Environmental Chemistry Letters
https://doi.org/10.1007/s10311-020-01033-6
REVIEW
Toxicity of metal and metal oxide nanoparticles: a review
Ayse Busra Sengul1 · Eylem Asmatulu2
Received: 22 August 2019 / Accepted: 7 June 2020
© Springer Nature Switzerland AG 2020
Abstract
Nanotechnology has recently found applications in many fields such as consumer products, medicine and environment. Nanoparticles display unique properties and vary widely according to their dimensions, morphology, composition, agglomeration
and uniformity states. Nanomaterials include carbon-based nanoparticles, metal-based nanoparticles, organic-based nanoparticles and composite-based nanoparticles. The increasing production and use of nanoparticles result in higher exposure
to humans and the environment, thus raising issues of toxicity. Here we review the properties, applications and toxicity of
metal and non-metal-based nanoparticles. Nanoparticles are likely to be accumulated in sensitive organs such as heart, liver,
spleen, kidney and brain after inhalation, ingestion and skin contact. In vitro and in vivo studies indicate that exposure to
nanoparticles could induce the production of reactive oxygen species (ROS), which is a predominant mechanism leading to
toxicity. Excessive production of ROS causes oxidative stress, inflammation and subsequent damage to proteins, cell membranes and DNA. ROS production induced by nanoparticles is controlled by size, shape, surface, composition, solubility,
aggregation and particle uptake. The toxicity of a metallic nanomaterial may differ depending on the oxidation state, ligands,
solubility and morphology, and on environmental and health conditions.
Keywords Nanotechnology · Nanoparticles · Metal nanoparticles · Toxicity mechanism · Reactive oxygen species
Introduction
Nanotechnology has become one of the most rapidly growing areas of science and technology in the USA as well as
other parts of the world. It has led to the increased production and applications of nanomaterials in a wide range of
fields such as automotive, biomedical, cosmetics, defense,
energy and electronics. The global market for nanotechnology products and applications was valued at $39.2 billion in
2016 and is expected to reach $90.5 billion in 2021 (McWilliams 2016).
The nanomaterial is defined as a material with any external dimension in the nanoscale or having an internal structure or surface structure in the nanoscale, approximately
1–100 nm size range (ISO 2015). They may be in the form
of nanoparticles, nanofibers, nanotubes, nanocomposites
* Eylem Asmatulu
[email protected]
1
Department of Civil and Construction Engineering,
Kennesaw State University, Kennesaw, GA 30144, USA
2
Department of Mechanical Engineering, Wichita State
University, 1845 Fairmount Street, Wichita, KS 67260, USA
and nanostructured materials. Figure 1 shows the classification of nanostructure materials according to dimensions,
morphology, composition, agglomeration and uniformity
states. Nanoparticle agglomeration, size and surface reactivity, along with shape and size, need to be considered
when choosing health and environmental regulations for new
materials (Asmatulu et al. 2013; Asmatulu 2013). Nanoparticles have been used in many products due to their unique
physicochemical properties, which include a large surfaceto-volume ratio, extremely small size and size-dependent
optical properties (Sajid et al. 2015). For example, cobalt
nanoparticles have gained great interest in biomedicalrelated fields such as drug delivery and magnetic resonance
imaging (Ansari et al. 2017). Silica nanoparticles are used in
chemical and electronic industries, building materials, foods,
and the biomedical and biotechnological fields (Kaphle et al.
2018; Zhou et al. 2019). Titanium dioxide and zinc oxide
nanoparticles are frequently used as suncare products to
minimize the unwanted skin whitening effect and to provide high UV protection efficacy and pleasant skin aesthetics
(Stark et al. 2015).
