila melanogaster ().

Table 4 Overview of the Literature Investigating Neurotoxic Effects of TiO2 NPs in Other Animal Models

The nervous system of the C. elegans model is structurally and functionally similar to that of mammals. Its small size, short life cycle, and high reproductive rate make C. elegans an advantageous model in neuroscience.Citation132 Long-term exposure to TiO2 NPs (10 nm) resulted in severe defects in the development of AVL and DVB neurons that control defecation in nematodes.Citation127 Neurons exposed to both anatase and rutile TiO2 NPs exhibited shorter axon growth, which may be the reason for defective locomotion behavior in nematodes.Citation128

Drosophila melanogaster is an excellent animal model for evaluating the neurotoxicity of various NPs due to its low-cost, physiological similarities to humans, and well-known behavioral and developmental characteristics.Citation133,Citation134 The results of a recent study indicated that chronic exposure to TiO2 NPs (approximately 20 nm, 20 mg/kg) induced deficits in motor behavior by disrupting the development of the neuromuscular junction (NMJ) in Drosophila.Citation129

Tegillarca granosa and Pheretima hawayana are rarely studied in neurotoxicological research involving NPs. Tegillarca granosa inhabits intertidal mudflat, where the concentration of NPs is predicted to be higher than in other parts of the ocean.Citation135 Selecting Tegillarca granosa as an animal model provides better insights into the neurotoxicity of NPs in marine bivalve mollusks.Citation130 Exposure to TiO2 NPs (200 nm) increased neurotransmitters concentrations, suppressed the activity of acetylcholinesterase (AChE), and decreased the expression of neurotransmitter-related genes, which may disrupt various physiological processes in Tegillarca granosa.Citation130 While TiO2 NPs are increasingly being released into the soil, their effects on soil biota remain largely unknown.Citation136 The traditional sentinel species for soil toxicity testing is the earthworm, and Pheretima hawayana is a species of Egyptian earthworm. Exposure to TiO2 NPs altered biochemical indices related to the function of the nervous system, such as inhibiting AChE, increasing antioxidant enzymes, and accumulating malondialdehyde (MDA).Citation131

Neurotoxic Effects of TiO2 NPs in vitro Models

In vitro models are widely used to assess neurotoxic effects on cellular functions.Citation137 Several studies have evaluated the neurotoxic effects of TiO2 NPs using in vitro models. Primary hippocampal and cortical neurons are widely used in vitro models for neurotoxicology testing as they are easily polarized and form unique axons and dendrites. In addition, these models are used to study neuronal polarization, axon/dendrite morphology, synaptic formation, and central nervous system (CNS) functions.Citation138 Exposure to TiO2 NPs impairs neuronal function, inhibits neuroblast proliferation, reduces cell viability, and increases cell apoptosis by promoting oxidative stress in both primary hippocampal and cortical neurons.Citation139–144 Furthermore, TiO2 NPs inhibit neurite outgrowth of hippocampal neurons by interfering with glutamate metabolism and impairing N-methyl-D-aspartic acid (NMDA) receptor function.Citation145 According to some previous studies, the suppression of axonal development, dendritic development, and synapse development by TiO2 NPs was associated with decreased expression of axon growth-related factors and inhibition of the Wnt/β-catenin and BDNF-TrkB pathways.Citation146–148 See for details.

Table 5 Overview of the Literature on Neurotoxic Effects of TiO2 NPs on Primary Neuron and Nerve Cell Lines

Rat pheochromocytoma (PC12) cell line and human SH-SY5Y neuroblastoma cell line have been used as models for neurotoxicity testing of TiO2 NPs (). PC12 cell line shows morphological and functional differentiation similar to sympathetic neurons. PC12 cell line is a suitable model for studying the chemical disruption of neuronal differentiation, synthesis, storage, and release of neurotransmitters, function and regulation of ion channels, and the interaction of compounds with membrane-bound receptors.Citation160 A previous study revealed that treatment of PC12 cells with TiO2 NPs (< 36 nm, < 200 μg/mL) decreased cell viability, increased cell apoptosis via oxidative stress, inhibited the neurite outgrowth, disturbed cell cycle, and disrupted the ubiquitin-proteasome system.Citation149–151 The human-derived SH-SY5Y cell line is preferred over the PC12 cell line as it avoids interspecific differences in chemical action.Citation161 The SH-SY5Y cell line is an excellent model for studying toxicity on proliferating or differentiated cells because it can be maintained as neuroblasts or induced to differentiate into more neuron-like morphologies.Citation161 TiO2 NPs were shown to cause endoplasmic reticulum (ER) stress, autophagy, inhibition of cell proliferation, disturbance of the microtubule dynamics, and membrane damage in SH-SY5Y cells.Citation153–158 Several in vivo studies investigated the neurotoxic effects of TiO2 NPs on mouse hippocampus. However, one in vitro study explored the neurotoxic effects of TiO2 NPs on mouse hippocampal neuronal HT22 cells. The study revealed that TiO2 NPs increased apoptosis of HT22 cells via oxidative stress- and calcium imbalance-mediated ER stress.Citation159

Acute or prolonged exposure to TiO2 NPs is associated with toxic effects on neuronal and glial cells.Citation162 Glial cells are critical cells of the nervous system, which serve as tissue-resident macrophages. Microglia are crucial regulators that influence nervous system development, maintenance of the neural environment, and response to injury and repair.Citation163 The immortalized mouse microglia cell line BV2 is often used as an alternative for primary microglia in cell experiments. Some previous studies showed that exposure of BV2 cells to TiO2 NPs was associated with mitochondrial dysfunction and increased oxidative stress.Citation28,Citation164 Astrocytes play a key role in innate and adaptive immune responses in CNS injury.Citation165 Due to advancements in cell culture technology, primary astrocytes have become a common primary cell model. Previous studies revealed that TiO2 NPs induced mitochondria damage, oxidative stress, autophagy, neuroinflammation, and cell apoptosis in primary rat cortical astrocytes.Citation166–168 Other studies employed human glial cell lines as in vitro models for neurotoxicity studies to eliminate species differences. Some previous studies revealed that TiO2 NPs inhibited cell proliferation, induced morphological changes, decreased immuno-location of F-actin fibers, and increased cell apoptosis in U374 cells.Citation169,Citation170 Furthermore, several studies have investigated the neurotoxic effects of TiO2 NPs in a co-culture of glial cells and other cells. For example, Yang et al showed that TiO2 NPs stimulate the inflammatory reaction in brain microglia and damage neuron using a co-culture model of primary microglia and PC12 cell line.Citation171 Similarly, TiO2 NPs was shown to stimulate the inflammatory reaction in brain microglia and damage neurons in co-culture models of BV2 and N27 mesencephalic neurons, and BV2 and N2a neuroblastoma cells.Citation172,Citation173 See for details.

Table 6 Overview of the Literature on Neurotoxic Effects of TiO2 NPs in Primary Glial Cells and Glial Cell Lines

Most in vivo and in vitro studies have evaluated the neurotoxic effects of TiO2 NPs in the cortex, hippocampus, and cerebellum. However, to the best of our knowledge, no studies have evaluated the neurotoxic effects of TiO2 NPs on other brain regions. The BBB is effective in protecting the brain from chemical damage. Therefore, there is a need to understand the effects of TiO2 NPS on the BBB. A previous study exploring the effects of TiO2 NPs on an in vitro model of BBB established by co-culturing primary human brain microvascular endothelial cells (HBMECs) and primary human astrocytes, revealed that TiO2 NPs increased the permeability of the BBB.Citation31 Another study showed that acute or long-term exposure of an in vitro model of the BBB established by co-culturing primary rat endothelial cells and glial cells to TiO2 NPs was associated with BBB dysfunction related to increased inflammatory response and altered expression of the ABC transporter.Citation174 Moreover, treatment of T98G human glioblastoma cells with TiO2 NPs was associated with changes in the transcriptome, suggesting that exposure to TiO2 NPs could compromise BBB integrity and cause neuroinflammation.Citation175 Furthermore, TiO2 NPs can be internalized by dorsal root ganglion cells (DRG) and cause damage via apoptosis.Citation176,Citation177 Yu et al showed an association between the toxic effects of TiO2 NPs on olfactory bulb neuron cells and its pathogenicity to neurodegenerative diseases.Citation178 Furthermore, exposure to TiO2 NPs was associated with varying degrees of cytotoxicity to the human cerebral endothelial cell line (HCECs), human neural stem cell line (hNSCs), and neuroectodermal stem cell line (1C11) models.Citation179–181 See for details.

