Skip to main content

Advertisement

Log in

Neurotoxicity of four frequently used nanoparticles: a systematic review to reveal the missing data

  • Review Article
  • Published:
Archives of Toxicology Aims and scope Submit manuscript

Abstract

Systemic exposure to nanoparticles (NPs) adversely affects different organs, including the nervous system. We systematically extracted data from publication on PubMed and Embase database up to the year 2020, and analyzed in vitro and in vivo neurotoxicity of 4 of the most well studied NPs (silver NPs, carbon-based NPs, iron NPs and silica NPs). A relatively good correlation was observed between in vitro and in vivo effects, including genotoxicity, oxidative stress, apoptosis and pro-inflammatory effects. However, crucial knowledge gap exists in current understanding of the underlying mechanisms. Some of the critical knowledge gaps and research needs identified in relation to neurotoxicity of nanoparticles include (1) lack of physio-chemical characteristics of NPs used, (2) cellular/tissue uptake of NP, (3) NP translocation across the blood-brain barrier (BBB), (4) Effect of exposure routes.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

References

  • Allen JL et al (2017) Developmental neurotoxicity of inhaled ambient ultrafine particle air pollution: parallels with neuropathological and behavioral features of autism and other neurodevelopmental disorders. Neurotoxicology 59:140–154

    Article  CAS  PubMed  Google Scholar 

  • Almeida JPM, Chen AL, Foster A, Drezek R (2011) In vivo biodistribution of nanoparticles. Nanomed 6:815–835

    Article  CAS  Google Scholar 

  • Antsiferova A, Kopaeva M, Kashkarov P (2018) Effects of prolonged silver nanoparticle exposure on the contextual cognition and behavior of mammals. Materials 11:558

    Article  CAS  PubMed Central  Google Scholar 

  • Badman RP et al (2020) Dextran-coated iron oxide nanoparticle-induced nanotoxicity in neuron cultures. Sci Rep 10:1–14

    Article  CAS  Google Scholar 

  • Bagchi S et al (2019) In-vitro blood-brain barrier models for drug screening and permeation studies: an overview. Drug Des Devel Ther 13:3591–3605

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Baruwati B, Simmons SO, Varma RS, Veronesi B (2013) ‘Green’ synthesized and coated nanosilver alters the membrane permeability of barrier (intestinal, brain endothelial) cells and stimulates oxidative stress pathways in neurons. ACS Sustain Chem Eng 1:753–759

    Article  CAS  Google Scholar 

  • Begum AN, Aguilar JS, Elias L, Hong Y (2016) Silver nanoparticles exhibit coating and dose-dependent neurotoxicity in glutamatergic neurons derived from human embryonic stem cells. Neurotoxicology 57:45–53

    Article  CAS  PubMed  Google Scholar 

  • Bergin IL, Witzmann FA (2013) Nanoparticle toxicity by the gastrointestinal route: evidence and knowledge gaps. Int J Biomed Nanosci Nanotechnol 3:163

    Article  CAS  Google Scholar 

  • Boche D, Perry VH, Nicoll JAR (2013) Review: Activation patterns of microglia and their identification in the human brain. Neuropathol Appl Neurobiol 39:3–18

    Article  CAS  PubMed  Google Scholar 

  • Bondarenko O, Juganson K, Ivask A et al (2013) Toxicity of Ag, CuO and ZnO nanoparticles to selected environmentally relevant test organisms and mammalian cells in vitro: a critical review. Arch Toxicol 87:1181–1200. https://doi.org/10.1007/s00204-013-1079-4

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Bussy C et al (2015) Microglia determine brain region-specific neurotoxic responses to chemically functionalized carbon nanotubes. ACS Nano 9:7815–7830

    Article  CAS  PubMed  Google Scholar 

  • Cannon JR, Greenamyre JT (2011) The role of environmental exposures in neurodegeneration and neurodegenerative diseases. Toxicol Sci 124:225–250

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Dąbrowska-Bouta B et al (2016) Influence of a low dose of silver nanoparticles on cerebral myelin and behavior of adult rats. Toxicology 363–364:29–36

    Article  CAS  PubMed  Google Scholar 

  • De Simone U, Caloni F, Gribaldo L, Coccini T (2017) Human co-culture model of neurons and astrocytes to test acute cytotoxicity of neurotoxic compounds. Int J Toxicol 36:463–477

    Article  CAS  PubMed  Google Scholar 

  • De Simone U et al (2018) Human 3D cultures as models for evaluating magnetic nanoparticle CNS cytotoxicity after short- and repeated long-term exposure. Int J Mol Sci 19:1993

    Article  CAS  PubMed Central  Google Scholar 

  • Dong L et al (2019) Shape-dependent toxicity of alumina nanoparticles in rat astrocytes. Sci Total Environ 690:158–166

    Article  CAS  PubMed  Google Scholar 

  • Du Q et al (2019) Assessment of neurotoxicity induced by different-sized Stöber silica nanoparticles: induction of pyroptosis in microglia. Nanoscale 11:12965–12972

    Article  CAS  PubMed  Google Scholar 

  • Fatemi M, Hayati-Roodbari N, Ghaedi K, Naderi G (2013) The effects of prenatal exposure to silver nanoparticles on the developing brain in neonatal rats. J Biol Res Greece 20:233–242

    Google Scholar 

  • Feng X et al (2018) Graphene oxide induces p62/SQSTM-dependent apoptosis through the impairment of autophagic flux and lysosomal dysfunction in PC12 cells. Acta Biomater 81:278–292

    Article  CAS  PubMed  Google Scholar 

  • Fernández-Bertólez N et al (2018) Neurotoxicity assessment of oleic acid-coated iron oxide nanoparticles in SH-SY5Y cells. Toxicology 406–407:81–91

    Article  CAS  PubMed  Google Scholar 

  • Fernández-Bertólez N et al (2019) Evaluation of cytotoxicity and genotoxicity induced by oleic acid-coated iron oxide nanoparticles in human astrocytes. Environ Mol Mutagen. https://doi.org/10.1002/em.22323

    Article  PubMed  Google Scholar 

  • Fu J et al (2018) Silica nanoparticle exposure during the neonatal period impairs hippocampal precursor proliferation and social behavior later in life. Int J Nanomedicine 13:3593–3608

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Ganjuri M, Moshtaghian J, Ghaedi K (2015) Effect of nanosilver particles on procaspase-3 expression in newborn rat brain. Cell J 17:489–493

    PubMed  PubMed Central  Google Scholar 

  • Gao J, Zhang X, Yu M, Ren G, Yang Z (2015) Cognitive deficits induced by multi-walled carbon nanotubes via the autophagic pathway. Toxicology 337:21–29

    Article  CAS  PubMed  Google Scholar 

  • Genc S, Zadeoglulari Z, Fuss SH, Genc K (2012) The adverse effects of air pollution on the nervous system. J Toxicol. 2012:e782462 https://www.hindawi.com/journals/jt/2012/782462/

  • Ghooshchian M, Khodarahmi P, Tafvizi F (2017) Apoptosis-mediated neurotoxicity and altered gene expression induced by silver nanoparticles. Toxicol Ind Health 33:757–764

    Article  CAS  PubMed  Google Scholar 

  • Gonzalez-Carter DA et al (2017) Silver nanoparticles reduce brain inflammation and related neurotoxicity through induction of H2S-synthesizing enzymes. Sci Rep 7:42871

    Article  PubMed  PubMed Central  Google Scholar 

  • Haghani A et al (2020) Toxicity of urban air pollution particulate matter in developing and adult mouse brain: comparison of total and filter-eluted nanoparticles. Environ Int 136:105510

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Hanada S et al (2014) Cell-based in vitro blood-brain barrier model can rapidly evaluate nanoparticles’ brain permeability in association with particle size and surface modification. Int J Mol Sci 15:1812–1825

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Huang H et al (2019) AMPK mediates the neurotoxicity of iron oxide nanoparticles retained in mitochondria or lysosomes. Metallomics 11:1200–1206

    Article  CAS  PubMed  Google Scholar 

  • Imam SZ et al (2015) Iron oxide nanoparticles induce dopaminergic damage: in vitro pathways and in vivo imaging reveals mechanism of neuronal damage. Mol Neurobiol 52:913–926