Although the uses of nanoparticles have contributed
significant advantages in many areas, nanoparticles raise
13
Vol.:(0123456789)
Environmental Chemistry Letters
Fig. 1 Classification of nanostructured materials. (1) Dimensionality of nanoparticles; one-dimensional (1D), two-dimensional (2D)
and three-dimensional (3D) nanoparticles. (2) Morphology; flatness,
sphericity and aspect ratio. (3) Composition; composed of a single-
constituent or composite materials. (4) Uniformity and agglomeration: nanomaterials can be dispersed in aerosols, suspensions/colloids
or an agglomerate state. Adapted from Buzea et al. (2007)
serious concern about exposure and adverse effects on
human and the environment. In the recent years, a number of in vitro and in vivo studies have been performed to
understand the toxicological impacts and possible hazards
of different nanoparticle exposures to human and the environment. There is still a major gap in knowledge about the
toxicity effects of nanoparticle exposures. In this review,
an overview of the properties and potential applications of
metal and non-metal-based nanoparticles is provided and
the most recent research studies related to toxic effects of
exposure to metal and non-metal-based nanoparticles are
highlighted.
This article is an abridged version of the chapter by Sengul, A.B., and Asmatulu, E. [Nanomaterials Causing Cellular Toxicity and Genotoxicity, In V. Kumar, P. Guleria, S.
Ranjan, N. Dasgupta, and E. Lichtfouse, Nanotoxicology and
Nanoecotoxicology] that will be published in the book series
Environmental Chemistry for a Sustainable Word (http://
www.springer.com/series/11480).
properties but also viewed as the fundamental building
blocks of various applications. Carbon-based particles
mostly consist of carbon nanotubes, fullerenes and their
derivatives, carbon black, nanodiamonds, graphite nanoparticles, graphene nanoparticles and graphene oxide
(Patel et al. 2019). These particles have a wide of applications ranging from biomedicine through nanoelectronics to
mechanical engineering because of their versatile shapes and
various properties like electronic and thermal conductivity
(Ema et al. 2016; Sardoiwala et al. 2018). Among all of
them, metal-based nanoparticles are the most widely used
nanoparticles and are extensively used in different applications such as solar cells, paints, coatings, cosmetics, UV
blockers in sunscreen and environmental remediation (EPA
2017). Physiochemical properties, high stability, high reactivity, photothermal and plasmonic properties make metallic
nanoparticles an attractive candidate as a therapeutic agent
(Sardoiwala et al. 2018). Another group of nanoparticles
is organic-based. These well-defined nano-sized polymers
are built from branched units capable of being tailored to
perform specific chemical functions (Roy and Bhattacharya 2015). Organic-based nanoparticles such as micelles,
liposomes, dendrimers and polymers are extensively used
in various biosensors/diagnostics, drug delivery, polymer
materials, chemical sensors, modified electrodes, DNAtransfecting agents and therapeutic agents for prion diseases
(Farré and Barceló 2012; Sandeep 2013). Composite-based
1. Applications of nanoparticles
Nanotechnology has attracted great attention in recent years
due to its innovative applications in many fields, ranging
from consumer products to medicine to improving the environment. Nanoparticles play an important role in nanotechnology, not only because of their unique physicochemical
13
Environmental Chemistry Letters
nanoparticles can combine with carbon–carbon nanoparticles, carbon-inorganic nanoparticles and inorganic–inorganic nanoparticles (Jeevanandam et al. 2018). They have
novel electrical, catalytic, magnetic, mechanical, thermal
and imaging features. Composite-based nanoparticles are
used in drug delivery and cancer detection, as well as in
auto parts and packaging materials to enhance mechanical
and flame-retardant properties. The specific properties and
applications of different nanoparticles are summarized in
Table 1. The advantageous and disadvantageous of metallic
and non-metallic nanoparticles are provided in Table 2.
2. Toxicity mechanisms of nanoparticles
The estimated increase in the production and use of nanoparticles will result in their enhanced exposure to humans and
the environment. Humans are generally exposed to nanoparticles via inhalation (respiratory tract), ingestion (gastrointestinal tract), skin contact and injection (blood circulation)
(Fu et al. 2014; Wu and Tang 2018). Toxicity mechanism of
nanoparticles is given in Fig. 2. When nanoparticles enter
into the human body, they may cross various cellular barriers
and reach the most sensitive organs such as lung, liver and
kidney, thus resulting in mitochondrial damage, deoxyribonucleic acid (DNA) mutations and eventually cell apoptosis/
death (Ahamed et al. 2010; Bahadar et al. 2016; Shin et al.