Table 7 Overview of the Literature on Neurotoxic Effects of TiO2 NPs in Other Cells

Factors Influencing the Neurotoxic Potential of TiO2 NPs

The neurotoxic effects of TiO2 NPs are influenced by various factors. The exposure characteristics, such as exposure dose, method, duration, and species, can influence the toxic effects of TiO2 NPs in vivo. A review of the literature showed that the exposure dose in vivo and in vitro experiments was larger than the actual exposure dose of the population. According to a previous study, the levels of TiO2 NPs in air and water ranged from 0.7 to 16 μg/L.Citation182 It is estimated that children have an intake of TiO2 NPs of about 2–3 mg/kg/day, while adults have a TiO2 NPs intake of about 1 mg/kg/day.Citation2 Human exposure to TiO2 NPs is mainly through dietary intake and air inhalation. Although the exposure methods selected in animal studies attempted to mimic human exposure closely, there are some gaps. For example, the system for intranasal administration is simple compared to inhalation administration. Furthermore, intranasal administration is significantly affected by the inhalational dose.Citation183 The intranasal administration volumes in rodents at a given time should be limited to approximately 5 μL per nostril since volumes greater than this are likely to become wasted.Citation184,Citation185 Furthermore, ingested TiO2 NPs first interacts with the oral mucosa. However, intragastric administration does not interact with the oral mucosa and is thus associated with significant differences in absorption, bioavailability, and metabolism with implications for assumptions and models of toxicity kinetics.Citation81 In addition, the exposure period and duration also influence the neurotoxic effects of TiO2 NPs.Citation45,Citation94 However, the exposure duration in experiments tends to be shorter than that in humans. Species differences are often unavoidable. Therefore, there is a need to conduct epidemiological studies exploring the neurotoxic effects of TiO2 NPs on humans.

Furthermore, the physical and chemical properties of TiO2 NPs can affect their neurotoxicity. Particle size is key. In general, small particles are more likely to be absorbed and thus exert toxic effects.Citation186 According to some previous studies, the neurotoxic effects of TiO2 NPs depend on particle size.Citation60,Citation168 The hydrodynamic diameter or secondary particle sizes of TiO2 NPs are important with respect to neurotoxicity. While smaller NPs may seem more neurotoxic, they are also more likely to clump together and form aggregates.Citation186 Theoretically, the particle aggregation would increase the effective particle size thus reducing the neurotoxic potential. Several studies have used dynamic light scattering (DLS) to determine the effects of hydrodynamics or secondary particle size of TiO2 NPs on neurotoxicity. However, no studies have explored the effect of aggregate particle size on the neurotoxicity of TiO2 NPs. The zeta potential of TiO2 NPs has also been investigated in most neurotoxicological studies. Since most cell membranes are negatively charged, the zeta potential affects the tendency of NPs to penetrate the membrane, with cationic particles generally exhibiting higher toxicity associated with cell wall damage.Citation187 Furthermore, the surface charge of the nanoparticles can determine the degree of aggregation.Citation122,Citation162,Citation164 However, further studies are needed to investigate whether the zeta potential affects the neurotoxicity of TiO2 NPs. In addition, the toxicity of TiO2 NPs is dependent on crystalline phases. The anatase form of TiO2 NPs is more neurotoxic than that of rutile TiO2 NPs and P25 TiO2 NPs since anatase has a higher ability to induce oxidative stress.Citation152,Citation166,Citation188

Taken together, various factors can affect the neurotoxic potential of TiO2 NPs, including physical and chemical properties of TiO2 NPs, and exposure dose, exposure duration, exposed species. However, the specific effects of these factors on the neurotoxic effects of TiO2 NPs still need to be systematically compared.

Reflections on Neurotoxicity Induced by TiO2 NPs

Most studies to date have focused on rodents, and most experimental exposures used are not very realistic for human exposure. In addition, there is currently limited information on the levels of TiO2 NPs in the environment, consumer goods, and food products. For humans, more accurate monitoring is needed to determine daily exposure levels, particle characteristics and exposure route, all of which affect the neurotoxic potential of TiO2 NPs. Evaluating and availing data on TiO2 NPs levels in different environmental media helps to reliably estimate human exposure and thus assess the risk of TiO2 NPs. Furthermore, the degree of uptake through the digestive system, respiratory system, potential BBB crossing, and potential translocation to or even accumulation in nervous system should be further investigated. This information will indicate which route of exposure mitigation is most valuable for human health protection. However, apart from the recommended exposure limits (REL) established by the National Institute for Occupational Safety and Health (NIOSH), no other regulatory agencies have set occupational or environmental exposure limits for TiO2 NPs.Citation9 There are limitations in the monitoring methods of TiO2 NPs. There is an urgent need to develop appropriate methods for reducing TiO2 NPs in environmental media and food to prevent their potentially harmful health effects.Citation189

The specific mechanisms behind the neurotoxic effects of TiO2 NPs have only been explored through animal and cell experiments. TiO2 NPs increase the formation of reactive oxygen species (ROS) in the brain, thus inducing oxidative stress. Ze et al reported that TiO2 NPs induced oxidative stress thus causing brain damage through overactivation of the p38-Nrf-2 signaling pathway.Citation78 Oxidative stress can induce neuroinflammation, thus further aggravating cell damage.Citation63,Citation73,Citation92 Cell damage, including structural and functional damage, is associated with increased onset and development of neurodevelopmental or neurodegenerative diseases, such as autism spectrum disorder (ASD) and PD.Citation87,Citation100 Cell damage is also linked to behavioral deficits.Citation46,Citation57 Abnormal motor ability could be caused by a decrease in the axon length of motor neurons.Citation120 In addition, changes in hippocampal synaptic plasticity could lead to decreased spatial recognition.Citation80 The development of axons, dendrites and synapses is regulated by various signaling pathways. TiO2 NPs impair the growth of axons and dendrites through excessive activation of the ERK1/2/MAPK signaling pathway.Citation84 In addition, impairment of dendritic growth by TiO2 NPs is also related to inhibition of the Wnt/β-catenin signaling pathway.Citation147 Moreover, suppression of the neuronal synaptic outgrowth by TiO2 NPs is linked to the inhibition of the BDNF-TrkB signaling pathway.Citation148 Furthermore, the accumulation of TiO2 NPs in the brain could cause alterations in brain biochemistry and changes in neurotransmitter levels, contributing to behavioral changes.Citation45,Citation57,Citation117 Although all of these studies confirm that TiO2 NPs cause neurotoxic effects through different mechanisms, most of the evidence on the neurotoxic effects of TiO2 NPs is fragmentary and is obtained from different species. Furthermore, few of these mechanism studies have explored whether the neurotoxic effects of TiO2 NPs are mediated through synergistic interactions of multiple brain regions, organs, and systems. Whether TiO2 NPs with different characteristics cause different degrees of toxic effects through different mechanisms remains be further explored. Extensive systematic studies are needed to fully elucidate the neurotoxic mechanisms of TiO2 NPs, which will be helpful for the prevention and treatment of neurotoxic effects of TiO2 NPs.

Conclusion

Animals and humans can be exposed to TiO2 NPs through different exposure pathways, thus posing health hazards. At present, the neurotoxic effects of TiO2 NPs have only been evaluated through animal models, including rats, mice, and zebrafish, and cell studies, including primary neurons, PC12, and SH-SY5Y cell lines. TiO2 NPs can induce oxidative stress, promote neuroinflammation, alter brain biochemistry, or damage neurons. Neuronal damage can further lead to various behavioral disorders and is closely associated with increased onset and development of neurodevelopmental or neurodegenerative diseases. However, due to the lack of relevant epidemiological studies, whether TiO2 NPs are linked to neurodevelopmental or neurodegenerative diseases in humans remains unknown. Furthermore, the neurotoxic potential of TiO2 NPs can be affected by various factors. There is a need for researchers to understand the neurotoxic effects of TiO2 NPs on humans and develop strategies for mitigating the effects of TiO2 NPs on human health.