    Article  CAS  PubMed  Google Scholar 

  • Joris F et al (2017) Choose your cell model wisely: the in vitro nanoneurotoxicity of differentially coated iron oxide nanoparticles for neural cell labeling. Acta Biomater 55:204–213

    Article  CAS  PubMed  Google Scholar 

  • Kettler K, Veltman K, van de Meent D, van Wezel A, Hendriks AJ (2014) Cellular uptake of nanoparticles as determined by particle properties, experimental conditions, and cell type. Environ Toxicol Chem 33:481–492

    Article  CAS  PubMed  Google Scholar 

  • Khan AM et al (2019) Silver nanoparticle-induced expression of proteins related to oxidative stress and neurodegeneration in an in vitro human blood-brain barrier model. Nanotoxicology 13:221–239

    Article  CAS  PubMed  Google Scholar 

  • Kim S, Ryu D-Y (2013) Silver nanoparticle-induced oxidative stress, genotoxicity and apoptosis in cultured cells and animal tissues. J Appl Toxicol 33:78–89

    Article  CAS  PubMed  Google Scholar 

  • Kim Y-J, Yang SI (2011) Neurotoxic effects by silica TM nanoparticle is independent of differentiation of SH-SY5Y cells. Mol Cell Toxicol 7:381–388

    Article  CAS  Google Scholar 

  • Kim Y et al (2013) In vivo nanoneurotoxicity screening using oxidative stress and neuroinflammation paradigms. Nanomed Nanotechnol Biol Med 9:1057–1066

    Article  CAS  Google Scholar 

  • Lee H-Y et al (2010) Genomics-based screening of differentially expressed genes in the brains of mice exposed to silver nanoparticles via inhalation. J Nanopart Res 12:1567–1578

    Article  CAS  Google Scholar 

  • Liu F et al (2015) Effects of silver nanoparticles on human and rat embryonic neural stem cells. Front Neurosci 9:00115

    Article  Google Scholar 

  • Liu X, Sui B, Sun J (2017) Blood-brain barrier dysfunction induced by silica NPs in vitro and in vivo: involvement of oxidative stress and Rho-kinase/JNK signaling pathways. Biomaterials 121:64–82

    Article  CAS  PubMed  Google Scholar 

  • Liu Y et al (2018) Characterization of superparamagnetic iron oxide nanoparticle-induced apoptosis in PC12 cells and mouse hippocampus and striatum. Toxicol Lett 292:151–161

    Article  CAS  PubMed  Google Scholar 

  • Ma W, Gehret PM, Hoff RE, Kelly LP, Suh WH (2019) The investigation into the toxic potential of iron oxide nanoparticles utilizing rat pheochromocytoma and human neural stem cells. Nanomaterials 9:453

    Article  CAS  PubMed Central  Google Scholar 

  • Manickam V, Dhakshinamoorthy V, Perumal E (2018) Iron oxide nanoparticles induces cell cycle-dependent neuronal apoptosis in mice. J Mol Neurosci 64:352–362

    Article  CAS  PubMed  Google Scholar 

  • Manickam V, Dhakshinamoorthy V, Perumal E (2019) Iron oxide nanoparticles affects behaviour and monoamine levels in mice. Neurochem Res. https://doi.org/10.1007/s11064-019-02774-9

    Article  PubMed  Google Scholar 

  • Meng L et al (2013) Inhibitory effects of multiwall carbon nanotubes with high iron impurity on viability and neuronal differentiation in cultured PC12 cells. Toxicology 313:49–58

    Article  CAS  PubMed  Google Scholar 

  • Missaoui WN, Arnold RD, Cummings BS (2018) Toxicological status of nanoparticles: what we know and what we don’t know. Chem Biol Interact 295:1–12

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Morris-Schaffer K et al (2019) Effects of neonatal inhalation exposure to ultrafine carbon particles on pathology and behavioral outcomes in C57BL/6J mice. Part Fibre Toxicol 16:1–15

    Article  PubMed  PubMed Central  Google Scholar 

  • Murugadoss S et al (2017) Toxicology of silica nanoparticles: an update. Arch Toxicol 91:2967–3010

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Oberdörster G et al (2004) Translocation of inhaled ultrafine particles to the brain. Inhal Toxicol 16:437–445

    Article  CAS  PubMed  Google Scholar 

  • Oberdörster G, Oberdörster E, Oberdörster J (2005) Nanotoxicology: an emerging discipline evolving from studies of ultrafine particles. Environ Health Perspect 113:823–839

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Oberdörster G, Elder A, Rinderknecht A (2009) Nanoparticles and the brain: cause for concern? J Nanosci Nanotechnol 9:4996–5007

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Oh J-H et al (2016) Integrative analysis of genes and miRNA alterations in human embryonic stem cells-derived neural cells after exposure to silver nanoparticles. Toxicol Appl Pharmacol 299:8–23

    Article  CAS  PubMed  Google Scholar 

  • Onoda A, Takeda K, Umezawa M (2017) Dose-dependent induction of astrocyte activation and reactive astrogliosis in mouse brain following maternal exposure to carbon black nanoparticle. Part Fibre Toxicol 14:1–16

    Article  CAS  Google Scholar 

  • Petters C, Dringen R (2015) Accumulation of iron oxide nanoparticles by cultured primary neurons. Neurochem Int 81:1–9

    Article  CAS  PubMed  Google Scholar 

  • Phillips CL et al (2013) The response effect of pheochromocytoma (PC12) cell lines to oxidized multi-walled carbon nanotubes (o-MWCMTs). Afr Health Sci 13:947–954

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Pulgar VM (2019) Transcytosis to cross the blood brain barrier. New advancements and challenges. Front Neurosci 12:01019

    Article  Google Scholar 

  • Raftis JB, Miller MR (2019) Nanoparticle translocation and multi-organ toxicity: a particularly small problem. Nano Today 26:8–12

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Rosas-Hernandez H et al (2019) Cytotoxicity profile of pristine graphene on brain microvascular endothelial cells. J Appl Toxicol 39:966–973

    Article  CAS  PubMed  Google Scholar 

  • Repar N, Li H, Aguilar JS et al (2018) Silver nanoparticles induce neurotoxicity in a human embryonic stem cell-derived neuron and astrocyte network. Nanotoxicology 12:104–116. https://doi.org/10.1080/17435390.2018.1425497

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Salatin S, Dizaj SM, Khosroushahi AY (2015) Effect of the surface modification, size, and shape on cellular uptake of nanoparticles. Cell Biol Int 39:881–890

    Article  CAS  PubMed  Google Scholar 

  • Sankowski R, Mader S, Valdés-Ferrer SI (2015) Systemic inflammation and the brain: novel roles of genetic, molecular, and environmental cues as drivers of neurodegeneration. Front Cell Neurosci 9:28

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Sawicki K et al (2019) Toxicity of metallic nanoparticles in the central nervous system. Nanotechnol Rev 8:175–200

    Article  CAS  Google Scholar 

  • SCoEaNIHR ES (2010) Scientific basis for the definition of the term “nanomaterial. Eur Commun Sci Comm Emerg New. Identified Health Risks SCENHR

  • Sharma HS, Ali SF, Hussain SM, Schlager JJ, Sharma A (2009) Influence of engineered nanoparticles from metals on the blood-brain barrier permeability, cerebral blood flow, brain edema and neurotoxicity. An experimental study in the rat and mice using biochemical and morphological approaches. J Nanosci Nanotechnol 9:5055–5072

    Article  CAS  PubMed  Google Scholar 

  • Sheida E et al (2017) The effect of iron nanoparticles on performance of cognitive tasks in rats. Environ Sci Pollut Res Int 24:8700–8710

    Article  CAS  PubMed  Google Scholar 

  • Skalska J, Frontczak-Baniewicz M, Struzyńska L (2015) Synaptic degeneration in rat brain after prolonged oral exposure to silver nanoparticles. Neurotoxicology 46:145–154

    Article  CAS  PubMed  Google Scholar 

  • Skalska J, Dąbrowska-Bouta B, Strużyńska L (2016) Oxidative stress in rat brain but not in liver following oral administration of a low dose of nanoparticulate silver. Food Chem Toxicol 97:307–315

    Article  CAS  PubMed  Google Scholar 

  • Stone V et al (2017) Nanomaterials versus ambient ultrafine particles: an opportunity to exchange toxicology knowledge. Environ Health Perspect 125:106002

