Journal Home > Volume 12 , Issue 1

The recent advances in nanoscience and nanotechnology have enhanced the synthesis of various forms of nanomaterials for practical applications. Undoubtedly, these nanomaterials are not without attendant toxicities to humans, environment, and other organisms. Moreover, the toxicity of nanomaterials dominates the landscape of current toxicity concerns highlighted by the FDA. Titanium dioxide nanoparticles (TiO2 NPs) contribute a large proportion of synthesized nanomaterials mainly due to their excellent photocatalytic activities, mechanical and chemical stability, bio- and chemical inertness, corrosion resistance, thin film transparency, and low production cost. These fascinating properties of TiO2 NPs have been extensively exploited and dramatically increased their utility for various applications such as in nanomedicine for cancer theranostics, nanobiotechnology, environment, pharmacy, energy, food, cosmetics, and paper industries. Owing to the poor understanding of the impacts of NPs on humans, no clear regulation has been implemented for NPs among international authorities. Over time, the toxicity state of TiO2 NPs is typical of a double-edged sword. Hitherto, there is no restriction on the use of TiO2 NPs irrespective of the toxicity concerns raised by some researchers. This may have been dampened by the low-to-no toxicity reports from other researchers on these NPs. This review therefore looks into the recent toxicity reports from various studies conducted with/on TiO2 NPs as to ascertain their present-day safety. To elucidate this, we discussed the possible exposure routes to these NPs and their effects on the environment, plants, soil organisms, and aquatic species. We also provided insights on the toxicity mechanisms of TiO2 NPs and proposed future perspectives for improving their safe applications.


menu
Abstract
Full text
Outline
About this article

Revisiting an Old Friend Nano-TiO2: A Crucial Assessment of its Current Safety

Show Author's information Ozioma Udochukwu Akakuru1Muhammad Zubair Iqbal1,3( )Chuang Liu1,2Zihou Li1,2Yang Gao1,2Chen Xu1Elvis Ikechukwu Nosike1,2Gohar Ijaz Dar1,2Fang Yang1Aiguo Wu1( )
Cixi Institute of Biomedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices, & Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, China
University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing 100049, China
School of Materials Science and Engineering, Zhejiang Sci-Tech University, No. 2 Road of Xiasha, Hangzhou 310018, China

Abstract

The recent advances in nanoscience and nanotechnology have enhanced the synthesis of various forms of nanomaterials for practical applications. Undoubtedly, these nanomaterials are not without attendant toxicities to humans, environment, and other organisms. Moreover, the toxicity of nanomaterials dominates the landscape of current toxicity concerns highlighted by the FDA. Titanium dioxide nanoparticles (TiO2 NPs) contribute a large proportion of synthesized nanomaterials mainly due to their excellent photocatalytic activities, mechanical and chemical stability, bio- and chemical inertness, corrosion resistance, thin film transparency, and low production cost. These fascinating properties of TiO2 NPs have been extensively exploited and dramatically increased their utility for various applications such as in nanomedicine for cancer theranostics, nanobiotechnology, environment, pharmacy, energy, food, cosmetics, and paper industries. Owing to the poor understanding of the impacts of NPs on humans, no clear regulation has been implemented for NPs among international authorities. Over time, the toxicity state of TiO2 NPs is typical of a double-edged sword. Hitherto, there is no restriction on the use of TiO2 NPs irrespective of the toxicity concerns raised by some researchers. This may have been dampened by the low-to-no toxicity reports from other researchers on these NPs. This review therefore looks into the recent toxicity reports from various studies conducted with/on TiO2 NPs as to ascertain their present-day safety. To elucidate this, we discussed the possible exposure routes to these NPs and their effects on the environment, plants, soil organisms, and aquatic species. We also provided insights on the toxicity mechanisms of TiO2 NPs and proposed future perspectives for improving their safe applications.

Keywords: TiO2 nanoparticles, Exposure routes, Environmental fates, Toxicity mechanism

References(140)

[1]

Y.X.J. Wáng, J.M. Idée, A comprehensive literatures update of clinical researches of superparamagnetic resonance iron oxide nanoparticles for magnetic resonance imaging. Quant Imaging Med Surg, 2017, 7: 88-122.

[2]

Y.X.J. Wang, Current status of superparamagnetic iron oxide contrast agents for liver magnetic resonance imaging. World J Gastroenterol, 2015, 21: 13400-13402.

[3]

J.E. Karlsson, W. El-Saadi, M. Ali, et al., Mangafodipir as a cardioprotective adjunct to reperfusion therapy: A feasibility study in patients with ST-segment elevation myocardial infarction. European Heart Journal - Cardiovascular Pharmacotherapy, 2015, 1: 39-45.

[4]

B. Jovanovic, Critical review of public health regulations of titanium dioxide, a human food additive. Integrated Environmental Assessment and Management, 2015, 11: 10-20.

[5]

H. Shi, R. Magaye, V. Castranova, et al., Titanium dioxide nanoparticles: a review of current toxicological data. Particle and Fibre Toxicology, 2013, 10: 15.