Abbreviations

NMs, nanomaterials; TiO2 NPs, titanium dioxide nanoparticles; FPs, fine particles; PB, placental barrier; BBB, blood-brain barrier; C. elegans, Caenorhabditis elegans; SD rats, Sprague-Dawley rats; PND, postnatal day; BDNF, brain-derived neurotrophic factor; BW, body weight; BDE-209, decabromodiphenyl oxide; NMJ, neuromuscular junction; AchE, acetylcholinesterase; MDA, malondialdehyde; CNS, central nervous system; NMDA, N-methyl-D-aspartic acid; ER, endoplasmic reticulum; HBMECs, human brain microvascular endothelial cells; DRG, dorsal root ganglion; HCECs, human cerebral endothelial cell line; HNSCs, human neural stem cell line; 1C11, neuroectodermal stem cell line; DLS, Dynamic light scattering; REL, recommended exposure limit; NIOSH, National Institute for Occupational Safety and Health; ROS, reactive oxygen species; ASD, autism spectrum disorder; PD, Parkinson’s disease.

Disclosure

The authors declare that they have no known competing interests.

Acknowledgment

The authors would like to thank all the reviewers who participated in the review, as well as MJEditor (www.mjeditor.com) for providing English editing services during the preparation of this manuscript.

Additional information

Funding

This work was supported by the Research Foundation for Talented Scholars, Nanjing Medical University (NMUR20210002).