2015; Tan et al. 2018).
The production of reactive oxygen species (ROS), which
could cause oxidative stress, inflammation and consequent
damage to proteins, cell membranes and DNA, is a predominant mechanism leading to toxicity (Fard et al. 2015; Fu
et al. 2014; He et al. 2015; Liu et al. 2013; Manke et al.
2013). The level of ROS production induced by nanoparticles is dependent on several factors such as size, shape,
surface, composition, solubility, aggregation/agglomeration,
particle uptake, the presence of mutagens and transition metals affiliated with the particles (Fu et al. 2014; Gatoo et al.
2014; He et al. 2015; Jeevanandam et al. 2018; Manke et al.
2013; Shvedova et al. 2012). Gliga et al. (2014) revealed
size-dependent cytotoxicity of silver nanoparticles since
only the 10-nm silver nanoparticles were cytotoxic for the
human lung cells starting at doses of 20 μg/mL. However,
no coating-dependent cytotoxicity was found between the
10-nm citrate and 10-nm polyvinylpyrrolidone-coated silver
nanoparticles. Rizk et al. (2017) found that the liver function enzymes, oxidative stress markers and liver histological pattern were greatly influenced with dose and time of
titanium oxide nanoparticles (21 nm), while the genetic disturbance started at the high dose (500 mg/kg body weight)
of exposure and for long duration (45 days). Steckiewicz
et al. (2019) also demonstrated that the cytotoxicity of gold
nanoparticles was shape-dependent. Gold nanostars with
the highest anticancer potential were found to be the most
cytotoxic, whereas gold nanospheres with small anticancer
potential were found to be less toxic. The following part will
explain metallic and non-metallic nanoparticles and their
toxicity effects.
3. Nanoparticles and Their Toxicity
Nanoparticle characteristics (e.g., size and shape), composition, concentration and related toxicity effects are given in
Table 3.
Human can exposure to nanomaterials with three ways;
inhalation into the pulmonary system, absorption through
the dermal system and ingestion through the gastrointestinal
system. Because of the size and shape, nanoparticles may
diffuse quicker in air than their larger counterparts and can
get further down the respiratory tract. Inhalation can create
great concerns owing to the primary impacts of particulate
nanomaterials. The primary consideration is being given to
examining impacts on the respiratory and the cardio-vascular framework. Considering dermal entry, healthy skin has a
superior obstruction when compared to the respiratory tract
system. The hindrance capacity could be constrained by
skin injuries, solid mechanical strain or little nanoparticles
(< 5–10 nm). Entry by ingestion is of lower concern among
the all. Many potential health effects are shown in Fig. 3.
Metallic nanoparticles
Gold
Gold nanoparticles have been widely used in biomedical
fields due to their various advantageous and properties
such as tunable sizes, facile synthesis, easy modification
and strong optical properties (Jia et al. 2017). The outcome
regarding gold nanoparticle toxicity is as yet conflicting.
Some researcher reported that these particles are biocompatible and have unimportant toxicity (Mukherjee et al. 2016;
Negahdary et al. 2015; Nelson et al. 2013; Sung et al. 2011),
while others emphasized that they cause ROS leading to
oxidative stress and are also highly toxic because of their
physicochemical properties (Li et al. 2010; Lopez-Chaves
et al. 2018; Thakor et al. 2011; Zhang et al. 2010).