[6]

I. Iavicoli, V. Leso, and A. Bergamaschi, Toxicological Effects of Titanium Dioxide Nanoparticles: A Review of In Vivo Studies. Journal of Nanomaterials, 2012, 2012: 36.

[7]

S.I.L. Gomes, C.P. Roca, F. von der Kammer, et al., Mechanisms of (photo) toxicity of TiO2 nanomaterials (NM103, NM104, NM105): using high-throughput gene expression in Enchytraeus crypticus. Nanoscale, 2018, 10: 21960-21970.

[8]

K. Kakinoki, K. Yamane, M. Igarashi, et al., Evaluation of Titanium Dioxide as a Pharmaceutical Excipient for Preformulation of a Photo-Labile Drug: Effect of Physicochemical Properties on the Photostability of Solid-State Nisoldipine. Chemical and Pharmaceutical Bulletin, 2005, 53: 811-815.

[9]

L.C. Pele, V. Thoree, S.F.A. Bruggraber, et al., Pharmaceutical/food grade titanium dioxide particles are absorbed into the bloodstream of human volunteers. Particle and fibre toxicology, 2015, 12: 26-26.

[10]

A. Weir, P. Westerhoff, L. Fabricius, et al., Titanium dioxide nanoparticles in food and personal care products. Environmental science & technology, 2012, 46: 2242-2250.

[11]

H.S. Bang, H.S. Bang, and Y.K. Lee, The functional TiO2-biodegradable plastic composite material produced by HVOF spraying process. Journal of nanoscience and nanotechnology, 2007, 7: 3830-3833.

[12]

A.M.A. Aljafery, Flexural Resistance and Impact Resistance of High-Impact Acrylic Resin with Addition of TiO3-Al2O2Nanoparticles. Nano Biomedicine and Engineering, 2018, 10: 40-45.

[13]

A.S. Jerabek, K.R. Wall, and C.D. Stallings, A practical application of reduced-copper antifouling paint in marine biological research. PeerJ, 2016, 4: e2213-e2213.

[14]

M. Simonin, A. Richaume, J.P. Guyonnet, et al., Titanium dioxide nanoparticles strongly impact soil microbial function by affecting archaeal nitrifiers. Scientific Reports, 2016, 6: 33643.

[15]

T.L.L. Carmo, P.R. Siqueira, V.C. Azevedo, et al., Overview of the toxic effects of titanium dioxide nanoparticles in blood, liver, muscles, and brain of a Neotropical detritivorous fish. Environmental toxicology, 2019.

[16]

Y.Y. Huang, H. Choi, Y. Kushida, et al., Broad-Spectrum Antimicrobial Effects of Photocatalysis Using Titanium Dioxide Nanoparticles Are Strongly Potentiated by Addition of Potassium Iodide. Antimicrobial Agents and Chemotherapy, 2016, 60: 5445.

[17]

M. Saeed, M.Z. Iqbal, W. Ren, et al., Controllable synthesis of Fe3O4 nanoflowers: enhanced imaging guided cancer therapy and comparison of photothermal efficiency with black-TiO2. Journal of Materials Chemistry B, 2018, 6: 3800-3810.

[18]

M. Saeed, M.Z. Iqbal, W. Ren, et al., Tunable fabrication of new theranostic Fe3O4-black TiO2 nanocomposites: dual wavelength stimulated synergistic imaging-guided phototherapy in cancer. Journal of Materials Chemistry B, 2019, 7: 210-223.

[19]

H. Su, X. Song, J. Li, et al., Biosafety evaluation of Janus Fe3O4-TiO2 nanoparticles in Sprague Dawley rats after intravenous injection. Int J Nanomedicine, 2018, 13: 6987-7001.

[20]

W. Ren, Y. Yan, L. Zeng, et al., A Near Infrared Light Triggered Hydrogenated Black TiO2 for Cancer Photothermal Therapy. Advanced Healthcare Materials, 2015, 4: 1526-1536.

[21]

X. Zhang, B. Pan, K. Wang, et al., Electrochemical Property and Cell Toxicity of Gold Electrode Modified by Monolayer PAMAM Encapsulated Gold Nanorods. Nano Biomedicine and Engineering, 2010, 2: 182-188.

[22]

X. Dai, D. Cui, Advances in the Toxicity of Nanomaterials. Nano Biomedicine and Engineering, 2012, 4: 150-156.

[23]

H. Liu, L. Ma, J. Zhao, et al., Biochemical toxicity of nano-anatase TiO2 particles in mice. Biological trace element research, 2009, 129: 170-180.

[24]

D. Wang, L. Zhao, H. Ma, et al., Quantitative Analysis of Reactive Oxygen Species Photogenerated on Metal Oxide Nanoparticles and Their Bacteria Toxicity: The Role of Superoxide Radicals. Environmental Science & Technology, 2017, 51: 10137-10145.