References

  • Grande F, Tucci P. Titanium dioxide nanoparticles: a risk for human health? Mini Rev Med Chem. 2016;16(9):762–769. doi:10.2174/1389557516666160321114341
  • Weir A, Westerhoff P, Fabricius L, et al. Titanium dioxide nanoparticles in food and personal care products. Environ Sci Technol. 2012;46(4):2242–2250. doi:10.1021/es204168d
  • Mueller NC, Nowack B. Exposure modeling of engineered nanoparticles in the environment. Environ Sci Technol. 2008;42(12):4447–4453. doi:10.1021/es7029637
  • Shakeel M, Jabeen F, Shabbir S, et al. Toxicity of Nano-Titanium Dioxide (TiO2-NP) through various routes of exposure: a review. Biol Trace Elem Res. 2016;172(1):1–36. doi:10.1007/s12011-015-0550-x
  • Ali SA, Rizk MZ, Hamed MA, et al. Assessment of titanium dioxide nanoparticles toxicity via oral exposure in mice: effect of dose and particle size. Biomarkers. 2019;24(5):492–498. doi:10.1080/1354750X.2019.1620336
  • Institute IRS. The relevance of the rat lung response to particle overload for human risk assessment: a workshop consensus report. Inhal Toxicol. 2000;12(1–2):1–17. doi:10.1080/08958370050029725
  • Lee KP, Trochimowicz HJ, Reinhardt CF. Pulmonary response of rats exposed to titanium dioxide (TiO2) by inhalation for two years. Toxicol Appl Pharmacol. 1985;79(2):179–192. doi:10.1016/0041-008x(85)90339-4
  • Racovita AD. Titanium dioxide: structure, impact, and toxicity. Int J Environ Res Public Health. 2022;19(9):5681. doi:10.3390/ijerph19095681
  • Shi H, Magaye R, Castranova V, et al. Titanium dioxide nanoparticles: a review of current toxicological data. Part Fibre Toxicol. 2013;10:15. doi:10.1186/1743-8977-10-15
  • Song B, Liu J, Feng X, et al. A review on potential neurotoxicity of titanium dioxide nanoparticles. Nanoscale Res Lett. 2015;10(1):1042. doi:10.1186/s11671-015-1042-9
  • Boyes WK, van Thriel C. Neurotoxicology of nanomaterials. Chem Res Toxicol. 2020;33(5):1121–1144. doi:10.1021/acs.chemrestox.0c00050
  • Li A, Zhuang T, Shi J, et al. Heavy metals in maternal and cord blood in Beijing and their efficiency of placental transfer. J Environ Sci (China). 2019;80:99–106. doi:10.1016/j.jes.2018.11.004
  • Jin Y, Li Z, An H, et al. Environmental titanium exposure and reproductive health: risk of low birth weight associated with maternal titanium exposure from a nested case-control study in northern China. Ecotoxicol Environ Saf. 2021;208:111632. doi:10.1016/j.ecoenv.2020.111632
  • Zheng G, Zhong H, Guo Z, et al. Levels of heavy metals and trace elements in umbilical cord blood and the risk of adverse pregnancy outcomes: a population-based study. Biol Trace Elem Res. 2014;160(3):437–444. doi:10.1007/s12011-014-0057-x
  • Kinghorn KJ, Grönke S, Castillo-Quan JI, et al. A Drosophila model of neuronopathic gaucher disease demonstrates lysosomal-autophagic defects and altered mTOR signalling and is functionally rescued by rapamycin. J Neurosci. 2016;36(46):11654–11670. doi:10.1523/JNEUROSCI.4527-15.2016
  • Feng W, Cui X, Liu B, et al. Association of urinary metal profiles with altered glucose levels and diabetes risk: a population-based study in China. PLoS One. 2015;10(4):e0123742. doi:10.1371/journal.pone.0123742
  • Xiao Y, Yuan Y, Liu Y, et al. Circulating multiple metals and incident stroke in Chinese adults. Stroke. 2019;50(7):1661–1668. doi:10.1161/STROKEAHA.119.025060
  • Yang L, Jiang Y, Zhao L, et al. Multiple metals exposure and arterial stiffness: a panel study in China. Chemosphere. 2021;263:128217. doi:10.1016/j.chemosphere.2020.128217
  • Yuan Y, Xiao Y, Feng W, et al. Plasma metal concentrations and incident coronary heart disease in Chinese adults: the Dongfeng-Tongji cohort. Environ Health Perspect. 2017;125(10):107007. doi:10.1289/EHP1521
  • Yamano S, Goto Y, Takeda T, et al. Pulmonary dust foci as rat pneumoconiosis lesion induced by titanium dioxide nanoparticles in 13-week inhalation study. Part Fibre Toxicol. 2022;19(1):58. doi:10.1186/s12989-022-00498-3
  • Sagawa T, Honda A, Ishikawa R, et al. Role of necroptosis of alveolar macrophages in acute lung inflammation of mice exposed to titanium dioxide nanoparticles. Nanotoxicology. 2021;15(10):1312–1330. doi:10.1080/17435390.2021.2022231
  • Wu Y, Chen L, Chen F, et al. A key moment for TiO(2): prenatal exposure to TiO(2) nanoparticles may inhibit the development of offspring. Ecotoxicol Environ Saf. 2020;202:110911. doi:10.1016/j.ecoenv.2020.110911
  • Chen Z, Wang Y, Zhuo L, et al. Effect of titanium dioxide nanoparticles on the cardiovascular system after oral administration. Toxicol Lett. 2015;239(2):123–130. doi:10.1016/j.toxlet.2015.09.013
  • Savi M, Rossi S, Bocchi L, et al. Titanium dioxide nanoparticles promote arrhythmias via a direct interaction with rat cardiac tissue. Part Fibre Toxicol. 2014;11(1):63. doi:10.1186/s12989-014-0063-3
  • Hong F, Zhao X, Chen M, et al. TiO2 nanoparticles-induced apoptosis of primary cultured Sertoli cells of mice. J Biomed Mater Res A. 2016;104(1):124–135. doi:10.1002/jbm.a.35548
  • Hussain S, Thomassen LC, Ferecatu I, et al. Carbon black and titanium dioxide nanoparticles elicit distinct apoptotic pathways in bronchial epithelial cells. Part Fibre Toxicol. 2010;7:10. doi:10.1186/1743-8977-7-10
  • Li X, Kang B, Eom Y, et al. Comparison of cytotoxicity effects induced by four different types of nanoparticles in human corneal and conjunctival epithelial cells. Sci Rep. 2022;12(1):155. doi:10.1038/s41598-021-04199-3
  • Rihane N, Nury T, M’rad I, et al. Microglial cells (BV-2) internalize titanium dioxide (TiO2) nanoparticles: toxicity and cellular responses. Environ Sci Pollut Res Int. 2016;23(10):9690–9699. doi:10.1007/s11356-016-6190-7
  • Teleanu DM, Chircov C, Grumezescu A, et al. Neurotoxicity of nanomaterials: an up-to-date overview. Nanomaterials. 2019;9(1):96. doi:10.3390/nano9010096
  • Liu Y, He Q. The route of nanomaterials entering brain. In: Neurotoxicity of Nanomaterials and Nanomedicine. Elsevier; 2017:33–57.
  • Liu X, Sui B, Sun J. Size- and shape-dependent effects of titanium dioxide nanoparticles on the permeabilization of the blood-brain barrier. J Mater Chem B. 2017;5(48):9558–9570. doi:10.1039/c7tb01314k
  • Jiang X, Gao H. Neurotoxicity of Nanomaterials and Nanomedicine. Academic Press; 2016.
  • Bencsik A, Lestaevel P, Guseva Canu I. Nano- and neurotoxicology: an emerging discipline. Prog Neurobiol. 2018;160:45–63. doi:10.1016/j.pneurobio.2017.10.003
  • Teschke K, Marion SA, Tsui JKC, et al. Parkinson’s disease and occupation: differences in associations by case identification method suggest referral bias. Am. J. Ind. Med. 2014;57(2):163–171. doi:10.1002/ajim.22272
  • Andujar P, Simon-Deckers A, Galateau-Sallé F, et al. Role of metal oxide nanoparticles in histopathological changes observed in the lung of welders. Part Fibre Toxicol. 2014;11(1):23. doi:10.1186/1743-8977-11-23
  • Iqubal A, Ahmed M, Ahmad S, et al. Environmental neurotoxic pollutants: review. Environ Sci Pollut Res Int. 2020;27(33):41175–41198. doi:10.1007/s11356-020-10539-z
  • Gottschalk F, Sonderer T, Scholz RW, et al. Modeled environmental concentrations of engineered nanomaterials (TiO 2, ZnO, Ag, CNT, Fullerenes) for different regions. Environ Sci Technol. 2009;43(24):9216–9222. doi:10.1021/es9015553
  • Zhang R, Zhang H, Tu C, et al. Facilitated transport of titanium dioxide nanoparticles by humic substances in saturated porous media under acidic conditions. J Nanopart Res. 2015;17(4):1–11. doi:10.1007/s11051-015-2972-y
  • Gosset P, Lassalle P, Vanhée D, et al. Production of tumor necrosis factor-α and Interleukin-6 by human alveolar macrophages exposed in vitro to coal mine dust. Am J Respir Cell Mol Biol. 1991;5(5):431–436. doi:10.1165/ajrcmb/5.5.431
  • Mao Y, Bajinka O, Tang Z, et al. Lung-brain axis: metabolomics and pathological changes in lungs and brain of respiratory syncytial virus-infected mice. J Med Virol. 2022;94(12):5885–5893. doi:10.1002/jmv.28061
  • Jiang Y, Wei Y, Guo W, et al. Prenatal titanium exposure and child neurodevelopment at 1 year of age: a longitudinal prospective birth cohort study. Chemosphere. 2023;311(Pt 1):137034. doi:10.1016/j.chemosphere.2022.137034