Nelson et al. (2013) demonstrated that the citrate-stabilized
gold nanoparticles (10, 30 or 60 nm) do not induce discrete,
dose-dependent oxidative damage to DNA in either a human
liver carcinoma cells or calf thymus-DNA at concentrations
less than 0.2 or 2 μg/mL, respectively, nor do they cause cell
death or produce free radicals. Senut et al. (2016) found that
human embryonic stem cells exposed to 1.5 nm mercaptosuccinic acid-capped gold nanoparticles exhibited loss of cohesiveness, rounding up and detachment resulting in ongoing
cell death at concentrations as high as 0.1 μg/mL, whereas
other gold nanoparticles (4 and 14 nm) exhibited almost no
13
13
Table 1 Properties and applications of different nanoparticles
Type of nanoparticle Examples
Carbon-based
Specific properties
Carbon nano- High thermal and electrical conductivity
tubes (CNTs) High tensile strength and elasticity
Aspect ratio
C60
Antioxidant capacity
Radical scavenging
Carbon black
Reinforcing effects
Thermal and electrical conductivity
Resistance to UV radiation
Antioxidation effect
Nanodiamonds Ease of functionalization
High biocompatibility
Graphene
Electrical and thermal conductivity
High surface area
High strength
Good elasticity
Ease of functionalization
Chemical inertness
Gas impermeability
Applications
References
Composites to consumer electronics, energy stor- Ema et al. (2016 and Ma-Hock et al. (2013)
age, health care
Targeted drug delivery, energy applications, elec- Aschberger et al. (2010) and Ming et al. (2018)
tronics, polymer modifications, environment,
cosmetic products
Sahu et al. (2014)
Reinforcement agent in rubber products, black
pigment in printing inks and lithography,
electrodes for batteries in electrical conductors,
finishing process of leather goods production
Drug and gene delivery
Mengesha and Youan (2013)
Gurunathan and Kim (2016 and Ma-Hock et al.
Sensors, batteries, fuel cells, supercapacitors,
(2013)
transistors, components of high-strength
machinery, display screens, biomedical applications
Environmental Chemistry Letters
Type of nanoparticle Examples
Specific properties
Metal-based
NIR-responsive drug delivery, NIR-responsive
controlled release, delivery of chemical drugs
and genes, theranostic application, promising
agents in bioimaging, biosensing and as immunotherapy carriers
Antibacterial, antifungal, antiviral and antifilarial Consumer products including electronics, cosmetics, household appliances, textiles and food
activity
production as well as in biomedical applicaOxidative activity
tions such as antimicrobial agents, drug delivery, molecular imaging, biomedical sensing and
even cancer photodynamic therapy
High magnetism
Pigments, catalysts, sensors, magnetic contrast
Electrical and catalytic properties
agents and in energy storage devices
Fuel cells, polymers, paints, coatings, textiles,
High hardness
biomaterials, batteries, adsorbent, grinding,
Mechanical strength
catalysis, polishing abrasives
Wear resistance
Chemical inertness with good biocompatibility
Paints, plastics, cosmetics, personal care products
Brightness
and as food additives and drug delivery agents,
High refractive index
coatings, papers, inks, medicines, pharmaceutiResistance to discoloration
cals, food products, toothpaste
Stronger catalytic activity
Catalysis, paints, wave filters, UV detectors,
Higher chemical activity
transparent conductive films, varistors, gas
Oxidation resistance
sensors, solar cells, sunscreens and cosmetic
Corrosion resistance, photocatalysis
products
Stronger absorption
Shielding ability to the ultraviolet rays
Superparamagnetic
Drug delivery, magnetic resonance imaging,