[25]

M. Skocaj, M. Filipic, J. Petkovic, et al., Titanium dioxide in our everyday life; is it safe?, Radiology and oncology, 2011, 45: 227-247.

[26]

G. Gao, Y. Ze, X. Zhao, et al., Titanium dioxide nanoparticle-induced testicular damage, spermatogenesis suppression, and gene expression alterations in male mice. Journal of hazardous materials, 2013, 258-259: 133-143.

[27]

M. Kosmulski, The pH-dependent surface charging and points of zero charge: V. Update. Journal of Colloid and Interface Science, 2011, 353: 1-15.

[28]

J. Zhao, V. Castranova, Toxicology of nanomaterials used in nanomedicine. Journal of toxicology and environmental health. Part B, Critical reviews, 2011, 14: 593-632.

[29]

J. Xu, H. Shi, M. Ruth, et al., Acute Toxicity of Intravenously Administered Titanium Dioxide Nanoparticles in Mice. PLOS ONE, 2013, 8: e70618.

[30]

K. Yamashita, Y. Yoshioka, K. Higashisaka, et al., Silica and titanium dioxide nanoparticles cause pregnancy complications in mice. Nature nanotechnology, 2011, 6: 321-328.

[31]

K. Onuma, Y. Sato, S. Ogawara, et al., Nano-scaled particles of titanium dioxide convert benign mouse fibrosarcoma cells into aggressive tumor cells. The American journal of pathology, 2009, 175: 2171-2183.

[32]

B. van Ravenzwaay, R. Landsiedel, E. Fabian, et al., Comparing fate and effects of three particles of different surface properties: nano-TiO2, pigmentary TiO2 and quartz. Toxicology letters, 2009, 186: 152-159.

[33]

A.E. Gonzalez-Esquivel, C.L. Charles-Nino, F.P. Pacheco-Moises, et al., Beneficial effects of quercetin on oxidative stress in liver and kidney induced by titanium dioxide (TiO2) nanoparticles in rats, Toxicology mechanisms and methods, 25(2015): 166-175.

[34]

L. Geraets, A.G. Oomen, P. Krystek, et al., Tissue distribution and elimination after oral and intravenous administration of different titanium dioxide nanoparticles in rats. Particle and fibre toxicology, 2014, 11: 30.

[35]

M.Z. Iqbal, W. Ren, M. Saeed, et al., A facile fabrication route for binary transition metal oxide-based Janus nanoparticles for cancer theranostic applications. Nano Research, 2018, 11: 5735-5750.

[36]

G. Shu, H. Wang, H.X. Zhao, et al., Microwave-Assisted Synthesis of Black Titanium Monoxide for Synergistic Tumor Phototherapy. ACS Applied Materials & Interfaces, 2019, 11: 3323-3333.

[37]

J. Mou, T. Lin, F. Huang, et al., Black titania-based theranostic nanoplatform for single NIR laser induced dual-modal imaging-guided PTT/PDT. Biomaterials, 2016, 84: 13-24.

[38]

M. Wang, K. Deng, W. Lü, et al., Rational Design of Multifunctional Fe@γ-Fe2O3@H-TiO2 Nanocomposites with Enhanced Magnetic and Photoconversion Effects for Wide Applications: From Photocatalysis to Imaging-Guided Photothermal Cancer Therapy. Advanced Materials, 2018, 30: 1706747.

[39]

O.U. Akakuru, M.Z. Iqbal, M. Saeed, et al., The Transition from Metal-Based to Metal-Free Contrast Agents for T1 Magnetic Resonance Imaging Enhancement. Bioconjugate Chemistry, 2019, 30: 2264-2286.

[40]

F. Imanparast, M. Doosti, and M.A. Faramarzi, Metal Nanoparticles in Atherosclerosis: Applications and Potential Toxicity. Nano Biomedicine and Engineering, 2015, 7: 111-127.

[41]

A.M.C. Swart, M.K.B. Parmar, S. Burdett, et al., Why i.p. therapy cannot yet be considered as a standard of care for the first-line treatment of ovarian cancer: a systematic review. Annals of Oncology, 2007, 19: 688-695.

[42]

M. Shakeel, F. Jabeen, S. Shabbir, et al., Toxicity of Nano-Titanium Dioxide (TiO2-NP) Through Various Routes of Exposure: a Review. Biological trace element research, 2016, 172: 1-36.

[43]

X. Valentini, P. Rugira, A. Frau, et al., Hepatic and Renal Toxicity Induced by TiO2 Nanoparticles in Rats: A Morphological and Metabonomic Study. Journal of Toxicology, 2019, 2019: 19.

[44]

N.R. Younes, S. Amara, I. Mrad, et al., Subacute toxicity of titanium dioxide (TiO2) nanoparticles in male rats: emotional behavior and pathophysiological examination. Environmental science and pollution research international, 2015, 22: 8728-8737.

[45]

L.L. Guo, X.H. Liu, D.X. Qin, et al., Effects of nanosized titanium dioxide on the reproductive system of male mice. Zhonghua nan ke xue = National journal of andrology, 2009, 15: 517-522.