    Article  PubMed  PubMed Central  Google Scholar 

  • Sun B, Liu R, Ye N, Xiao Z-D (2015) Comprehensive evaluation of microrna expression profiling reveals the neural signaling specific cytotoxicity of superparamagnetic iron oxide nanoparticles (SPIONs) through N-methyl-d-aspartate Receptor. PLoS ONE 10:e0121671

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Sun C et al (2016) Silver nanoparticles induced neurotoxicity through oxidative stress in rat cerebral astrocytes is distinct from the effects of silver ions. Neurotoxicology 52:210–221

    Article  CAS  PubMed  Google Scholar 

  • Sun D et al (2017) Evaluating the toxicity of silicon dioxide nanoparticles on neural stem cells using RNA-Seq. RSC Adv 7:47552–47564

    Article  CAS  Google Scholar 

  • Tabatabaei SRF, Moshrefi M, Askaripour M (2015) Prenatal exposure to silver nanoparticles causes depression like responses in mice. Indian J Pharm Sci 77:681–686

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Tang Y, Le W (2016) Differential roles of M1 and M2 microglia in neurodegenerative diseases. Mol Neurobiol 53:1181–1194

    Article  CAS  PubMed  Google Scholar 

  • Tang W et al (2019) Emerging blood–brain-barrier-crossing nanotechnology for brain cancer theranostics. Chem Soc Rev 48:2967–3014

    Article  CAS  PubMed  Google Scholar 

  • Teleanu DM, Chircov C, Grumezescu AM, Volceanov A, Teleanu RI (2018) Impact of nanoparticles on brain health: an up to date overview. J Clin Med 7:490

    Article  PubMed Central  Google Scholar 

  • Trickler WJ et al (2010) Silver nanoparticle induced blood-brain barrier inflammation and increased permeability in primary rat brain microvessel endothelial cells. Toxicol Sci 118:160–170

    Article  CAS  PubMed  Google Scholar 

  • van Berlo D et al (2014) Investigation of the effects of short-term inhalation of carbon nanoparticles on brains and lungs of c57bl/6j and p47(phox−/−) mice. Neurotoxicology 43:65–72

    Article  CAS  PubMed  Google Scholar 

  • Visalli G et al (2017) In vitro assessment of neurotoxicity and neuroinflammation of homemade MWCNTs. Environ Toxicol Pharmacol 56:121–128

    Article  CAS  PubMed  Google Scholar 

  • Wang B et al (2009) Neurotoxicity of low-dose repeatedly intranasal instillation of nano- and submicron-sized ferric oxide particles in mice. J Nanopart Res 11:41–53

    Article  CAS  Google Scholar 

  • Wang J, Sun P, Bao Y, Liu J, An L (2011) Cytotoxicity of single-walled carbon nanotubes on PC12 cells. Toxicol in Vitro 25:242–250

    Article  CAS  PubMed  Google Scholar 

  • Wang Y, Xiong L, Tang M (2017) Toxicity of inhaled particulate matter on the central nervous system: neuroinflammation, neuropsychological effects and neurodegenerative disease. J Appl Toxicol 37:644–667

    Article  CAS  PubMed  Google Scholar 

  • Wei H, Deng F, Chen Y et al (2014) Ultrafine carbon black induces glutamate and ATP release byactivating connexin and pannexin hemichannels in cultured astrocytes. Toxicology 323:32–41. https://doi.org/10.1016/j.tox.2014.06.005

    Article  CAS  PubMed  Google Scholar 

  • Weichenthal S et al (2020) Within-city spatial variations in ambient ultrafine particle concentrations and incident brain tumors in adults. Epidemiology 31:177–183

    Article  PubMed  PubMed Central  Google Scholar 

  • Weldon BA et al (2018) Using primary organotypic mouse midbrain cultures to examine developmental neurotoxicity of silver nanoparticles across two genetic strains. Toxicol Appl Pharmacol 354:215–224

    Article  CAS  PubMed  Google Scholar 

  • Wu J, Wang C, Sun J, Xue Y (2011) Neurotoxicity of silica nanoparticles: brain localization and dopaminergic neurons damage pathways. ACS Nano 5:4476–4489

    Article  CAS  PubMed  Google Scholar 

  • Wu J, Ding T, Sun J (2013) Neurotoxic potential of iron oxide nanoparticles in the rat brain striatum and hippocampus. Neurotoxicology 34:243–253

    Article  CAS  PubMed  Google Scholar 

  • Xie H, Wu J (2016) Silica nanoparticles induce alpha-synuclein induction and aggregation in PC12-cells. Chem Biol Interact 258:197–204

    Article  CAS  PubMed  Google Scholar 

  • Xu L et al (2015a) Neurotoxicity of silver nanoparticles in rat brain after intragastric exposure. J Nanosci Nanotechnol 15:4215–4223

    Article  CAS  PubMed  Google Scholar 

  • Xu L et al (2015b) Silver nanoparticles induce tight junction disruption and astrocyte neurotoxicity in a rat blood-brain barrier primary triple coculture model. Int J Nanomed 10:6105–6119

    CAS  Google Scholar 

  • Xue Y, Wu J, Sun J (2012) Four types of inorganic nanoparticles stimulate the inflammatory reaction in brain microglia and damage neurons in vitro. Toxicol Lett 214:91–98

    Article  CAS  PubMed  Google Scholar 

  • Yamada S, Yamazaki D, Kanda Y (2018) Silver nanoparticles inhibit neural induction in human induced pluripotent stem cells. Nanotoxicology 12:836–846

    Article  CAS  PubMed  Google Scholar 

  • Yamashita K et al (2010) Carbon nanotubes elicit DNA damage and inflammatory response relative to their size and shape. Inflammation 33:276–280

    Article  CAS  PubMed  Google Scholar 

  • Yamashita K et al (2011) Silica and titanium dioxide nanoparticles cause pregnancy complications in mice. Nat Nanotechnol 6:321–328

    Article  CAS  PubMed  Google Scholar 

  • Yan Z et al (2017) Zinc oxide nanoparticle-induced atherosclerotic alterations in vitro and in vivo. Int J Nanomed 12:4433–4442

    Article  CAS  Google Scholar 

  • Yang X et al (2014) Uptake of silica nanoparticles: Neurotoxicity and Alzheimer-like pathology in human SK–N–SH and mouse neuro2a neuroblastoma cells. Toxicol Lett 229:240–249

    Article  CAS  PubMed  Google Scholar 

  • Ye Y et al (2019) Effects of silica nanoparticles on endolysosome function in primary cultured neurons. Can J Physiol Pharmacol 97:297–305

    Article  CAS  PubMed  Google Scholar 

  • Yin N et al (2015) Silver nanoparticle exposure induces rat motor dysfunction through decrease in expression of calcium channel protein in cerebellum. Toxicol Lett 237:112–120

    Article  CAS  PubMed  Google Scholar 

  • Yin N et al (2018) Assessment of the developmental neurotoxicity of silver nanoparticles and silver ions with mouse embryonic stem cells in vitro. J Interdiscip Nanomed 3:133–145

    Article  CAS  Google Scholar 

  • Zeinabad HA, Zarrabian A, Saboury AA, Alizadeh AM, Falahati M (2016) Interaction of single and multi wall carbon nanotubes with the biological systems: tau protein and PC12 cells as targets. Sci Rep 6:26508

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Zhang Y et al (2019) Exposure to carbon black nanoparticles during pregnancy persistently damages the cerebrovascular function in female mice. Toxicology 422:44–52

    Article  CAS  PubMed  Google Scholar 

  • Zhang X et al (2020) Surface functionalization of pegylated gold nanoparticles with antioxidants suppresses nanoparticle-induced oxidative stress and neurotoxicity. Chem Res Toxicol 33:1195–1205

    Article  CAS  PubMed  Google Scholar 

  • Zhou M et al (2016) Implications for blood-brain-barrier permeability, in vitro oxidative stress and neurotoxicity potential induced by mesoporous silica nanoparticles: effects of surface modification. RSC Adv 6:2800–2809

    Article  CAS  Google Scholar 

  • Ziemińska E, Stafiej A, Struzyńska L (2014) The role of the glutamatergic NMDA receptor in nanosilver-evoked neurotoxicity in primary cultures of cerebellar granule cells. Toxicology 315:38–48

    Article  CAS  PubMed  Google Scholar 

Download references

Funding

This study is funded by Ministry of Education.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Peter H. M. Hoet.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Appendices

Appendix

In vitro result

Cellular uptake

AgNPs

Pristine AgNPs (6.5 nm, 10 µg/ml) were taken up by rat primary BME cells, astrocytes and pericytes in triple culture (Xu et al. 2015a). PVP-coated AgNPs of different sizes (23–102 nm) at 1–50 µg/ml were internalized by rat primary BME cells and rat astrocytes (Sun et al. 2016; Trickler et al. 2010). The internalization of citrate-coated AgNPs (49.7 nm, 50 µg/ml) was observed in N9 cells, a mouse microglia-derived cell line (Gonzalez-Carter et al. 2017).