  • Li Z, Huo W, Li Z, et al. Association between titanium and silver concentrations in maternal hair and risk of neural tube defects in offspring: a case-control study in north China. Reprod Toxicol. 2016;66:115–121. doi:10.1016/j.reprotox.2016.10.006
  • Zhang X, Mei D, Li Y, et al. Arsenic exposure via drinking water during pregnancy and lactation induces autism-like behaviors in male offspring mice. Chemosphere. 2022;290:133338. doi:10.1016/j.chemosphere.2021.133338
  • Mohammadipour A, Hosseini M, Fazel A, et al. The effects of exposure to titanium dioxide nanoparticles during lactation period on learning and memory of rat offspring. Toxicol Ind Health. 2016;32(2):221–228. doi:10.1177/0748233713498440
  • Mortensen NP, Pathmasiri W, Snyder RW, et al. Oral administration of TiO(2) nanoparticles during early life impacts cardiac and neurobehavioral performance and metabolite profile in an age- and sex-related manner. Part Fibre Toxicol. 2022;19(1):3. doi:10.1186/s12989-021-00444-9
  • Mohammadipour A, Fazel A, Haghir H, et al. Maternal exposure to titanium dioxide nanoparticles during pregnancy; impaired memory and decreased hippocampal cell proliferation in rat offspring. Environ Toxicol Pharmacol. 2014;37(2):617–625. doi:10.1016/j.etap.2014.01.014
  • Cui Y, Chen X, Zhou Z, et al. Prenatal exposure to nanoparticulate titanium dioxide enhances depressive-like behaviors in adult rats. Chemosphere. 2014;96:99–104. doi:10.1016/j.chemosphere.2013.07.051
  • Engler-Chiurazzi EB, Stapleton PA, Stalnaker JJ, et al. Impacts of prenatal nanomaterial exposure on male adult Sprague-Dawley rat behavior and cognition. J Toxicol Environ Health A. 2016;79(11):447–452. doi:10.1080/15287394.2016.1164101
  • Asghari A, Hosseini M, Beheshti F, et al. Inducible nitric oxide inhibitor aminoguanidine, ameliorated oxidative stress, interleukin-6 concentration and improved brain-derived neurotrophic factor in the brain tissues of neonates born from titanium dioxide nanoparticles exposed rats. J Matern Fetal Neonatal Med. 2019;32(23):3962–3973. doi:10.1080/14767058.2018.1480602
  • Ebrahimzadeh Bideskan A, Mohammadipour A, Fazel A, et al. Maternal exposure to titanium dioxide nanoparticles during pregnancy and lactation alters offspring hippocampal mRNA BAX and Bcl-2 levels, induces apoptosis and decreases neurogenesis. Exp Toxicol Pathol. 2017;69(6):329–337. doi:10.1016/j.etp.2017.02.006
  • Gao X, Yin S, Tang M, et al. Effects of developmental exposure to TiO2 nanoparticles on synaptic plasticity in hippocampal dentate gyrus area: an in vivo study in anesthetized rats. Biol Trace Elem Res. 2011;143(3):1616–1628. doi:10.1007/s12011-011-8990-4
  • Grissa I, Guezguez S, Ezzi L, et al. The effect of titanium dioxide nanoparticles on neuroinflammation response in rat brain. Environ Sci Pollut Res Int. 2016;23(20):20205–20213. doi:10.1007/s11356-016-7234-8
  • Grissa I, ElGhoul J, Mrimi R, et al. In deep evaluation of the neurotoxicity of orally administered TiO(2) nanoparticles. Brain Res Bull. 2020;155:119–128. doi:10.1016/j.brainresbull.2019.10.005
  • Nalika N, Waseem M, Kaushik P, et al. Role of melatonin and quercetin as countermeasures to the mitochondrial dysfunction induced by titanium dioxide nanoparticles. Life Sci. 2023;328:121403. doi:10.1016/j.lfs.2023.121403
  • Krawczynska A, Dziendzikowska K, Gromadzka-Ostrowska J, et al. Silver and titanium dioxide nanoparticles alter oxidative/inflammatory response and renin-angiotensin system in brain. Food Chem Toxicol. 2015;85:96–105. doi:10.1016/j.fct.2015.08.005
  • Valentini X, Deneufbourg P, Paci P, et al. Morphological alterations induced by the exposure to TiO(2) nanoparticles in primary cortical neuron cultures and in the brain of rats. Toxicol Rep. 2018;5:878–889. doi:10.1016/j.toxrep.2018.08.006
  • Naima R, Imen M, Mustapha J, et al. Acute titanium dioxide nanoparticles exposure impaired spatial cognitive performance through neurotoxic and oxidative mechanisms in Wistar rats. Biomarkers. 2021;26(8):760–769. doi:10.1080/1354750X.2021.1999501
  • Horvath T, Papp A, Kovács D, et al. Electrophysiological alterations and general toxic signs obtained by subacute administration of titanium dioxide nanoparticles to the airways of rats. Ideggyogy Sz. 2017;70(3–4):127–135. doi:10.18071/isz.70.0127
  • Papp A, Horváth T, Igaz N, et al. Presence of titanium and toxic effects observed in rat lungs, kidneys, and central nervous system in vivo and in cultured astrocytes in vitro on exposure by titanium dioxide nanorods. Int J Nanomedicine. 2020;15:9939–9960. doi:10.2147/IJN.S275937
  • Horvath T, Vezér T, Kozma G, et al. Functional neurotoxicity and tissue metal levels in rats exposed subacutely to titanium dioxide nanoparticles via the airways. Ideggyogy Sz. 2018;71(1–02):35–42. doi:10.18071/isz.71.0035
  • Younes NR, Amara S, Mrad I, et al. Subacute toxicity of titanium dioxide (TiO2) nanoparticles in male rats: emotional behavior and pathophysiological examination. Environ Sci Pollut Res Int. 2015;22(11):8728–8737. doi:10.1007/s11356-014-4002-5
  • Nalika N, Parvez S. Mitochondrial dysfunction in titanium dioxide nanoparticle-induced neurotoxicity. Toxicol Mech Methods. 2015;25(5):355–363. doi:10.3109/15376516.2015.1020183
  • Cui Y, Che Y, Wang H. Nono-titanium dioxide exposure during the adolescent period induces neurotoxicities in rats: ameliorative potential of bergamot essential oil. Brain Behav. 2021;11(5):e02099. doi:10.1002/brb3.2099
  • Latif MA, Jabeen F, Ali M, et al. Neurotoxic effects of titanium dioxide nanoparticles on the brain of male sprague dawley rats. Pak J Pharm Sci. 2019;32(5(Supplementary)):2311–2316.
  • Disdier C, Chalansonnet M, Gagnaire F, et al. Brain inflammation, blood brain barrier dysfunction and neuronal synaptophysin decrease after inhalation exposure to titanium dioxide nano-aerosol in aging rats. Sci Rep. 2017;7(1):12196. doi:10.1038/s41598-017-12404-5
  • Halawa A, Elshopakey G, El‐Adl M, et al. Chitosan attenuated the neurotoxicity-induced titanium dioxide nanoparticles in brain of adult rats. Environ Toxicol Int J. 2022;37(3):612–626. doi:10.1002/tox.23429
  • Herting MM, Sowell ER. Puberty and structural brain development in humans. Front Neuroendocrinol. 2017;44:122–137. doi:10.1016/j.yfrne.2016.12.003
  • Lees B, Meredith LR, Kirkland AE, et al. Effect of alcohol use on the adolescent brain and behavior. Pharmacol Biochem Behav. 2020;192:172906. doi:10.1016/j.pbb.2020.172906
  • Jiang M, Jang SE, Zeng L. The effects of extrinsic and intrinsic factors on neurogenesis. Cells. 2023;12(9):1285. doi:10.3390/cells12091285
  • Ribeiro FF, Xapelli S. An overview of adult neurogenesis. Adv Exp Med Biol. 2021;1331:77–94. doi:10.1007/978-3-030-74046-7_7
  • Erdo F, Bors LA, Farkas D, Bajza Á, Gizurarson S. Evaluation of intranasal delivery route of drug administration for brain targeting. Brain Res Bull. 2018;143:155–170. doi:10.1016/j.brainresbull.2018.10.009
  • Ze Y, Hu R, Wang X, et al. Neurotoxicity and gene-expressed profile in brain-injured mice caused by exposure to titanium dioxide nanoparticles. J Biomed Mater Res A. 2014;102(2):470–478. doi:10.1002/jbm.a.34705
  • Wang J, Liu Y, Jiao F, et al. Time-dependent translocation and potential impairment on central nervous system by intranasally instilled TiO(2) nanoparticles. Toxicology. 2008;254(1–2):82–90. doi:10.1016/j.tox.2008.09.014
  • Zhang L, Bai R, Li B, et al. Rutile TiO(2) particles exert size and surface coating dependent retention and lesions on the murine brain. Toxicol Lett. 2011;207(1):73–81. doi:10.1016/j.toxlet.2011.08.001
  • Ze X, Su M, Zhao X, et al. TiO2 nanoparticle-induced neurotoxicity may be involved in dysfunction of glutamate metabolism and its receptor expression in mice. Environ Toxicol: Int J. 2016;31(6):655–662. doi:10.1002/tox.22077
  • Ze Y, Sheng L, Zhao X, et al. TiO2 nanoparticles induced hippocampal neuroinflammation in mice. PLoS One. 2014;9(3):e92230. doi:10.1371/journal.pone.0092230