[46]

J. Chen, X. Dong, J. Zhao, et al., In vivo acute toxicity of titanium dioxide nanoparticles to mice after intraperitioneal injection. Journal of applied toxicology : JAT, 2009, 29: 330-337.

[47]

H. Kim, H. Park, and S.J. Lee, Effective method for drug injection into subcutaneous tissue. Scientific reports, 2017, 7: 9613-9613.

[48]

R.D. Brohi, L. Wang, H.S. Talpur, et al., Toxicity of Nanoparticles on the Reproductive System in Animal Models: A Review. Frontiers in pharmacology, 2017, 8: 606-606.

[49]

M. Umezawa, H. Tainaka, N. Kawashima, et al., Effect of fetal exposure to titanium dioxide nanoparticle on brain development - brain region information. The Journal of toxicological sciences, 2012, 37: 1247-1252.

[50]

M. Shimizu, H. Tainaka, T. Oba, et al., Maternal exposure to nanoparticulate titanium dioxide during the prenatal period alters gene expression related to brain development in the mouse. Particle and fibre toxicology, 2009, 6: 20.

[51]

Y. Cui, X. Chen, Z. Zhou, et al., Prenatal exposure to nanoparticulate titanium dioxide enhances depressive-like behaviors in adult rats. Chemosphere, 2014, 96: 99-104.

[52]

Y. Takahashi, K. Mizuo, Y. Shinkai, et al., Prenatal exposure to titanium dioxide nanoparticles increases dopamine levels in the prefrontal cortex and neostriatum of mice. The Journal of toxicological sciences, 2010, 35: 749-756.

[53]

L.K. Braydich-Stolle, N.M. Schaeublin, R.C. Murdock, et al., Crystal structure mediates mode of cell death in TiO2 nanotoxicity. Journal of Nanoparticle Research, 2009, 11: 1361-1374.

[54]

F. Hong, X. Yu, N. Wu, et al., Progress of in vivo studies on the systemic toxicities induced by titanium dioxide nanoparticles. Toxicology Research, 2017, 6: 115-133.

[55]

J. Schulz, H. Hohenberg, F. Pflucker, et al., Distribution of sunscreens on skin. Advanced drug delivery reviews, 2002, 54 Suppl 1: S157-163.

[56]

A.O. Gamer, E. Leibold, and B. van Ravenzwaay, The in vitro absorption of microfine zinc oxide and titanium dioxide through porcine skin. Toxicology in vitro : an international journal published in association with BIBRA, 2006, 20: 301-307.

[57]

N. Sadrieh, A.M. Wokovich, N.V. Gopee, et al., Lack of significant dermal penetration of titanium dioxide from sunscreen formulations containing nano- and submicron-size TiO2 particles, Toxicological sciences: an official journal of the Society of Toxicology, 115(2010): 156-166.

[58]

C. Bennat, C.C. Muller-Goymann, Skin penetration and stabilization of formulations containing microfine titanium dioxide as physical UV filter. International journal of cosmetic science, 2000, 22: 271-283.

[59]

J.G. Rouse, J. Yang, J.P. Ryman-Rasmussen, et al., Effects of Mechanical Flexion on the Penetration of Fullerene Amino Acid-Derivatized Peptide Nanoparticles through Skin. Nano Letters, 2007, 7: 155-160.

[60]

J.P. Ryman-Rasmussen, J.E. Riviere, and N.A. Monteiro-Riviere, Penetration of intact skin by quantum dots with diverse physicochemical properties. Toxicological sciences : an official journal of the Society of Toxicology, 2006, 91: 159-165.

[61]

J. Unnithan, M.U. Rehman, F.J. Ahmad, et al., Aqueous synthesis and concentration-dependent dermal toxicity of TiO2 nanoparticles in Wistar rats. Biological trace element research, 2011, 143: 1682-1694.

[62]

Y. Sagawa, M. Futakuchi, J. Xu, et al., Lack of promoting effect of titanium dioxide particles on chemically-induced skin carcinogenesis in rats and mice. The Journal of toxicological sciences, 2012, 37: 317-327.

[63]

F. Furukawa, Y. Doi, M. Suguro, et al., Lack of skin carcinogenicity of topically applied titanium dioxide nanoparticles in the mouse. Food and chemical toxicology: an international journal published for the British Industrial Biological Research Association, 2011, 49: 744-749.

[64]

M. Crosera, A. Prodi, M. Mauro, et al., Titanium Dioxide Nanoparticle Penetration into the Skin and Effects on HaCaT Cells. Int J Environ Res Public Health, 2015, 12: 9282-9297.

[65]

J. Wu, W. Liu, C. Xue, et al., Toxicity and penetration of TiO2 nanoparticles in hairless mice and porcine skin after subchronic dermal exposure. Toxicology letters, 2009, 191: 1-8.

[66]

F.U. Rehman, C. Zhao, H. Jiang, et al., Biomedical applications of nano-titania in theranostics and photodynamic therapy. Biomaterials Science, 2016, 4: 40-54.