Carbon-based NPs

Graphene oxide (50 µg/ml) was taken up by PC12 cells time-dependently (1–24 h) (Feng et al. 2018). Different types of MWCNTs at 10–30 µg/ml were internalized by neurons and mixed glia cells (Bussy et al. 2015; Visalli et al. 2017; Meng et al. 2013; Phillips et al. 2013). The internalized MWCNTs were found in cytoplasm, vacuoles, endosome, lysosome and nucleus.

IONPs

Fe3O4 NPs were internalized and found in cytoplasm and the perinuclear area of PC12 cells (Liu et al. 2018). O-IONPs were internalized by human astrocytes (Fernández-Bertólez et al. 2019). Cellular uptake of O-IONPs in human neurons was dose- (10–200 µg/ml) and time- (3–24 h) dependent (Fernández-Bertólez et al. 2018). Monocultures of D384 and SH-SY5Y cells and co-cultures/3D cultures of SH-SY5Y and D384 cells treated with PVP-coated Fe3O4 NPs showed dose- (1–100 µg/ml) and time- (24–48 h) dependent cellular uptake (Simone et al. 2018). SPIONs at 214 µg/ml were internalized by PC12 cells (SCoEaNIHR ES 2010). Uptake of MIRB by primary mouse neurons was also detected by FITC microscopy (Badman et al. 2020).

SiNPs

SiO2 NPs (12.1–25 nm, 10–200 µg/ml) were internalized by SK–N–SH cells and PC12 cells and found in cytoplasm or vacuoles (Yang et al. 2014; Xie and Wu 2016). Amorphous SiO2 NPs (100 µg/ml) were internalized via macropinocytosis and found in cytoplasm of primary BME cells (Liu et al. 2017). Internalization of FITC-labeled Stöber SiO2 NPs of different sizes (46–306 nm) by microglia cells and BME cells was size- and dose- (100–400 µg/ml) dependent. Neurons barely took up these NPs (Du et al. 2019; Sun et al. 2017). RuBpy-doped SiO2 NPs at 0.1 µg/ml were taken up by rat primary hippocampal neurons and found in endolysosomes (Ye et al. 2019).

Oxidative stress

AgNPs

Oxidative stress was induced by AgNPs (18.3 nm, 10 µg/ml), AgNPs (20 nm, 1 µg/ml), GSH-coated AgNPs (7.5 nm, 1 µg/ml) and green tea-coated AgNPs (12.5 nm, 1 µg/ml) in rat PC12 cells and NF-κB reporter gene-transfected rat N27 cells (Wang et al. 2009; Baruwati et al. 2013). Exposure to PVP-coated AgNPs of different sizes (15–23.18 nm) resulted in dose- (0.01–20 µg/ml) and time- (0.5–24 h) dependent oxidative stress responses in human ESCs, rat NSCs and rat primary astrocytes (Sun et al. 2016; Begum et al. 2016). PVP-coated (15 nm) AgNPs at 75 µg/ml also induced oxidative stress in rat primary cerebellar granule cells (Ziemińska et al. 2014). Citrate-coated AgNPs of different sizes (7.9–20.3 nm) induced a dose-dependent (0.1–50 µg/ml) oxidative stress response in human ESCs (Oh et al. 2016; Sun et al. 2016; Begum et al. 2016), with no oxidative stress observed when citrate-coated AgNPs of different sizes (34.08–49.7 nm, 1–50 µg/ml) were applied to mouse ESCs and mouse microglia cells (Yin et al. 2018; Gonzalez-Carter et al. 2017).

Carbon-based NPs

Different types of MWCNTs induced dose-dependent (5–50 µg/ml) nitric oxide (NO) production in primary mixed glia cells and increased reactive oxygen species (ROS) levels in retinoid acid (RA)-differentiated SH-SY5Y cells (Bussy et al. 2015; Visalli et al. 2017). Long (20 µm) SWCNTs (5–400 µg/ml) increase ROS production, lipid peroxidation, glutathione (GSH) depletion and decreased superoxide dismutase, catalase and glutathione peroxidase (GSH-Px) levels in PC12 cells (Wang et al. 2011).

IONPs

Fe3O4 NPs (30 nm) dose-dependently (25–200 µg/ml) induced oxidative stress in PC12 cells (Wu et al. 2013), but 45 nm of Fe3O4 NPs at 250–500 µg/ml did not produce oxidative stress in rat primary microglia cells (Xue et al. 2012). IONPs (10.8, 30.1 nm) in SH-SY5Y cells and rat primary BME cells dose-dependently (0.1–10 µg/ml) induced oxidative stress (Imam et al. 2015). PMA-coated IONPs dose-dependently (7–140 nM) induced oxidative stress in mNSCs and ReNcell VM, LAN-N-2 and Neuron-2a cells and dose-dependently (7–140 nM) decreased oxidative stress in hNSCs and C17.2 cells (Joris et al. 2017). DMSA-coated IONPs dose-dependently (7–140 nM) increased oxidative stress in hNSCs and ReNcell VM, LAN-N-2 and Neuron-2a cells and dose-dependently (7–140 nM) decreased oxidative stress in mNSCs and C17.2 cells (Joris et al. 2017). No obvious oxidative stress was observed after exposure of primary rat neurons to MIRB (Badman et al. 2020).

SiNPs

SiO2 NPs (12.1–15 nm) dose-dependently (2.5–200 µg/ml) increased the oxidative stress response in rat, mouse and human neurons (Wu et al. 2011; Yang et al. 2014), but 20 nm of SiO2 NPs at 250–500 µg/ml did not induce oxidative stress in rat primary microglia cells (Xue et al. 2012). Exposure to all sizes of Stöber SiO2 NPs (46–306 nm) at 50 µg/ml induced oxidative stress in microglia cells (Du et al. 2019). Amorphous SiO2 NPs (100 µg/ml) induced oxidative stress in human primary BME cells (Liu et al. 2017). Different types of mesoporous SiO2 NPs induced oxidative stress in NGF-treated PC12 cells (Zhou et al. 2016). LUDOX® TM SiO2 NPs at 100 µg/ml did not induce oxidative stress in SH-SY5Y or RA-induced SH-SY5Y cells (Kim and Yang 2011).

Pro-inflammatory responses

AgNPs

Exposure to different-sized PVP-coated AgNPs (1–50 µg/ml) induced secretion of cytokines, including CINC-2a/b, CINC-3, fratalkine, L-selectin, thymus chemokine, prostaglandin E2 (PGE2), IL-10, IL-1β, IP-10 and TNF-α, in rat primary astrocytes and rat primary BME cells (Sun et al. 2016; Trickler et al. 2010). Citrate-coated AgNPs at 50 µg/ml reduced lipopolysaccharide-induced TNF-α release in mouse microglia cells (Gonzalez-Carter et al. 2017).

Carbon-based NPs

Levels of TNF-α, IL-6 and IL-8 were increased in RA-treated SH-SY5Y cells after exposure to 12.5 µg/ml pristine and carboxyl-functionalized MWCNTs (Visalli et al. 2017).

IONPs

Fe3O4 NPs at 500 µg/ml induced TNF-α, IL-6 and IL-1β secretion in rat primary microglia cells (Xue et al. 2012).

SiNPs

SiO2 NPs (20 nm) at 500 µg/ml induced TNF-α, IL-6 and IL-1β release in rat primary microglia cells (Xue et al. 2012). Stöber SiO2 NPs (50 nm) at 100 µg/ml induced IL-1β secretion from mouse primary microglia cells and a macrophage cell line (J744A.1) (Du et al. 2019).