thermal ablation therapy, in vivo cell tracking, magnetic separation of cells or molecules
and remediation of different environmental
contaminants such as heavy metals, chlorinated
organic solvents
Au
Ag
Co
Al2O3
TiO2
ZnO
IO
Tunable sizes
Facile synthesis
Easy modification
Optical properties
Applications
References
Jia et al. (2017)
Akter et al. (2018), Cameron et al. (2018), Chen
et al. (2015), Foldbjerg et al. (2011) and Guo
et al. (2018)
Cappellini et al. (2018) and Wan et al. (2017)
Future Markets (2013), Kim et al. (2010) and
Poborilova et al. (2013)
Ranjan and Ramalingam (2016), Shi et al. (2013),
Ursini et al. (2014), Weir et al. (2012) and Yin
et al. (2012)
Guan et al. (2012) and Huang et al. (2010)
Feng et al. (2018), Guerra et al. (2018) and Naqvi
et al. (2010)
Environmental Chemistry Letters
Table 1 (continued)
13
13
Table 1 (continued)
Type of nanoparticle Examples
Specific properties
Applications
References
Organic-based
Series of branches
Multivalency
Globular structure
Well-defined molecular weight with controlled
surface
Small size
Biodegradability
Water solubility
Non-toxicity
Long shelf life
Stability
Fluorescence brightness
High photostability
Good biocompatibility
Large surface area-to-volume ratios
Tunable pore size and connectivity
Biodegradability
Ease of surface modification
Biosensors/diagnostics, drug delivery, gene
transfection, catalysis
Azmi and Shad (2017) and Sandeep (2013)
Delivery of drugs, proteins and DNA or genes to
specific targeted tissues or organs
Yadav et al. (2019)
Dendrimers
Polymers
Composites-based
Quantum dots
Silica
Havrdova et al. (2016) and Matea et al. (2017)
Solar cells, photovoltaic devices, light-emitting
diode fabrication, photodetectors, computing,
biomedical imaging and so on
Duan et al. (2013) and Kim et al. (2015)
Biomedical and biotechnological fields, such
as medical diagnostics, drug delivery, gene
therapy, biomolecules detection, photodynamic
therapy and bioimaging
C60: Fullerene; NP: nanoparticle; Ag: silver; Al2O3: aluminum oxide; Au: gold; Co: cobalt; IO: iron oxide; TiO2: titanium dioxide; ZnO: zinc oxide
Environmental Chemistry Letters
Environmental Chemistry Letters
Table 2 The advantages and disadvantages of metallic and non-metallic nanoparticles
Nanoparticle Advantages
Metallic
Al2O3
Au
CuO
Ag
ZnO
IO
TiO2
Disadvantages
Particles instability: nanomaterials can experience
Enrich Rayleigh scattering
a transformation since they are in high energy
Surface-enhanced Raman scattering
local minima and thermodynamically unstable.
Great plasma absorption.
These cause deterioration of quality, weak corroBiological system imaging
sion resistance, and keeping the structure is hard
Reveal chemical information on metallic nanoscale
Impurity: during the synthesis of nanoparticles,
substrate
nitrides, oxides, the generation can aggravate
Uniformity in shape, size and branch length
from the impure nature. Since nanoparticles are
highly reactive, there can be high chances of
impurity too
Biologically harmful: nanomaterials have been
toxic, carcinogenic and cause irritation
Explosion: exothermic combustion can follow up
with an explosion since the fine metal particles
behave as strong explosives
Difficulty in synthesis: synthesizing of nanoparticles is extremely difficult, so they should be
encapsulated
Non-metallic
C
Multiple functions, chemical alteration, biocompatible and water-soluble, effective loading
Conductivity and high surface area
Carbon
nanotube
(CNT)
Mesoporous High surface area, decent electrostatic immosilica (MS) bilization, immediate adjustment for covalent
immobilization