[67]

B. Feng, J. Weng, B.C. Yang, et al., Characterization of surface oxide films on titanium and adhesion of osteoblast. Biomaterials, 2003, 24: 4663-4670.

[68]

A. Tautzenberger, A. Kovtun, and A. Ignatius, Nanoparticles and their potential for application in bone. Int J Nanomedicine, 2012, 7: 4545-4557.

[69]

Y.T. Sul, Electrochemical growth behavior, surface properties, and enhanced in vivo bone response of TiO2 nanotubes on microstructured surfaces of blasted, screw-shaped titanium implants. Int J Nanomedicine, 2010, 5: 87-100.

[70]

A.R. Ribeiro, S. Gemini-Piperni, R. Travassos, et al., Trojan-Like Internalization of Anatase Titanium Dioxide Nanoparticles by Human Osteoblast Cells. Scientific Reports, 2016, 6: 23615.

[71]

L.M. Bjursten, L. Rasmusson, S. Oh, et al., Titanium dioxide nanotubes enhance bone bonding in vivo. Journal of biomedical materials research. Part A, 2010, 92: 1218-1224.

[72]

D. Cadosch, E. Chan, O.P. Gautschi, et al., Metal is not inert: role of metal ions released by biocorrosion in aseptic loosening--current concepts. Journal of biomedical materials research. Part A, 2009, 91: 1252-1262.

[73]

J.X. Wang, Y.B. Fan, Y. Gao, et al., TiO2 nanoparticles translocation and potential toxicological effect in rats after intraarticular injection. Biomaterials, 2009, 30: 4590-4600.

[74]

M.B. Heringa, L. Geraets, J.C.H. van Eijkeren, et al., Risk assessment of titanium dioxide nanoparticles via oral exposure, including toxicokinetic considerations. Nanotoxicology, 2016, 10: 1515-1525.

[75]

J.F. Hillyer, R.M. Albrecht, Gastrointestinal persorption and tissue distribution of differently sized colloidal gold nanoparticles. Journal of pharmaceutical sciences, 2001, 90: 1927-1936.

[76]

S. Vanden-Hehir, W.J. Tipping, M. Lee, et al., Raman Imaging of Nanocarriers for Drug Delivery. Nanomaterials (Basel), 2019, 9.

[77]

O.U. Akakuru, H. Louis, R. Uwaoma, et al., Novel highly-swellable and pH-responsive slow release formulations of clotrimazole with chitosan-g-PEG/starch microparticles. Reactive and Functional Polymers, 2019, 135: 32-43.

[78]

O.U. Akakuru, H. Louis, O.C. Akakuru, et al., Facile fabrication of pH-responsive and swellable slow release microparticles of chlorpheniramine maleate with chitosan-starch matrices and their crosslinks. International Journal of Polymeric Materials and Polymeric Biomaterials, 2019: 1-15.

[79]

Y. Duan, J. Liu, L. Ma, et al., Toxicological characteristics of nanoparticulate anatase titanium dioxide in mice Biomaterials, 2010, 31: 894-899.

[80]

R. Tassinari, F. Cubadda, G. Moracci, et al., Oral, short-term exposure to titanium dioxide nanoparticles in Sprague-Dawley rat: focus on reproductive and endocrine systems and spleen Nanotoxicology, 2014, 8: 654-662.

[81]

L.P. Sycheva, V.S. Zhurkov, V.V. Iurchenko, et al., Investigation of genotoxic and cytotoxic effects of micro- and nanosized titanium dioxide in six organs of mice in vivo. Mutation research, 2011, 726: 8-14.

[82]

H. Chen, R. Zhao, B. Wang, et al., The effects of orally administered Ag, TiO2 and SiO2 nanoparticles on gut microbiota composition and colitis induction in mice. NanoImpact, 2017, 8: 80-88.

[83]

A. MacNicoll, M. Kelly, H. Aksoy, et al., A study of the uptake and biodistribution of nano-titanium dioxide using in vitro and in vivo models of oral intake. Journal of Nanoparticle Research, 2015, 17.

[84]

W. Dudefoi, K. Moniz, E. Allen-Vercoe, et al., Impact of food grade and nano-TiO2 particles on a human intestinal community. Food and Chemical Toxicology, 2017, 106: 242-249.

[85]

W.S. Cho, B.C. Kang, J.K. Lee, et al., Comparative absorption, distribution, and excretion of titanium dioxide and zinc oxide nanoparticles after repeated oral administration. Particle and fibre toxicology, 2013, 10: 9.

[86]

H.C. Winkler, T. Notter, U. Meyer, et al., Critical review of the safety assessment of titanium dioxide additives in food. Journal of nanobiotechnology, 2018, 16: 51.

[87]

K. Sanders, L.L. Degn, W.R. Mundy, et al., In vitro phototoxicity and hazard identification of nano-scale titanium dioxide. Toxicol Appl Pharmacol, 2012, 258: 226-236.