Morphology

Pristine AgNPs at 1–10 µg/ml commonly induced mitochondrial shrinkage in triple cultures of rat primary BME cells, astrocytes and pericytes (Xu et al. 2015b). PVP-coated AgNPs of different sizes induced cell perforation and dose-dependently (0.1–25 µg/ml) inhibited neurite outgrowth in rat primary BME cells and human ESCs (Trickler et al. 2010; Begum et al. 2016). Human ESCs treated with citrate-coated AgNPs showed a similar dose-dependent (0.1–50 µg/ml) reduction in neurite outgrowth and cell detachment and a round/irregular cell shape (Oh et al. 2016; Begum et al. 2016).

Carbon-based NPs

Different types of MWCNTs at 30 µg/ml induced organelle damage in PC12 and NGF-induced PC12 neurons (Meng et al. 2013). The PC12 neurons also tended to adhere to and grew on the MWCNTs (Phillips et al. 2013). Apoptotic markers were observed after PC12 neurons were exposed to SWCNTs (20 µm) at 100–400 µg/ml (SCoEaNIHR ES 2010).

IONPs

Fe3O4 NPs (17.9 nm) and n-octyltriethoxysilane-coated Fe3O4 NPs (18.7 nm) at 6–98 µg/ml both induced shape changes and inhibited neurite outgrowth in ReNcell VM cells (Ma et al. 2019). However, Fe3O4 NPs or n-octyltriethoxysilane-coated Fe3O4 NPs at 6–98 µg/ml induced no significant morphological changes in PC12 cells. PVP-coated Fe3O4 NPs at 1–100 µg/ml induced changes in cell shape and reduced cell density when applied to D384 cells in monoculture, in 3D culture or when co-cultured with SH-SY5Y cells (Simone et al. 2017; Simone et al. 2018). SH-SY5Y cells in 3D culture disaggregated in the presence of PVP-coated Fe3O4 NPs at 1–100 µg/ml. PMA-coated IONPs (3.5–70 nM) induced contractions in cell size and morphological changes in hNSCs, mNSCs and ReNcell VM, C17.2, Neuro-2a and LA-N-2 cells (Joris et al. 2017). DMSA-coated IONPs (3.5–70 nM) produced similar results in hNSCs, mNSCs and ReNcell VM, C17.2, Neuro-2a, and LA-N-2 cells (Joris et al. 2017). In contrast, DMSA-coated IONPs (2 mM) induced no observable morphological changes in rat primary cerebellar granule neurons (Petters and Dringen 2015).

SiNPs

Different types of pristine SiO2 NPs (12.1–15 nm) at 2.5–200 µg/ml induced morphological changes, inhibition of neurite outgrowth and cell detachment in cultures of mouse neuro-2 rat PC12 cells (Wu et al. 2011; Yang et al. 2014). Stöber SiO2 NPs (200 µg/ml) of different sizes (46–306 nm) induced a pyroptotic cell morphology in a microglia cell line, a primary microglia cell line and a macrophage cell line (Du et al. 2019). SH-SY5Y cells detached and became more irregularly shaped in the presence of LUDOX® TM SiO2 NPs (1.5 nm, 100 µg/ml) (Kim and Yang 2011). However, neurite outgrowth in RA-treated SH-SY5Y cells was not altered by exposure to LUDOX® TM SiO2 NPs (Kim and Yang 2011). Exposure to 100–400 µg/ml FITC-Stöber SiO2 NPs induced disruption of the plasma membrane, vacuolated cytoplasm, damaged organelles and apoptotic cell morphology in C17.2 neurons (Sun et al. 2017).

Cytotoxicity and cell death

AgNPs

PVP-coated AgNPs of different sizes dose-dependently (0.01–80 µg/ml) induced cytotoxicity in human ESCs, human NSCs and rat NSCs and primary astrocytes (Liu et al. 2015; Begum et al. 2016). In human BME cells, cytotoxicity was induced time-dependently (24–48 h) with NPs applied at 10 µg/ml, whereas in rat primary BME cells size-dependent cytotoxicity was induced with NPs applied at 50 µg/ml (Trickler et al. 2010; Khan et al. 2019). Cytotoxicity was also induced by PVP-coated AgNPs (15 nm, 10 µg/ml) in rat primary cerebellar granule cells (Ziemińska et al. 2014). Citrate-coated AgNPs of different sizes (8–34 nm) dose- (0.1–200 µg/ml) and time-dependently (6–24 h) induced cytotoxicity in human ESCs (Begum et al. 2016). In contrast, no cytotoxicity was observed in mouse ESCs and mouse microglia cells with citrate-coated AgNPs (34–50 nm) at 1–50 µg/ml (Yin et al. 2018; Gonzalez-Carter et al. 2017). Citrate-coated (20 and 110 nm) and PVP-coated (110 nm) gold core AgNPs at 50 µg/ml induced cytotoxicity in mouse primary midbrain cells (Weldon et al. 2018).

PVP-coated AgNPs of different sizes (23–23.18 nm, 5–10 µg/ml) induced apoptosis in rat primary astrocytes and NSCs (Sun et al. 2016; Liu et al. 2015). Exposure to citrate-coated AgNPs (7.9 nm, 25–50 µg/ml) time-dependently (6–24 h) induced apoptosis in human ESCs (Oh et al. 2016).

Carbon-based NPs

Graphene dose-dependently induced cytotoxicity (10–100 µg/ml) in rat BME cells (Rosas-Hernandez et al. 2019). Graphene oxide dose- (5–60 µg/ml) and time-dependently (6–24 h) induced cytotoxicity in rat neurons. Carbon black NPs induced cytotoxicity in umbilical vein endothelial cells (14 nm, 100 µg/ml) but had no cytotoxic effect on primary astrocytes (19 nm, 128 µg/ml) (Zhang et al. 2019). PC12 cells exposed to different types of SWCNTs exhibited dose-dependent cytotoxicity (0.01–600 µg/ml) (Wang et al. 2011; Zeinabad et al. 2016). Different types of MWCNTs (pristine, high iron impunities, carboxyl functionalized) dose- (0.01–200 µg/ml) or time-dependently (6–72 h) induced cytotoxicity in PC12 and SH-SY5Y cells (Zeinabad et al. 2016; Phillips et al. 2013). MWCNTS (pristine or functionalized) induced cytotoxicity in rat primary mixed glia cells, related to the brain region from where cells were isolated (Bussy et al. 2015).

Different types of MWCNT at 10–66.5 µg/ml induced apoptosis in primary mixed glia cells or necrosis in PC12 cells (Bussy et al. 2015; Zeinabad et al. 2016). Different types of SWCNTs at 22.7–100 µg/ml induced apoptosis in PC12 cells (Wang et al. 2011; Zeinabad et al. 2016). Graphene oxide at 40–60 µg/ml induced autophagy in PC12 cells (Feng et al. 2018).

IONPs

O-IONPs dose-dependently (5–200 µg/ml) induced cytotoxicity in human astrocytes and neurons (Fernández-Bertólez et al. 2018; Fernández-Bertólez et al. 2019). PVP-coated Fe3O4 NPs dose- (1–100 µg/ml) and time-dependently induced cytotoxicity in 3D cultures of D384 and SH-SY5Y cells, monocultures of D384 and SH-SY5Y cells and D384 and SH-SY5Y co-cultures (Simone et al. 2017, 2018). Repeated dosing of PVP-coated Fe3O4 NPs dose-dependently (0.1–25 µg/ml) induced cytotoxicity in both 3D cultures of D384 and SH-SY5Y cells. Lysosomal-targeted IONPs (200 µg/ml) but not mitochondria-targeted IONPs or pristine IONPs induced cytotoxicity in SH-SY5Y cells (Huang et al. 2019). PMA-coated IONPs dose-dependently (3–140 nM) induced cytotoxicity in hNSCs, mNSCs, and ReNcell VM, C17.2, LAN-N-2 and Neuron-2a cells (Joris et al. 2017). Exposure to DMSA-coated IONPs dose-dependently (3–140 nM) induced cytotoxicity in hNSCs, mNSCs and ReNcell VM and LAN-N-2 cells (Joris et al. 2017). DMSA (2 mM)-coated IONPs did not induce cytotoxicity but rather delayed cytotoxicity in rat primary cerebellar granule neurons (Petters and Dringen 2015).