Effective carrier system for hydrophilic drug,
Polymeric
biocompatible, functional alteration, targeting the
micelles
potential
(PMs)
References
Kumar et al. (2018)
Toxicity
Kahraman et al. (2017)
Difficult functionalization, fragile and aggregation
Lamberti et al. (2014)
pH sensitivity
Kahraman et al. (2017)
Occasional cytotoxicity, need of surface alterations Kahraman et al. (2017)
Ag: silver; Al2O3: aluminum oxide; Au: gold; C: carbon; CNT: carbon nanotube; CuO: copper oxide; IO: iron oxide; TiO2: titanium oxide; ZnO:
zinc oxide
Fig. 2 Toxicity mechanism
of nanoparticles mediated by
reactive oxygen species (ROS)
generation. The model describes
extracellular sources of ROS
as exposure routes for the engineered nanoparticles. Intracellular ROS can be generated from
the mitochondria, which later
causes lipid peroxidation, DNA
damage and protein denaturation
toxic effects on the human embryonic stem cells at concentrations as high as 10 μg/mL. Hanna et al. (2018) observed
that the majority of gold nanoparticles with a range of sizes
(30, 60 and 100 nm) and surface coatings (polyvinylpyrrolidinone, polyethylene glycol, citrate, dendrimers and branched
polyethylenimine) tested had little to no impact on nematode
13
13
Table 3 Nanoparticles characteristics and related toxicity mechanisms
Physicochemical characterizations
Toxicity assays
Cells
Results
Au
Shape: rods, stars, spheres
Particle size (nm): ≈ 39 nm length,
18 nm width (rods), ≈ 215 nm
(stars) ≈ 6.3 nm (spheres)
Concentration (μg/mL.): 0.3–5
MTT
NR
hFOB 1.19
143B
MG-63
Ag
Allium cepa
Shape: Spherical
Particle size (nm): ≈ 5, 25, 50, 75
Concentration (mg/L): 100
Hydrodynamic diameter (nm):
42.6 ± 19.2 (5 nm Au), 77.1 ± 26.2
(25 nm Au), 80.5 ± 30.4 (50 nm Au),
124.4 ± 48.1 (75 nm Au)
Allium cepa root
Co
Compositions: Co3O4 nanograins,
Co(OH)2 nanoflakes and Co3(PO4)2
microflakes
Particle size: 35–40 nm (nanograins),
100 nm (nanoflakes) and 1 μm
(microflakes)
Concentration (μg/mL): 100
HaCaT
Au nanoparticles stars were the most Steckiewicz et al. (2019)
cytotoxic, whereas Au nanoparticles
spheres were the less cytotoxic ones
IC50 values were the lowest for 143B
cell line in comparison with hFOB
1.19 and MG-63 cell lines for Au
nanoparticles in all investigated
shapes
Au nanoparticles-induced apoptosis in
human osteosarcoma cells, both in
143B and MG-63
Au nanoparticles penetrated through
the cell membrane and caused ultrastructural changes
Ag nanoparticles-induced alterations Scherer et al. (2019)
on the root elongation, germination,
mitotic, nuclear abnormality and
micronucleus indices
MPs permeated the plant cell wall,
ending in an increase of the cytotoxic and genotoxic with decreasing
the Ag nanoparticles diameter
Anwar et al. (2019)
Co3(PO4)2 microflakes and Co(OH)2
nanoflakes showed potent amoebicidal activity at 100 and 10 μg/ml
against Acanthamoeba castellanii as
compared to Co3O4
Encystation and excystation assays
showed consistent inhibition at
100 μg/ml
Co nanoparticles inhibited amoebaemediated host cell cytotoxicity as
determined by LDH release without
causing significant damage to
human cells when treated alone
Amoebicidal
Encystation
Excystation
LDH
References
Environmental Chemistry Letters
Nanoparticles
Nanoparticles
Physicochemical characterizations
Toxicity assays
Cells
Results
Al2O3
Particle size (nm): 20–80
Surface area (m2/g): 40
Hydrodynamic diameter (nm):
303–512 (in Millipore water)
Zeta potential (mV): 12.41 (in Millipore water)
Concentration (μg/mL): 10, 25, 50
and 100
MTT
SOD
CAT
GSH
GST
LPO
CHSE-214
TiO2
WST-1
Shape: bipyramids, rods, platelets
LDH
Particle size (nm): 50 ± 9