[88]

S.N.A. Shah, Z. Shah, M. Hussain, et al., Hazardous Effects of Titanium Dioxide Nanoparticles in Ecosystem. Bioinorganic Chemistry and Applications, 2017, 2017: 12.

[89]

E.J. Park, J. Yoon, K. Choi, et al., Induction of chronic inflammation in mice treated with titanium dioxide nanoparticles by intratracheal instillation. Toxicology, 2009, 260: 37-46.

[90]

D. Zheng, N. Wang, X. Wang, et al., Effects of the interaction of TiO2 nanoparticles with bisphenol A on their physicochemical properties and in vitro toxicity. Journal of hazardous materials, 2012, 199-200: 426-432.

[91]

C. Mühlfeld, M. Geiser, N. Kapp, et al., Re-evaluation of pulmonary titanium dioxide nanoparticle distribution using the "relative deposition index": Evidence for clearance through microvasculature. Particle and fibre toxicology, 2007, 4: 7.

[92]

M. Geiser, W.G. Kreyling, Deposition and biokinetics of inhaled nanoparticles. Particle and fibre toxicology, 2010, 7: 2.

[93]

A. Nemmar, P.H. Hoet, B. Vanquickenborne, et al., Passage of inhaled particles into the blood circulation in humans. Circulation, 2002, 105: 411-414.

[94]

M. Husain, D. Wu, A.T. Saber, et al., Intratracheally instilled titanium dioxide nanoparticles translocate to heart and liver and activate complement cascade in the heart of C57BL/6 mice. Nanotoxicology, 2015, 9: 1013-1022.

[95]

W.G. Kreyling, U. Holzwarth, N. Haberl, et al., Quantitative biokinetics of titanium dioxide nanoparticles after intratracheal instillation in rats: Part 3. Nanotoxicology, 2017, 11: 454-464.

[96]

B.L. Baisch, N.M. Corson, P. Wade-Mercer, et al., Equivalent titanium dioxide nanoparticle deposition by intratracheal instillation and whole body inhalation: the effect of dose rate on acute respiratory tract inflammation. Particle and fibre toxicology, 2014, 11: 5.

[97]

H. Li, Y. Zhang, and W. Huang, Novel Selective Sensors Based on TiO2 Nanotubes Supported MS(TiO2@MS, M=Cd, Zn) for Their Gas Sensing Properties. Nano Biomedicine and Engineering, 2010, 2: 143-148.

[98]

J. Carbajo, A. Bahamonde, and M. Faraldos, Photocatalyst performance in wastewater treatment applications: Towards the role of TiO2 properties. Molecular Catalysis, 2017, 434: 167-174.

[99]

M.M. Higarashi, W.F. Jardim, Remediation of pesticide contaminated soil using TiO2 mediated by solar light. Catalysis Today, 2002, 76: 201-207.

[100]

Nasikhudin, M. Diantoro, A. Kusumaatmaja, et al., Study on Photocatalytic Properties of TiO2 Nanoparticle in various pH condition. Journal of Physics: Conference Series, 2018, 1011: 012069.

[101]

K. Dai, T. Peng, H. Chen, et al., Photocatalytic Degradation and Mineralization of Commercial Methamidophos in Aqueous Titania Suspension. Environmental Science & Technology, 2008, 42: 1505-1510.

[102]

I.K. Konstantinou, T.M. Sakellarides, V.A. Sakkas, et al., Photocatalytic Degradation of Selected s-Triazine Herbicides and Organophosphorus Insecticides over Aqueous TiO2 Suspensions. Environmental Science & Technology, 2001, 35: 398-405.

[103]

S. Parra, J. Olivero, and C. Pulgarin, Relationships between physicochemical properties and photoreactivity of four biorecalcitrant phenylurea herbicides in aqueous TiO2 suspension. Applied Catalysis B: Environmental, 2002, 36: 75-85.

[104]

A.P. Gondikas, F. von der Kammer, R.B. Reed, et al., Release of TiO2 nanoparticles from sunscreens into surface waters: a one-year survey at the old Danube recreational Lake. Environ Sci Technol, 2014, 48: 5415-5422.

[105]

C. Larue, H. Castillo-Michel, S. Sobanska, et al., Fate of pristine TiO2 nanoparticles and aged paint-containing TiO2 nanoparticles in lettuce crop after foliar exposure. Journal of hazardous materials, 2014, 273: 17-26.

[106]

M. Ghosh, M. Bandyopadhyay, and A. Mukherjee, Genotoxicity of titanium dioxide (TiO2) nanoparticles at two trophic levels: plant and human lymphocytes. Chemosphere, 2010, 81: 1253-1262.

[107]

C.W. Hu, M. Li, Y.B. Cui, et al., Toxicological effects of TiO2 and ZnO nanoparticles in soil on earthworm Eisenia fetida. Soil Biology & Biochemistry, 2010, 42: 586-591.