  • Wang J, Chen C, Liu Y, et al. Potential neurological lesion after nasal instillation of TiO(2) nanoparticles in the anatase and rutile crystal phases. Toxicol Lett. 2008;183(1–3):72–80. doi:10.1016/j.toxlet.2008.10.001
  • Ze Y, Zheng L, Zhao X, et al. Molecular mechanism of titanium dioxide nanoparticles-induced oxidative injury in the brain of mice. Chemosphere. 2013;92(9):1183–1189. doi:10.1016/j.chemosphere.2013.01.094
  • Tachibana K, Kawazoe S, Onoda A, et al. Effects of prenatal exposure to titanium dioxide nanoparticles on DNA methylation and gene expression profile in the mouse brain. Front Toxicol. 2021;3:705910. doi:10.3389/ftox.2021.705910
  • Ze Y, Sheng L, Zhao X, et al. Neurotoxic characteristics of spatial recognition damage of the hippocampus in mice following subchronic peroral exposure to TiO2 nanoparticles. J Hazard Mater. 2014;264:219–229. doi:10.1016/j.jhazmat.2013.10.072
  • Vandenberg LN, Welshons WV, Vom Saal FS, Toutain PL, Myers JP. Should oral gavage be abandoned in toxicity testing of endocrine disruptors? Environ Health. 2014;13(1):46. doi:10.1186/1476-069X-13-46
  • Su J, Duan X, Qiu Y, et al. Pregnancy exposure of titanium dioxide nanoparticles causes intestinal dysbiosis and neurobehavioral impairments that are not significant postnatally but emerge in adulthood of offspring. J Nanobiotechnology. 2021;19(1):234. doi:10.1186/s12951-021-00967-5
  • Yang C, Xue J, Qin Q, et al. Prenatal exposure to titanium dioxide nanoparticles induces persistent neurobehavioral impairments in maternal mice that is associated with microbiota-gut-brain axis. Food Chem Toxicol. 2022;169:113402. doi:10.1016/j.fct.2022.113402
  • Zhou Y, Ji J, Chen C, Hong F. Retardation of axonal and dendritic outgrowth is associated with the MAPK signaling pathway in offspring mice following maternal exposure to nanosized titanium dioxide. J Agric Food Chem. 2019;67(9):2709–2715. doi:10.1021/acs.jafc.8b06992
  • Hong F, Zhou Y, Ji J, et al. Nano-TiO(2) inhibits development of the central nervous system and its mechanism in offspring mice. J Agric Food Chem. 2018;66(44):11767–11774. doi:10.1021/acs.jafc.8b02952
  • Zhou Y, Hong F, Tian Y, et al. Nanoparticulate titanium dioxide-inhibited dendritic development is involved in apoptosis and autophagy of hippocampal neurons in offspring mice. Toxicol Res. 2017;6(6):889–901. doi:10.1039/c7tx00153c
  • Heidari Z, Mohammadipour A, Haeri P, Ebrahimzadeh-Bideskan A. The effect of titanium dioxide nanoparticles on mice midbrain substantia nigra. Iran J Basic Med Sci. 2019;22(7):745–751. doi:10.22038/ijbms.2019.33611.8018
  • Umezawa M, Tainaka H, Kawashima N, et al. Effect of fetal exposure to titanium dioxide nanoparticle on brain development − brain region information. J Toxicol Sci. 2012;37(6):1247–1252. doi:10.2131/jts.37.1247
  • Shimizu M, Tainaka H, Oba T, et al. Maternal exposure to nanoparticulate titanium dioxide during the prenatal period alters gene expression related to brain development in the mouse. Part Fibre Toxicol. 2009;6(1):20. doi:10.1186/1743-8977-6-20
  • Takahashi Y, Mizuo K, Shinkai Y, et al. Prenatal exposure to titanium dioxide nanoparticles increases dopamine levels in the prefrontal cortex and neostriatum of mice. J Toxicol Sci. 2010;35(5):749–756. doi:10.2131/jts.35.749
  • Tarlan M, Sajedianfard J, Fathi M. Effect of titanium dioxide nanoparticles administered during pregnancy on depression-like behavior in forced swimming and tail suspension tests in offspring mice. Toxicol Ind Health. 2020;36(4):297–304. doi:10.1177/0748233720925707
  • Ma L, Liu J, Li N, et al. Oxidative stress in the brain of mice caused by translocated nanoparticulate TiO2 delivered to the abdominal cavity. Biomaterials. 2010;31(1):99–105. doi:10.1016/j.biomaterials.2009.09.028
  • Habibi P, Ostad SN, Monazzam MR, et al. Thermal stress and TiO(2) nanoparticle-induced oxidative DNA damage and apoptosis in mouse hippocampus. Environ Sci Pollut Res Int. 2022;29(60):90128–90139. doi:10.1007/s11356-022-21796-5
  • Wang J-X, Li Y-F, Zhou G-Q, et al. [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(2):91–95. Chinese.
  • Hu R, Gong X, Duan Y, et al. Neurotoxicological effects and the impairment of spatial recognition memory in mice caused by exposure to TiO2 nanoparticles. Biomaterials. 2010;31(31):8043–8050. doi:10.1016/j.biomaterials.2010.07.011
  • Hu R, Zheng L, Zhang T, et al. Molecular mechanism of hippocampal apoptosis of mice following exposure to titanium dioxide nanoparticles. J Hazard Mater. 2011;191(1–3):32–40. doi:10.1016/j.jhazmat.2011.04.027
  • Jia X, Wang S, Zhou L, et al. The potential liver, brain, and embryo toxicity of titanium dioxide nanoparticles on mice. Nanoscale Res Lett. 2017;12(1):478. doi:10.1186/s11671-017-2242-2
  • Zhang S, Jiang X, Cheng S, et al. Titanium dioxide nanoparticles via oral exposure leads to adverse disturbance of gut microecology and locomotor activity in adult mice. Arch Toxicol. 2020;94(4):1173–1190. doi:10.1007/s00204-020-02698-2
  • Shin JA, Lee EJ, Seo SM, et al. Nanosized titanium dioxide enhanced inflammatory responses in the septic brain of mouse. Neuroscience. 2010;165(2):445–454. doi:10.1016/j.neuroscience.2009.10.057
  • Notter T, Aengenheister L, Weber-Stadlbauer U, et al. Prenatal exposure to TiO(2) nanoparticles in mice causes behavioral deficits with relevance to autism spectrum disorder and beyond. Transl Psychiatry. 2018;8(1):193. doi:10.1038/s41398-018-0251-2
  • Sofranko A, Wahle T, Heusinkveld HJ, et al. Evaluation of the neurotoxic effects of engineered nanomaterials in C57BL/6J mice in 28-day oral exposure studies. Neurotoxicology. 2021;84:155–171. doi:10.1016/j.neuro.2021.03.005
  • Mohamadzadeh N, Zirak Javanmard M, Karimipour M, et al. Developmental toxicity of the neural tube induced by titanium dioxide nanoparticles in mouse embryos. Avicenna J Med Biotechnol. 2021;13(2):74–80. doi:10.18502/ajmb.v13i2.5524
  • Shrivastava R, Raza S, Yadav A, et al. Effects of sub-acute exposure to TiO 2, ZnO and Al2 O3 nanoparticles on oxidative stress and histological changes in mouse liver and brain. Drug Chem Toxicol. 2014;37(3):336–347. doi:10.3109/01480545.2013.866134
  • Mohamed HR, Hussien NA. Genotoxicity studies of Titanium Dioxide Nanoparticles (TiO2NPs) in the brain of mice. Scientifica. 2016;2016:6710840. doi:10.1155/2016/6710840
  • Liu W, Huang G, Su X, et al. Zebrafish: a promising model for evaluating the toxicity of carbon dot-based nanomaterials. ACS Appl Mater Interfaces. 2020;12(43):49012–49020. doi:10.1021/acsami.0c17492
  • Hu S, Han J, Yang L, et al. Impact of co-exposure to titanium dioxide nanoparticles and Pb on zebrafish embryos. Chemosphere. 2019;233:579–589. doi:10.1016/j.chemosphere.2019.06.009
  • Miao W, Zhu B, Xiao X, et al. Effects of titanium dioxide nanoparticles on lead bioconcentration and toxicity on thyroid endocrine system and neuronal development in zebrafish larvae. Aquat Toxicol. 2015;161:117–126. doi:10.1016/j.aquatox.2015.02.002
  • Wang Q, Chen Q, Zhou P, et al. Bioconcentration and metabolism of BDE-209 in the presence of titanium dioxide nanoparticles and impact on the thyroid endocrine system and neuronal development in zebrafish larvae. Nanotoxicology. 2014;8 Suppl 1:196–207. doi:10.3109/17435390.2013.875232
  • Li M, Wu Q, Wang Q, Xiang D, Zhu G. Effect of titanium dioxide nanoparticles on the bioavailability and neurotoxicity of cypermethrin in zebrafish larvae. Aquat Toxicol. 2018;199:212–219. doi:10.1016/j.aquatox.2018.03.022
  • Fan B, Dai L, Liu C, et al. Nano-TiO(2) aggravates bioaccumulation and developmental neurotoxicity of triphenyl phosphate in zebrafish larvae. Chemosphere. 2022;287(Pt 3):132161. doi:10.1016/j.chemosphere.2021.132161
  • Fu J, Guo Y, Yang L, Han J, Zhou B. Nano-TiO(2) enhanced bioaccumulation and developmental neurotoxicity of bisphenol a in zebrafish larvae. Environ Res. 2020;187:109682. doi:10.1016/j.envres.2020.109682
  • Guo Y, Chen L, Wu J, et al. Parental co-exposure to bisphenol A and nano-TiO(2) causes thyroid endocrine disruption and developmental neurotoxicity in zebrafish offspring. Sci Total Environ. 2019;650(Pt 1):557–565. doi:10.1016/j.scitotenv.2018.09.007