Fe3O4 NPs of different sizes dose-dependently (25–200 µg/ml) induced apoptosis in PC12 cells (Fernández-Bertólez et al. 2018; Wu et al. 2013). Exposure to 10.8 nm IONPs at 2.5–40 µg/ml triggered apoptosis in SH-SY5Y cells (Imam et al. 2015). In contrast, exposure to 30.1 nm IONPs did not induce apoptosis in SH-SY5Y cells (Imam et al. 2015). MIRB (10 and 20 µg/ml) after 24-h exposure induced significant cytotoxicity in primary mouse neurons (Badman et al. 2020).

SPIONs (214 µg/ml) induced apoptosis in PC12 cells (Sun et al. 2015). O-IONPs dose-dependently (5–200 µg/ml) induced apoptosis in human astrocytes and human neurons (Fernández-Bertólez et al. 2018; Fernández-Bertólez et al. 2019).

SiNPs

Stöber SiO2 NPs size- (46–306 nm) and dose- (25–200 µg/ml) dependently induced cytotoxicity in microglia cells and macrophages (Du et al. 2019). Stöber SiO2 NPs (46 nm) at 100 µg/ml also induced cytotoxicity in primary microglia cells (Du et al. 2019). In monocultures of human primary BME cells, amorphous SiO2 NPs dose-dependently (25–800 µg/ml) induced cytotoxicity (Liu et al. 2017). In co-cultures of human primary BME cells and human primary astrocytes, exposure to amorphous SiO2 NPs dose-dependently (25–200 µg/ml) induced cytotoxicity in only BME cells. LUDOX® TM SiO2 NPs dose-dependently (10–1000 µg/ml) induced cytotoxicity in SH-SY5Y and RA-induced SH-SY5Y cells (Kim and Yang 2011). Different types of mesoporous SiO2 NPs (266–498.4 nm) induced cytotoxicity dose-dependently (12.5–100 µg/ml) (Zhou et al. 2016). Different sizes of SiO2 NPs (15–68 nm) all dose-dependently (0.01–200 µg/ml) induced cytotoxicity in rat primary hippocampal neurons, rat PC12 cells and mouse neuro-2a and human SK-N-SH cells (Wu et al. 2011; Ye et al. 2019).

Different sizes of SiO2 NPs (12.1–15 nm) dose-dependently (2.5–200 µg/ml) induced apoptosis in rat, mouse and human neuronal cell lines (Wu et al. 2011; Yang et al. 2014). SiO2 NPs (25 nm) at 25 to 200 µg/ml induced autophagy in rat neurons (SCoEaNIHR ES 2010). Stöber SiO2 NPs size- (46–306 nm) and dose-dependently (25–200 µg/ml) induced pyroptosis in mouse microglia cells (Du et al. 2019). Exposure to LUDOX® TM SiO2 NPs induced apoptosis in both SH-SY5Y and RA-treated SH-SY5Y cells (Kim and Yang 2011). FITC-Stöber SiO2 NPs dose-dependently (100–400 µg/ml) induced apoptosis in microglia.

Genotoxicity

AgNPs

PVP-coated (23 nm) AgNPs at 5 µg/ml induced DNA damage in rat ESCs (Liu et al. 2015).

Carbon-based NPs

Pristine and carboxyl-functionalized MWCNTs at 12.5–25 µg/ml induced DNA damage in RA-treated SH-SY5Y cells (Visalli et al. 2017). SWCNTs dose-dependently (50–100 µg/ml) induced cell cycle arrest (G2/M phase) in PC12 cells (Wang et al. 2011).

IONPs

O-IONPs dose-dependently (5–100 µg/ml) induced DNA damage and cell cycle arrest (S phase) in human astrocytes (Fernández-Bertólez et al. 2019). O-IONPs induced DNA damage and dose-dependent (10–200 µg/ml) cell cycle arrest (G0/G1 phase) in human neurons (Fernández-Bertólez et al. 2018). Fe3O4 NPs (30 nm) at 25–200 µg/ml induced cell cycle arrest (G2/M phase) in PC12 cells (Wu et al. 2013).

SiNPs

Exposure to SiO2 NPs (15 nm, 25–200 µg/ml) induced cell cycle arrest (G2/M phase) in NGF-treated PC12 cells (Wu et al. 2011). FITC-Stöber SiO2 NP exposure induced cell cycle arrest (G2/M phase) in mouse microglia as shown by a dose-dependent decrease in proliferation rate (Sun et al. 2017). Exposure to LUDOX® TM SiO2 NPs at 100 µg/ml induced DNA damage and cell cycle arrest in SH-SY5Y cells (G0/G1 phase) and RA-treated SH-SY5Y cells (G2/M phase) (Kim and Yang 2011).

In vivo results

Distribution of NPs into the nerve system

AgNPs

Sprague–Dawley (SD) rats (male/female) orally administered pristine AgNPs (6 nm; 1 and 10 mg/kg body weight [bw]t/day for 14 days) showed elevated Ag level in blood and a dose-dependent accumulation of Ag in the brain (Xu et al. 2015a). Ag level was not significantly elevated in the forebrain of male Wistar rats administered citrate-coated AgNPs (10 nm) via oral gavage (0.2 mg/kg bw/day for 14 days). TEM revealed elevated Ag level in the blood and nanosized dark granules in the hippocampus, which could be AgNPs (Skalska et al. 2015). IP-administered PVP-coated AgNPs in Male Wistar rats (14.3 nm; 200 µg/per rat/day, daily for 5 days) were found in cytoplasm and nuclei of hippocampal neurons (Ghooshchian et al. 2017). Ag level was elevated in the cerebellum of SD rats (male/female) after 14 weeks daily of NI of citrate-coated AgNPs (20 nm; 0.1, 0.2, 0.5, 1 mg/kg bw) (Yin et al. 2015). Fatemi et al. investigated possible prenatal neurotoxicity by exposing pregnant Wistar rats to citrate-coated AgNPs (20 nm; 25 mg/kg bw/day) starting from day 9 of gestation until the day of delivery. Ag level was elevated in the brain of pups (Fatemi et al. 2013).

Carbon-based NPs

No biodistribution of carbon-based NPS to the CNS or peripheral nervous system (PNS) was assessed in the studies reviewed.

IONPs

Fe level was increased dose-dependently in serum, frontal cortex, hippocampus and cerebellum in male albino (Mus musculus) mice orally administered Fe2O3 NPs (45 nm; 25, 50 mg/kg bw/day for 30 days) (Manickam et al. 2019). Fe level was also increased in serum, frontal cortex, hippocampus and cerebellum of Swiss albino mice (male/female) orally administered Fe2O3 NPs (50 nm) for 30 days (25, 50 mg/kg bw/day) (Manickam et al. 2018). Male CD ICR mice with NI of Fe2O3 NPs (21 nm) for 30 days (130 µg/per animal/every 2 days) had detectable levels of NPs in neurons of axons in the olfactory bulb and hippocampal mitochondria and lysosome (Wang et al. 2009). With NI of 125I-radiolabeled Fe3O4 NPs (36 nm; 20 µg/per male SD rat/day for 1 and 7 days), NPs were observed in the olfactory bulb, striatum, hippocampus, brain stem and cerebellum; half of the NPs still remained in striatum and hippocampus after 14 days of administration. Higher Fe content was related to longer exposure (Wu et al. 2013). In male SD rats, IV-injected IONPs (10.8 nm, 50 mg/kg bw) translocated to the brain after 1 h (Imam et al. 2015). Wistar rats sacrificed 1, 7 or 21 days after IP administration of IONPs (62.5 nm; 2, 14 mg/kg bw) showed a dose-dependent increase in Fe level in the cerebral cortex. Prussian blue staining was positive at blood vessel intima and in macrophages of the soft meninges in the high-dose groups at both 1 day and 7 days’ post-injection (Sheida et al. 2017). Kim et al. investigated neurotoxicity in the PNS by injecting (iron concentration at 0.5, 15 mM) DMSA-coated Fe2O3 NPs (9 nm) into the sciatic nerve of female SD rats. NPs were accumulated at 48 h post-administration (Kim et al. 2013).