[108]

A. Burton, Titanium dioxide photocleans polluted air. Environmental health perspectives, 2012, 120: A229-A229.

[109]

P.G. Sayre, K.G. Steinhäuser, and T. van Teunenbroek, Methods and data for regulatory risk assessment of nanomaterials: Questions for an expert consultation. NanoImpact, 2017, 8: 20-27.

[110]

L. Li, M. Sillanpaa, and M. Risto, Influences of water properties on the aggregation and deposition of engineered titanium dioxide nanoparticles in natural waters. Environmental pollution (Barking, Essex : 1987), 2016, 219: 132-138.

[111]

X. Shi, Z. Li, W. Chen, et al., Fate of TiO2 nanoparticles entering sewage treatment plants and bioaccumulation in fish in the receiving streams. NanoImpact, 2016, 3-4: 96-103.

[112]

G. Federici, B.J. Shaw, and R.D. Handy, Toxicity of titanium dioxide nanoparticles to rainbow trout (Oncorhynchus mykiss): Gill injury, oxidative stress, and other physiological effects. Aquatic Toxicology, 2007, 84: 415-430.

[113]

T.L.L. do Carmo, V.C. Azevedo, P.R. de Siqueira, et al., Reactive oxygen species and other biochemical and morphological biomarkers in the gills and kidneys of the Neotropical freshwater fish, Prochilodus lineatus, exposed to titanium dioxide (TiO2) nanoparticles. Environmental Science and Pollution Research, 2018, 25: 22963-22976.

[114]

L. Hao, Z. Wang, and B. Xing, Effect of sub-acute exposure to TiO2 nanoparticles on oxidative stress and histopathological changes in Juvenile Carp (Cyprinus carpio). J Environ Sci (China), 2009, 21: 1459-1466.

[115]

C.P. Vignardi, F.M. Hasue, P.V. Sartorio, et al., Genotoxicity, potential cytotoxicity and cell uptake of titanium dioxide nanoparticles in the marine fish Trachinotus carolinus (Linnaeus, 1766). Aquatic toxicology (Amsterdam, Netherlands), 2015, 158: 218-229.

[116]

Q. Fang, Q. Shi, Y. Guo, et al., Enhanced Bioconcentration of Bisphenol A in the Presence of Nano-TiO2 Can Lead to Adverse Reproductive Outcomes in Zebrafish. Environ Sci Technol, 2016, 50: 1005-1013.

[117]

J. Hou, L. Wang, C. Wang, et al., Toxicity and mechanisms of action of titanium dioxide nanoparticles in living organisms. Journal of environmental sciences (China), 2019, 75: 40-53.

[118]

Y. Wang, X. Zhu, Y. Lao, et al., TiO2 nanoparticles in the marine environment: Physical effects responsible for the toxicity on algae Phaeodactylum tricornutum. The Science of the total environment, 2016, 565: 818-826.

[119]

M. Sendra, D. Sanchez-Quiles, J. Blasco, et al., Effects of TiO2 nanoparticles and sunscreens on coastal marine microalgae: Ultraviolet radiation is key variable for toxicity assessment. Environment international, 2017, 98: 62-68.

[120]
Szymańska R. Kołodziej K. Ślesak I. Titanium dioxide nanoparticles (100–1000 mg/l) can affect vitamin E response in Arabidopsis thalianaEnvironmental Pollution2016213957965 10.1016/j.envpol.2016.03.026

R. Szymańska, K. Kołodziej, I. Ślesak, et al., Titanium dioxide nanoparticles (100–1000 mg/l) can affect vitamin E response in Arabidopsis thaliana. Environmental Pollution, 2016, 213: 957-965.

[121]

Z.G. Doğaroğlu, N. Köleli, TiO2 and ZnO Nanoparticles Toxicity in Barley (Hordeum vulgare L.). CLEAN – Soil, Air, Water, 2017, 45: 1700096.

[122]

E. Topuz, L. Sigg, and I. Talinli, A systematic evaluation of agglomeration of Ag and TiO2 nanoparticles under freshwater relevant conditions. Environmental pollution (Barking, Essex : 1987), 2014, 193: 37-44.

[123]

B. Jovanović, Review of titanium dioxide nanoparticle phototoxicity: Developing a phototoxicity ratio to correct the endpoint values of toxicity tests. Environmental toxicology and chemistry, 2015, 34: 1070-1077.

[124]

K. Schlich, K. Hund-Rinke, Influence of soil properties on the effect of silver nanomaterials on microbial activity in five soils. Environmental Pollution, 2015, 196: 321-330.

[125]

A. Bermejo-Nogales, M. Connolly, P. Rosenkranz, et al., Negligible cytotoxicity induced by different titanium dioxide nanoparticles in fish cell lines. Ecotoxicol Environ Saf, 2017, 138: 309-319.

[126]

B.P. Lankadurai, E.G. Nagato, and M.J. Simpson, Environmental metabolomics: an emerging approach to study organism responses to environmental stressors. Environmental Reviews, 2013, 21: 180-205.