  • Zhu R, Liu C, Wang J, et al. Nano-TiO(2) aggravates bioaccumulation and developmental neurotoxicity of difenoconazole in zebrafish larvae via oxidative stress and apoptosis: protective role of vitamin C. Ecotoxicol Environ Saf. 2023;251:114554. doi:10.1016/j.ecoenv.2023.114554
  • Xu L, Yang X, He Y, Hu Q, Fu Z. Combined exposure to titanium dioxide and tetracycline induces neurotoxicity in zebrafish. Comp Biochem Physiol C. 2023;267:109562. doi:10.1016/j.cbpc.2023.109562
  • Wu Q, Yan W, Liu C, et al. Co-exposure with titanium dioxide nanoparticles exacerbates MCLR-induced brain injury in zebrafish. Sci Total Environ. 2019;693:133540. doi:10.1016/j.scitotenv.2019.07.346
  • Lei L, Qiao K, Guo Y, et al. Titanium dioxide nanoparticles enhanced thyroid endocrine disruption of pentachlorophenol rather than neurobehavioral defects in zebrafish larvae. Chemosphere. 2020;249:126536. doi:10.1016/j.chemosphere.2020.126536
  • Sheng L, Wang L, Su M, et al. Mechanism of TiO2 nanoparticle-induced neurotoxicity in zebrafish (D anio rerio). Environ Toxicol Int J. 2016;31(2):163–175. doi:10.1002/tox.22031
  • Chen J, Li J, Jiang H, et al. Developmental co-exposure of TBBPA and titanium dioxide nanoparticle induced behavioral deficits in larval zebrafish. Ecotoxicol Environ Saf. 2021;215:112176. doi:10.1016/j.ecoenv.2021.112176
  • Chen J, Lei L, Mo W, et al. Developmental titanium dioxide nanoparticle exposure induces oxidative stress and neurobehavioral changes in zebrafish. Aquat Toxicol. 2021;240:105990. doi:10.1016/j.aquatox.2021.105990
  • Gu J, Guo M, Huang C, et al. Titanium dioxide nanoparticle affects motor behavior, neurodevelopment and axonal growth in zebrafish (Danio rerio) larvae. Sci Total Environ. 2021;754:142315. doi:10.1016/j.scitotenv.2020.142315
  • Sun X, Yang Q, Jing M, et al. Environmentally relevant concentrations of organic (benzophenone-3) and inorganic (titanium dioxide nanoparticles) UV filters co-exposure induced neurodevelopmental toxicity in zebrafish. Ecotoxicol Environ Saf. 2023;249:114343. doi:10.1016/j.ecoenv.2022.114343
  • Hu Q, Guo F, Zhao F, et al. 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
  • Caruso G, Scalisi EM, Pecoraro R, et al. Effects of carnosine on the embryonic development and TiO(2) nanoparticles-induced oxidative stress on Zebrafish. Front Vet Sci. 2023;10:1148766. doi:10.3389/fvets.2023.1148766
  • Palaniappan PR, Pramod KS. The effect of titanium dioxide on the biochemical constituents of the brain of Zebrafish (Danio rerio): an FT-IR study. Spectrochim Acta A Mol Biomol Spectrosc. 2011;79(1):206–212. doi:10.1016/j.saa.2011.02.038
  • Kaur K, Narang RK, Singh S. Glabridin mitigates TiO(2)NP induced cognitive deficit in adult zebrafish. Neurochem Int. 2023;169:105585. doi:10.1016/j.neuint.2023.105585
  • Bauer B, Mally A, Liedtke D. Zebrafish embryos and larvae as alternative animal models for toxicity testing. Int J Mol Sci. 2021;22(24):13417. doi:10.3390/ijms222413417
  • Zhao Y, Wu Q, Tang M, et al. The in vivo underlying mechanism for recovery response formation in nano-titanium dioxide exposed Caenorhabditis elegans after transfer to the normal condition. Nanomedicine. 2014;10(1):89–98. doi:10.1016/j.nano.2013.07.004
  • Hu CC, Wu G-H, Hua T-E, et al. Uptake of TiO 2 nanoparticles into C. elegans neurons negatively affects axonal growth and worm locomotion behavior. ACS Appl Mater Interfaces. 2018;10(10):8485–8495. doi:10.1021/acsami.7b18818
  • Zhang X, Song Y, Wang J, et al. Chronic exposure to titanium dioxide nanoparticles induces deficits of locomotor behavior by disrupting the development of NMJ in Drosophila. Sci Total Environ. 2023;888:164076. doi:10.1016/j.scitotenv.2023.164076
  • Guan X, Shi W, Zha S, et al. Neurotoxic impact of acute TiO(2) nanoparticle exposure on a benthic marine bivalve mollusk, Tegillarca granosa. Aquat Toxicol. 2018;200:241–246. doi:10.1016/j.aquatox.2018.05.011
  • Khalil AM. Neurotoxicity and biochemical responses in the earthworm Pheretima hawayana exposed to TiO2NPs. Ecotoxicol Environ Saf. 2015;122:455–461. doi:10.1016/j.ecoenv.2015.09.010
  • Ruszkiewicz JA, Pinkas A, Miah MR, et al. C. elegans as a model in developmental neurotoxicology. Toxicol Appl Pharmacol. 2018;354:126–135. doi:10.1016/j.taap.2018.03.016
  • Chifiriuc MC, Ratiu A, Popa M, et al. Drosophotoxicology: an emerging research area for assessing nanoparticles interaction with living organisms. Int J Mol Sci. 2016;17(2):36. doi:10.3390/ijms17020036
  • Ong C, Yung L-YL, Cai Y, et al. Drosophila melanogaster as a model organism to study nanotoxicity. Nanotoxicology. 2015;9(3):396–403. doi:10.3109/17435390.2014.940405
  • Sun TY, Bornhöft NA, Hungerbühler K, et al. Dynamic probabilistic modeling of environmental emissions of engineered nanomaterials. Environ Sci Technol. 2016;50(9):4701–4711. doi:10.1021/acs.est.5b05828
  • McShane H, Sarrazin M, Whalen JK, et al. Reproductive and behavioral responses of earthworms exposed to nano-sized titanium dioxide in soil. Environ Toxicol Chem. 2012;31(1):184–193. doi:10.1002/etc.714
  • Tiffany-Castiglioni E, Ehrich M, Dees L, et al. Bridging the gap between in vitro and in vivo models for neurotoxicology. Toxicol Sci. 1999;51(2):178–183. doi:10.1093/toxsci/51.2.178
  • Viesselmann C, Ballweg J, Lumbard D, Dent EW. Nucleofection and primary culture of embryonic mouse hippocampal and cortical neurons. J Vis Exp. 2011;47:e2373.
  • Sheng L, Ze Y, Wang L, et al. Mechanisms of TiO2 nanoparticle-induced neuronal apoptosis in rat primary cultured hippocampal neurons. J Biomed Mater Res A. 2015;103(3):1141–1149. doi:10.1002/jbm.a.35263
  • Gerber LS, Heusinkveld HJ, Langendoen C, et al. Acute, sub-chronic and chronic exposures to TiO(2) and Ag nanoparticles differentially affects neuronal function in vitro. Neurotoxicology. 2022;93:311–323. doi:10.1016/j.neuro.2022.10.010
  • Strickland JD, Lefew WR, Crooks J, et al. In vitro screening of metal oxide nanoparticles for effects on neural function using cortical networks on microelectrode arrays. Nanotoxicology. 2016;10(5):619–628. doi:10.3109/17435390.2015.1107142
  • Sun Y, Wang S, Zheng J. Biosynthesis of TiO(2) nanoparticles and their application for treatment of brain injury-an in-vitro toxicity study towards central nervous system. J Photochem Photobiol B. 2019;194:1–5. doi:10.1016/j.jphotobiol.2019.02.008
  • Gramowski A, Flossdorf J, Bhattacharya K, et al. Nanoparticles induce changes of the electrical activity of neuronal networks on microelectrode array neurochips. Environ Health Perspect. 2010;118(10):1363–1369. doi:10.1289/ehp.0901661
  • Winter M, Beer H-D, Hornung V, et al. Activation of the inflammasome by amorphous silica and TiO2 nanoparticles in murine dendritic cells. Nanotoxicology. 2011;5(3):326–340. doi:10.3109/17435390.2010.506957
  • Hong F, Sheng L, Ze Y, et al. Suppression of neurite outgrowth of primary cultured hippocampal neurons is involved in impairment of glutamate metabolism and NMDA receptor function caused by nanoparticulate TiO2. Biomaterials. 2015;53:76–85. doi:10.1016/j.biomaterials.2015.02.067
  • Mu X, Li W, Ze X, et al. Molecular mechanism of nanoparticulate TiO2 induction of axonal development inhibition in rat primary cultured hippocampal neurons. Environ Toxicol: Int J. 2020;35(8):895–905. doi:10.1002/tox.22926
  • Hong F, Ze Y, Zhou Y, et al. Nanoparticulate TiO 2-mediated inhibition of the Wnt signaling pathway causes dendritic development disorder in cultured rat hippocampal neurons. J Biomed Mater Res A. 2017;105(8):2139–2149. doi:10.1002/jbm.a.36073
  • Hong F, Ze X, Mu X, et al. Titanium dioxide inhibits hippocampal neuronal synapse growth through the brain-derived neurotrophic factor-tyrosine kinase receptor B signaling pathway. J Biomed Nanotechnol. 2021;17(1):37–52. doi:10.1166/jbn.2021.2999
  • Liu S, Xu L, Zhang T, et al. Oxidative stress and apoptosis induced by nanosized titanium dioxide in PC12 cells. Toxicology. 2010;267(1–3):172–177. doi:10.1016/j.tox.2009.11.012