SiNPs

After NI of 125I-radiolabeled SiO2 NPs (20 µg/per rat/day for 1 and 7 days), accumulation of particles was time-dependently increased in the olfactory bulb, striatum, hippocampus, brain stem, cerebellum and frontal cortex of SD rats (male/female) (Wu et al. 2011). Yamashita et al. IV-injected cyanine dye (DY-676)-labeled SiO2 NPs (70, 300, 1000 nm), carboxyl-functionalized SiO2 NPs (70 nm) and amine-functionalized SiO2 NPs (70 nm) in pregnant BALB/c mice (800 µg/per mouse); all types of SiO2 NPs accumulated in liver, whereas only 70 nm-SiO2 NPs were also found in placenta (regardless of whether they were functionalized or not) (Yamashita et al. 2011).

Histopathology and ultrastructural change

AgNPs

Female SD rats showed shrinkage of neurons and swelling of both astrocytes and extravascular lymphocytes after oral administration of pristine AgNPs (6 nm; 1 mg/kw bw/day for 7 days) (Xu et al. 2015a). The forebrain of male Wistar rats showed myelin damage after oral administration of citrate-coated AgNPs (10 nm) (Dąbrowska-Bouta et al. 2016). Orally administered citrate-coated AgNPs (10 nm; 0.2 mg/kg bw/daily for 14 days) induced synaptic vesicles, meylin-like bodies, swollen synapses and blurred synaptic cleft in the forebrain and hippocampus of male Wistar rats (Skalska et al. 2015).

NI of citrate-coated AgNPs (20 nm) induced a loosen and separated granular layer with Purkinje cell deficiency in the cerebellum of SD rats (male/female) (Yin et al. 2015). The cerebrum of C57BL/6 mice (male/female) showed no significant histopathological change after inhalation administration (1.919 × 107 particle number/m3, 6 h/day for 5 days/week for 2 weeks) of pristine AgNPs (22.18 nm) (Lee et al. 2010). Demyelination and BBB leakage were induced after IP, IV, ICA and IB injection of pristine AgNPs (55 nm). (Sharma et al. 2009).

Carbon-based NPs

The cerebral cortex of C57BL/6J mice (male/female) with NI of Printex 90 carbon black NPs (14 nm; 21, 103, 515 µg/per mouse/1 h/day for 10 days) showed decreased neuronal numbers (indicated as a reduction in microtubule-associated protein 2 staining), demyelination and fewer and shallower Nissl bodies (Zhang et al. 2019). IP-injected MWCNTs (diameter 15 nm, length 2 µm; 2.5 mg/kg/bw/day for 14 days) induced loosen and disordered hippocampal CA1 neurons in the brain of male Wistar rats (Gao et al. 2015). Prenatal neurotoxicity was measured by exposing pregnant ICR mice to NI of Printex 90 carbon black NPs (2.9, 15 or 73 μg/kg bw/day, on days 5 and 9 of gestation). The cerebral cortex of pups showed a dose-dependent increase in aquaporin-4 level (Onoda et al. 2017).

IONPs

The frontal cortex, hippocampus and cerebellum of male albino mice (M. musculus) showed demyelination and mitochondrial damage after oral administration of Fe2O3 NPs (45 nm) (Manickam et al. 2019). NI of Fe2O3 NPs (21 nm) induced dilated rough endoplasmic reticulum (ER)-degenerated dendrite and disrupted membrane structure in the olfactory bulb and hippocampus of male CD ICR mice (Wang et al. 2009). No brain lesions were observed in male SD rats after NI of Fe3O4 NPs (30 nm) (Wu et al. 2011). Striatum of male SD rats showed damage to the brain vasculature after IV injection of IONPs (10.8 nm) (Imam et al. 2015). The cerebral cortex of Wistar rats (male/female) showed neuronal damage, dose-dependent edema, cell dissociation, and glia and BME cell proliferation after IP injection of IONPs (62.5 nm) (Sheida et al. 2017). C57BL/6J mice IB injected with Fe3O4 NPs (49 nm; 10 μg/μl, daily for 7 and 14 days) showed a decrease in tyrosine hydroxylase-positive fibers in the dorsal striatum and in hematoxylin-labeled nuclei in hippocampus. However, no significant brain lesions were observed in either region (Liu et al. 2018). PNS neurotoxicity was reported in the sciatic nerve of female SD rats injected with DMSA-coated Fe2O3 NPs (9 nm); NP-laden phagocytes and edema were observed in the sub-perineurial area alongside severe edema, blood–nerve barrier disruption, loss of spindle-like cells and fiber in the perivascular area (Kim et al. 2013).

SiNPs

Male SD rats with NI of 125I-radiolabeled SiO2 NPs (20 nm) showed no significant histopathology changes (Wu et al. 2011). IP-administered amorphous SiO2 NPs (20 nm; 50 mg/kg bw/day for 28 days) induced neuronophagia, perivascular edema and satellitosis in the cerebral cortex of male SD rats (Liu et al. 2017).

DNA damage

AgNPs and SiNPs

DNA damage was not studied in the studies reviewed.

Carbon-based NPs

No significant DNA damage was observed in the olfactory bulb, cerebellum or remaining brain areas of wild-type and p47phox-/- C57BL/6J mice (male/female) after inhalation delivery of pristine carbon NPs (58.8 nm; 154 µg/m3, 4 h) (Berlo et al. 2014).

IONPs

Male albino mice (M. musculus) orally exposed to Fe2O3 NPs (50 nm) showed G1/S cell-cycle checkpoint activation in the frontal cortex, hippocampus and cerebellum (Manickam et al. 2018).

Oxidative stress

AgNPs

Male Wistar rats orally administered a repeating dose of citrate-coated AgNPs (10 nm; 0.2 mg/kg bw/day for 14 days) showed elevated ROS levels in brain and liver (Skalska et al. 2016). GSH and GSH-Px in the brains of pups were depleted with citrate-coated AgNPs (20 nm) prenatally administered by oral gavage (25 mg/kg bw/day, from day 9 of gestation to the last day of pregnancy) (Fatemi et al. 2013).

Carbon-based NPs

The levels of oxidative biomarkers (mRNA expression of 8-oxoguanine DNA glycosylase and heme oxygenase 1 [HO-1]) were not altered in the brains of wild type or p47 phox−/− C57BL/6J mice after inhalation (154 μg/m3, for 4 h) of pristine carbon-based NPs (58.8 nm) (Berlo et al. 2014).

IONPs

Levels of ROS in the frontal cortex, hippocampus and cerebellum were dose-dependently increased in male albino mice (M. musculus) orally administered Fe2O3 NPs (50 nm; 25 and 50 mg/kg bw/day for 30 days) (Manickam et al. 2018). Levels of oxidative stress biomarkers were changed in striatum (GSH level decreased, H2O2 level increased) and hippocampus (H2O2 level increased) after NI of 125I-radiolabeled Fe3O4 NPs (30 nm; 25 and 50 mg/kg bw/day, for 1 and 7 days). Superoxide dismutase and malondialdehyde (MDA) levels were not changed in either brain region in this study (Wu et al. 2013). NI of Fe2O3 NPs (21 nm; 130 μg/animal, every other day for 12 h, 72 h and 14 days) induced various levels of oxidative stress in hippocampus, cortex, cerebellum and brain stem of male albino mice (M. musculus) (Wang et al. 2009). HO-1 protein expression was increased in female SD rats injected with DMSA-coated Fe2O3 NPs (9 nm) in the sciatic nerve (Kim et al. 2013).

SiNPs

MDA level was significantly increased in male SD rats IP injected with amorphous SiO2 NPs (50 mg/kg bw, daily for 28 days).

Significant oxidative stress biomarkers were found in striatum (H2O2 and MDA levels increased and GSH level decreased) and hippocampus (GSH level decreased) in male SD rats after NI of SiO2 NPs (15 nm; 20 µg/per rat, daily for 1 and 7 days).