[127]

Y. Du, W. Ren, Y. Li, et al., The enhanced chemotherapeutic effects of doxorubicin loaded PEG coated TiO2 nanocarriers in an orthotopic breast tumor bearing mouse model. Journal of Materials Chemistry B, 2015, 3: 1518-1528.

[128]

S. Wang, W. Ren, J. Wang, et al., Black TiO2-based nanoprobes for T1-weighted MRI-guided photothermal therapy in CD133 high expressed pancreatic cancer stem-like cells. Biomaterials Science, 2018, 6: 2209-2218.

[129]

S. Senthilkumar, A. Rajendran, Biosynthesis of TiO2 nanoparticles using Justicia gendarussa leaves for photocatalytic and toxicity studies. Research on Chemical Intermediates, 2018, 44: 5923-5940.

[130]

X.Y. Deng, J. Cheng, X.L. Hu, et al., Biological effects of TiO2 and CeO2 nanoparticles on the growth, photosynthetic activity, and cellular components of a marine diatom Phaeodactylum tricornutum. The Science of the total environment, 2017, 575: 87-96.

[131]

F. Li, Z. Liang, X. Zheng, et al., Toxicity of nano-TiO2 on algae and the site of reactive oxygen species production. Aquatic toxicology (Amsterdam, Netherlands), 2015, 158: 1-13.

[132]

J. Chen, X. Dong, Y. Xin, et al., Effects of titanium dioxide nano-particles on growth and some histological parameters of zebrafish (Danio rerio) after a long-term exposure. Aquatic Toxicology, 2011, 101: 493-499.

[133]

X. Jia, S. Wang, L. Zhou, et al., The Potential Liver, Brain, and Embryo Toxicity of Titanium Dioxide Nanoparticles on Mice. Nanoscale Research Letters, 2017, 12: 478.

[134]

R. Sadiq, J.A. Bhalli, J. Yan, et al., Genotoxicity of TiO2 anatase nanoparticles in B6C3F1 male mice evaluated using Pig-a and flow cytometric micronucleus assays. Mutation Research/Genetic Toxicology and Environmental Mutagenesis, 2012, 745: 65-72.

[135]

M.M. Dobrzyńska, A. Gajowik, J. Radzikowska, et al., Genotoxicity of silver and titanium dioxide nanoparticles in bone marrow cells of rats in vivo. Toxicology, 2014, 315: 86-91.

[136]

A. A. El-Ghor, M.M. Noshy, A. Galal, et al., Normalization of Nano-Sized TiO2-Induced Clastogenicity, Genotoxicity and Mutagenicity by Chlorophyllin Administration in Mice Brain, Liver, and Bone Marrow Cells. Toxicological Sciences, 2014, 142: 21-32.

[137]

K.E. Driscoll, L.C. Deyo, J.M. Carter, et al., Effects of particle exposure and particle-elicited inflammatory cells on mutation in rat alveolar epithelial cells. Carcinogenesis, 1997, 18: 423-430.

[138]

R. Liu, X. Zhang, Y. Pu, et al., Small-sized titanium dioxide nanoparticles mediate immune toxicity in rat pulmonary alveolar macrophages in vivo. Journal of nanoscience and nanotechnology, 2010, 10: 5161-5169.

[139]

S. Bettini, E. Boutet-Robinet, C. Cartier, et al., Food-grade TiO2 impairs intestinal and systemic immune homeostasis, initiates preneoplastic lesions and promotes aberrant crypt development in the rat colon. Scientific Reports, 2017, 7: 40373.

[140]

Y. Fu, Y. Zhang, X. Chang, et al., Systemic immune effects of titanium dioxide nanoparticles after repeated intratracheal instillation in rat. International journal of molecular sciences, 2014, 15: 6961-6973.

Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Received: 26 October 2019
Accepted: 07 February 2020
Published: 14 February 2020
Issue date: March 2020

Copyright

© Ozioma Udochukwu Akakuru, Muhammad Zubair Iqbal, Chuang Liu, Zihou Li, Yang Gao, Chen Xu, Elvis Ikechukwu Nosike, Gohar Ijaz Dar, Fang Yang, and Aiguo Wu.

Acknowledgements

Acknowledgements

The authors acknowledge the support of National Key R&D Program of China (2018YFC0910601, and 2019YFA04000803), National Natural Science Foundation of China (31971292, U1432114, 81650410654, 8161101589), the Hundred Talents Program of Chinese Academy of Sciences (2010-735), Key Breakthrough Program of Chinese Academy of Sciences (KGZD-EW-T06), Special Program for Applied Research on Super Computation of the NSFC-Guangdong Joint Fund (the second phase to Aiguo Wu, U1501501), and the Science & Technology Bureau of Ningbo City (2015B11002). Ozioma Udochukwu Akakuru also appreciates the Chinese Academy of Sciences (CAS) and The World Academy of Sciences (TWAS) for the award of the CAS-TWAS President's Fellowship (2017A8017422001) for PhD studies.

Rights and permissions

This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Return