  • Wu J, Xie H. Effects of titanium dioxide nanoparticles on alpha-synuclein aggregation and the ubiquitin-proteasome system in dopaminergic neurons. Artif Cells Nanomed Biotechnol. 2016;44(2):690–694. doi:10.3109/21691401.2014.980507
  • Irie T, Kawakami T, Sato K, et al. Sub-toxic concentrations of nano-ZnO and nano-TiO2 suppress neurite outgrowth in differentiated PC12 cells. J Toxicol Sci. 2017;42(6):723–729. doi:10.2131/jts.42.723
  • Wu J, Sun J, Xue Y. Involvement of JNK and P53 activation in G2/M cell cycle arrest and apoptosis induced by titanium dioxide nanoparticles in neuron cells. Toxicol Lett. 2010;199(3):269–276. doi:10.1016/j.toxlet.2010.09.009
  • Ferraro SA, Domingo MG, Etcheverrito A, et al. Neurotoxicity mediated by oxidative stress caused by titanium dioxide nanoparticles in human neuroblastoma (SH-SY5Y) cells. J Trace Elem Med Biol. 2020;57:126413. doi:10.1016/j.jtemb.2019.126413
  • Lojk J, Repas J, Veranič P, et al. Toxicity mechanisms of selected engineered nanoparticles on human neural cells in vitro. Toxicology. 2020;432:152364. doi:10.1016/j.tox.2020.152364
  • Valdiglesias V, Costa C, Sharma V, et al. Comparative study on effects of two different types of titanium dioxide nanoparticles on human neuronal cells. Food Chem Toxicol. 2013;57:352–361. doi:10.1016/j.fct.2013.04.010
  • Mao Z, Xu B, Ji X, et al. Titanium dioxide nanoparticles alter cellular morphology via disturbing the microtubule dynamics. Nanoscale. 2015;7(18):8466–8475. doi:10.1039/c5nr01448d
  • Rosario F, Costa C, Lopes CB, et al. In vitro hepatotoxic and neurotoxic effects of titanium and cerium dioxide nanoparticles, arsenic and mercury co-exposure. Int J Mol Sci. 2022;23(5):2737. doi:10.3390/ijms23052737
  • Zhou T, Huang W-K, Xu Q-Y, et al. Nec-1 attenuates neurotoxicity induced by titanium dioxide nanomaterials on Sh-Sy5y cells through RIP1. Nanoscale Res Lett. 2020;15(1):65. doi:10.1186/s11671-020-03300-5
  • He Q, Zhou X, Liu Y, et al. Titanium dioxide nanoparticles induce mouse hippocampal neuron apoptosis via oxidative stress- and calcium imbalance-mediated endoplasmic reticulum stress. Environ Toxicol Pharmacol. 2018;63:6–15. doi:10.1016/j.etap.2018.08.003
  • Shafer TJ, Atchison WD. Transmitter, ion channel and receptor properties of pheochromocytoma (PC12) cells: a model for neurotoxicological studies. Neurotoxicology. 1991;12(3):473–492.
  • Lopez-Suarez L, Awabdh SA, Coumoul X, et al. The SH-SY5Y human neuroblastoma cell line, a relevant in vitro cell model for investigating neurotoxicology in human: focus on organic pollutants. Neurotoxicology. 2022;92:131–155. doi:10.1016/j.neuro.2022.07.008
  • Coccini T, Grandi S, Lonati D, et al. 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
  • Orihuela R, McPherson CA, Harry GJ. Microglial M1/M2 polarization and metabolic states. Br J Pharmacol. 2016;173(4):649–665. doi:10.1111/bph.13139
  • Long TC, Saleh N, Tilton RD, et al. Titanium dioxide (P25) produces reactive oxygen species in immortalized brain microglia (BV2): implications for nanoparticle neurotoxicity. Environ Sci Technol. 2006;40(14):4346–4352. doi:10.1021/es060589n
  • Colombo E, Farina C. Astrocytes: key regulators of neuroinflammation. Trends Immunol. 2016;37(9):608–620. doi:10.1016/j.it.2016.06.006
  • Wilson CL, Natarajan V, Hayward SL, et al. Mitochondrial dysfunction and loss of glutamate uptake in primary astrocytes exposed to titanium dioxide nanoparticles. Nanoscale. 2015;7(44):18477–18488. doi:10.1039/c5nr03646a
  • Perez-Arizti JA, Ventura-Gallegos JL, Galván Juárez RE, et al. Titanium dioxide nanoparticles promote oxidative stress, autophagy and reduce NLRP3 in primary rat astrocytes. Chem Biol Interact. 2020;317:108966. doi:10.1016/j.cbi.2020.108966
  • Liu Y, Xu Z, Li X. Cytotoxicity of titanium dioxide nanoparticles in rat neuroglia cells. Brain Inj. 2013;27(7–8):934–939. doi:10.3109/02699052.2013.793401
  • Marquez-Ramirez SG, Delgado-Buenrostro NL, Chirino YI, et al. Titanium dioxide nanoparticles inhibit proliferation and induce morphological changes and apoptosis in glial cells. Toxicology. 2012;302(2–3):146–156. doi:10.1016/j.tox.2012.09.005
  • Huerta-Garcia E, Pérez-Arizti JA, Márquez-Ramírez SG, et al. 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
  • 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(2):91–98. doi:10.1016/j.toxlet.2012.08.009
  • Long TC, Tajuba J, Sama P, et al. Nanosize titanium dioxide stimulates reactive oxygen species in brain microglia and damages neurons in vitro. Environ Health Perspect. 2007;115(11):1631–1637. doi:10.1289/ehp.10216
  • Hsiao I-L, Chang -C-C, Wu C-Y, et al. Indirect effects of TiO2 nanoparticle on neuron-glial cell interactions. Chem Biol Interact. 2016;254:34–44. doi:10.1016/j.cbi.2016.05.024
  • 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(3):886–896. doi:10.1016/j.biomaterials.2011.10.025
  • Fuster E, Candela H, Estévez J, et al. Titanium dioxide, but not zinc oxide, nanoparticles cause severe transcriptomic alterations in T98G human glioblastoma cells. Int J Mol Sci. 2021;22(4):2084. doi:10.3390/ijms22042084
  • Erriquez J, Bolis V, Morel S, et al. Nanosized TiO2 is internalized by dorsal root ganglion cells and causes damage via apoptosis. Nanomedicine. 2015;11(6):1309–1319. doi:10.1016/j.nano.2015.04.003
  • Bolis V, Busco C, Ciarletta M, et al. Hydrophilic/hydrophobic features of TiO2 nanoparticles as a function of crystal phase, surface area and coating, in relation to their potential toxicity in peripheral nervous system. J Colloid Interface Sci. 2012;369(1):28–39. doi:10.1016/j.jcis.2011.11.058
  • Yu Y, Ren W, Ren B. Nanosize titanium dioxide cause neuronal apoptosis: a potential linkage between nanoparticle exposure and neural disorder. Neurol Res. 2008;30(10):1115–1120. doi:10.1179/0161641215Z.000000000587
  • Halamoda Kenzaoui B, Chapuis Bernasconi C, Guney-Ayra S, et al. Induction of oxidative stress, lysosome activation and autophagy by nanoparticles in human brain-derived endothelial cells. Biochem J. 2012;441(3):813–821. doi:10.1042/BJ20111252
  • Fujioka K, Hanada S, Inoue Y, et al. Effects of silica and titanium oxide particles on a human neural stem cell line: morphology, mitochondrial activity, and gene expression of differentiation markers. Int J Mol Sci. 2014;15(7):11742–11759. doi:10.3390/ijms150711742
  • Ribeiro LW, Pietri M, Ardila-Osorio H, et al. Titanium dioxide and carbon black nanoparticles disrupt neuronal homeostasis via excessive activation of cellular prion protein signaling. Part Fibre Toxicol. 2022;19(1):48. doi:10.1186/s12989-022-00490-x
  • Heringa MB, Peters RJB, Bleys RLAW, et al. Detection of titanium particles in human liver and spleen and possible health implications. Part Fibre Toxicol. 2018;15(1):15. doi:10.1186/s12989-018-0251-7
  • Wolff RK. Toxicology studies for inhaled and nasal delivery. Mol Pharm. 2015;12(8):2688–2696. doi:10.1021/acs.molpharmaceut.5b00146
  • Miller MA, Stabenow JM, Parvathareddy J, et al. Visualization of murine intranasal dosing efficiency using luminescent Francisella tularensis: effect of instillation volume and form of anesthesia. PLoS One. 2012;7(2):e31359. doi:10.1371/journal.pone.0031359
  • Southam DS, Dolovich M, O’Byrne PM, et al. Distribution of intranasal instillations in mice: effects of volume, time, body position, and anesthesia. Am J Physiol Lung Cell Mol Physiol. 2002;282(4):L833–L839. doi:10.1152/ajplung.00173.2001
  • Prust M, Meijer J, Westerink RHS. The plastic brain: neurotoxicity of micro- and nanoplastics. Part Fibre Toxicol. 2020;17(1):24. doi:10.1186/s12989-020-00358-y
  • Clogston JD, Patri AK. Zeta potential measurement. Methods Mol Biol. 2011;697:63–70. doi:10.1007/978-1-60327-198-1_6
  • Jin C, Tang Y, Yang FG, et al. Cellular toxicity of TiO2 nanoparticles in anatase and rutile crystal phase. Biol Trace Elem Res. 2011;141(1–3):3–15. doi:10.1007/s12011-010-8707-0
  • Abbott Chalew TEA, Ajmani GS, Huang H, et al. Evaluating nanoparticle breakthrough during drinking water treatment. Environ Health Perspect. 2013;121(10):1161–1166. doi:10.1289/ehp.1306574