Cell death mechanism

AgNPs

Three studies reported apoptotic cell death induced by AgNPs; no studies reported autophagy induced by AgNPs. Male Wistar rats IP-injected with PVP-coated AgNPs (14.3 nm; 100–400 µg/per rat) showed a dose-dependent increase in apoptosis (downregulation of Bcl-2 and overexpression of Bax) in hippocampus (Ghooshchian et al. 2017). Ganjuri et al. studied the prenatal neurotoxicity of pristine AgNPs (30 nm) by orally exposing pregnant Wistar rats to NPs (1–10 µg/ml NPs in drinking water over the entire gestational period). Pro-caspase 3 mRNA level was dose-dependently increased in the brains of pups (Ganjuri et al. 2015). Fatemi et al. also observed a significant increase in level of the apoptotic marker caspase 9 in the brains of pups exposed to citrate-coated AgNPs (20 nm) in utero (Fatemi et al. 2013).

Carbon-based NPs

Apoptosis was not studied in the reports covered by this review. One study reported induction of autophagy with the administration of carbon-based NPs.

Repeated IP injection of MWCNTs (diameter 10–20 nm, length 2 μm; 2.5 mg/kg bw/day for 14 days) induced autophagy biomarkers (ratio of LC II/LC I mRNAs and increased Beclin-1 mRNA level) in brains of male Wistar rats (Gao et al. 2015).

IONPs

Two studies reported induction of apoptosis by IONP administration; no studies investigated the possible link between IONPs and autophagy. Apoptotic biomarkers (upregulation of Bax and cleaved caspase 3, downregulation of total Bcl-2) were changed in the frontal cortex, hippocampus and cerebellum of albino mice orally administered Fe2O3 NPs (50 nm; 25 and 50 mg/kg bw, daily for 30 days) (Manickam et al. 2018). Apoptotic cells were increased in number in the dorsal striatum and hippocampus of C57BL/6J mice injected with Fe3O4 NPs (49 nm; 2 μg per animal for 7 and 14 days) (Liu et al. 2018). The sciatic nerve of female SD rats injected with DMSA-coated Fe2O3 NPs (9 nm) showed elevated levels of the apoptotic biomarkers cleaved and uncleaved caspase 3 in both S100+ Schwann cells and TCR+ lymphocytes (Kim et al. 2013).

SiNPs

SiNP-induced apoptosis and autophagy were not studied.

Pro-inflammatory responses

AgNPs

Female SD rats orally administered pristine AgNPs (6 nm; 1, 10 mg/kg bw, daily for 14 days) showed increased IL-4 level in the brain (Xu et al. 2015a).

Carbon-based NPs

No significant pro-inflammatory responses were observed in the brain of wild type or p47phox−/− C57BL/6J mice (male/female) exposed to pristine carbon NPs (58.8 nm) via inhalation (154 µg/m3, 4 h) (Berlo et al. 2014).

IONPs

The pro-inflammatory response was evaluated in only one study looking at the effects of peripheral NP administration. Pro-inflammatory biomarker levels were increased (IL-1β and phosphorylated p38; gelatinolytic matrix metalloproteinase 9) in S100+ Schwann cells, CD68+ macrophages and endothelial cells of the sciatic nerve in treated female SD rats (Kim et al. 2013).

SiNPs

NI of pristine SiO2 NPs (15 nm; 20 µg/per animal, daily for 1 or 7 days) increased TNF-R, IL-1β, and IL-8 levels in the striatum of male SD rats, with no significant effects in the hippocampus (Wu et al. 2011). mRNA levels of NF-κB, P65 and TNF-α were increased in hippocampus dentate gyrus of C57BL/6J mice exposed to FITC-labeled SiO2 NPs (91 nm) via inhalation (Fu et al. 2018).

Astrocyte activation

AgNPs

Astrocytes were activated in SD rats (male/female) with NI of citrate-coated AgNPs (20 nm; 0.1, 0.2, 0.5, 1 mg/kg bw, daily for 98 days) (Yin et al. 2015). Injected (IP, IV, ICA, IB) pristine AgNPs (55 nm) activated astrocytes in male C57Balb mice and male SD rats (Sharma et al. 2009).

Carbon-based NPs

No astrocyte activation was found in C57BL/6J mice (male/female) after inhalation of UFCP (37.5 nm) (Morris-Schaffer et al. 2019). Astrocytes were dose-dependently activated at the perivascular area of the cerebral cortex in 6-week-old ICR mouse offspring with prenatal NI of Printex 90 carbon black (14 nm; 2.9, 15, 73 µg/kg bw/day, on days 5 and 9 of gestation) (Onoda et al. 2017).

IONPs

Oral administration of Fe2O3 NPs activated astrocytes in the frontal cortex, hippocampus and cerebellum of male albino mice (M. musculus) (Manickam et al. 2019).

SiNPs

Astrocytes were activated in the cerebral cortex and hippocampus of male SD rats intraperitoneally administered amorphous SiO2 NPs (20 nm; 50 mg/kg bw, daily for 28 days) (Liu et al. 2017).

Microglia activation

AgNPs, carbon-based NPs, IONPs

No study investigated microglia activation.

SiNPs

Fu et al. investigated microglia activation by exposing pregnant C57BL/6J mice to FITC-labeled SiO2 NPs (91 nm) via inhalation. Microglia activation was observed in the dentate gyrus of pups (Fu et al. 2018).

Function assessment (animal behavior)

AgNPs

Male Wistar rats orally administered citrate-coated AgNPs (10 nm) showed slight anxiety-like behaviors and changes in nociception reaction but no significant differences in motor function (locomotor activity and motor coordination) or memory performance (Dąbrowska-Bouta et al. 2016) Motor activity (locomotor activity and motor coordination) was dose-dependently impaired in SD rats (male/female) after NI of citrate-coated AgNPs (20 nm) (Yin et al. 2015). Antsiferova et al. orally administered male C57BL/6 mice with PVP-coated AgNPs in distilled water (31 nm; 50 µg/day for 30, 60, 120, 180 days); cautious and anxiety-related behavior were observed after 30–60 days but disappeared after 120 days. Exploratory behavior was increased at 120 days post-administration, with impaired contextual memory and loss of cautious behavior reported after 180 days (Antsiferova et al. 2018). Tabatabaei et al. investigated prenatal functional impairment of pregnant NMRI mice by subcutaneous administration of citrate-coated AgNPs (10 nm; 0.2–2 mg/kg bw/day, every 3 days until delivery); sex-specific depressive-like behaviors were observed in offspring (Zhang et al. 2019)

Carbon-based NPs

Newborn C57BL/6J mice (male/female) administered UCFP (37.5 nm) via inhalation (50 µg/m3, postnatal days 4–7 and 10–13, 4 h/day) showed altered locomotor activity only in females. The authors detected no significant alterations in memory, learning, anxiety-like behavior, social preference and impulsivity (Morris-Schaffer et al. 2019). Gao et al. found spatial learning and functional memory impairments in male Wistar rats after repeated IP injection of MWCNTs (diameter 10–20 nm, length 2 μm; 2.5 mg/kg bw/day for 14 days) (Gao et al. 2015).

IONPs

Dose-dependent impairment of motor activity, significant alteration in gait pattern and memory defects were noted in male albino mice (M. musculus) orally administered Fe3O4 NPs (45 nm; 25 or 50 mg/kg bw/day for 30 days) (Liu et al. 2017). Wistar rats (male/female) with IP injection of IONPs (62.5 nm; 2 or 14 mg/kw bw, for 1, 7 or 21 days) showed dose-dependent anxiety and inhibited locomotory activity (Sheida et al. 2017). Manickam et al. injected Fe2O3 NPs (49 nm; 10 µg/µl/day for 7 or 14 days) in the dorsal striatum or hippocampus of C57BL/6J mice (male/female); Both spatial learning and memory were impaired. Motor coordination was decreased in mice injected in the striatum, with impairment appearing at 7 days post-injection (Zhang et al. 2019).

SiNPs

C57BL/6J mouse (male/female) offspring prenatally exposed to FITC-labeled SiO2 NPs (91 nm) via inhalation (55.56, 111.11, 277.78 mg/m2, 30 min/day for 7 days) showed decreased social novelty preference and increased anxiety-like behavior (Fu et al. 2018).

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gong, JY., Holt, M. ., Hoet, P.H.M. et al. Neurotoxicity of four frequently used nanoparticles: a systematic review to reveal the missing data. Arch Toxicol 96, 1141–1212 (2022). https://doi.org/10.1007/s00204-022-03233-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00204-022-03233-1

Navigation