(bipyramids), 108 ± 47 (rods), 75 ± 25 Comet assay
(platelets), 20 ± 5 (P25), 150 ± 50
(food grade)
Hydrodynamic diameter (nm): 66 ± 20
(bipyramids), 36 ± 12 (rods),
233 ± 70 (platelets), 107 ± 31 (P25),
184 ± 61 (food grade)
Concentration (μg/mL): 5, 10, 20, 50
and 80
20, 50, 80, 120, and 160 μg/mL exposure with light
A dose-dependent decline in cell
Srikanth et al. (2015)
viability was observed in CHSE-214
cells exposed to Al2O3 nanoparticles
Oxidative stress induced by Al2O3
nanoparticles in CHSE-214 cells has
resulted in the significant reduction of SOD, CAT and GSH in a
dose-dependent manner. However, a
significant increase in GST and LPO
was observed in CHSE-214 cells
exposed to Al2O3 nanoparticles in a
dose-dependent manner
Significant morphological changes
in CHSE-214 cells were observed
when exposed to Al2O3 nanoparticles at 6, 12 and 24 h
Gea et al. (2019)
After light exposure, the largest
cytotoxicity was observed for rods
starting from the dose of 10 μg/
ml; P25, bipyramids and platelets
showed a similar cytotoxic effect
(doses of 50 μg/ml); no effect was
induced by food grade at the tested
concentrations
No significant LDH release was
detected
Food grade and platelets induced
direct genotoxicity, while P25
(50 μg/ml), food grade (50 μg/ml)
and platelets (80 μg/ml) caused
oxidative DNA damage
No genotoxic or oxidative damage was
induced by bipyramids and rods
Biological effects were overall lower
in darkness than after light exposure.
Only food grade, P25 and platelets
were internalized by cells, and the
uptake resulted correlated with
genotoxicity
BEAS-2B
References
Environmental Chemistry Letters
Table 3 (continued)
13
13
Table 3 (continued)
Nanoparticles
Physicochemical characterizations
Toxicity assays
Cells
Results
References
ZnO
Shape: Spherical
Size (nm): 30–50
Concentration (μg/mL): 1,5, 10, 15
and 20
Hydrodynamic diameter (nm) in
media: 284.76 ± 8.03
Zeta potential (mV): − 12.46 ± 0.28
Flow cytometry
MTT
DCFH-DA
HPRT gene forward mutation
Comet assay
Micronucleus
V-79
ZnO nanoparticles induced oxidative Jain et al. (2019)
stress and HGPRT gene mutation
All the genotoxicity endpoints such as
chromosomal break, DNA damage
and mutagenicity were observed at
6 h of ZnO nanoparticle exposure
ZnO nanoparticles manifested the cell
cycle arrest, ultrastructural modifications and further cell death
A significant increase in the apoptotic
cells was detected
Environmental Chemistry Letters
Nanoparticles
Physicochemical characterizations
Toxicity assays
Cells
Results
References
IO
Shape: spherical
Compositions: PEI and PEG
Particle size (nm): 10 and 30 (PEG),
10 (PEI)
Concentration (μg/mL): 3.125, 6.250,
12.500, 25.000, 50.00 and 100.000
Hydrodynamic diameter (nm):
17.2 ± 5.0 (PEI 10), 16.5 ± 4.7 (PEG
10), 35.8 ± 10.3 (PEG 30)
Zeta potential (mV): + 29.28 (PEI 10),
− 0.52 (PEG 10) and − 0.52 (PEG
30)
MTS
Hoechst 33342 and PI staining
LDH
Annexin V-FITC/PI staining
DCF-DA
PEI-coated IO nanoparticles exhibited Feng et al. (2018)
In vitro: RAW264.7
significantly higher uptake (after 4-h
SKOV-3
incubation) than PEGylated ones
In vivo:
Nu/Nu strain, BALB/c mice in both RAW264.7 and SKOV-3
cells and caused dose-dependent
severe cytotoxicity through multiple
mechanisms such as ROS production (at the concentrations ranging
from 5 to 20 μg/mL) and apoptosis
(after 24-h incubation)
10 nm PEGylated IO nanoparticles
showed higher cellular uptake than
30 nm ones and were slightly cytotoxic only at high concentrations
PEGylated IO nanoparticles but not
PEI-coated IO nanoparticles were
able to induce autophagy
All the IO nanoparticles tended to
distribute in the liver and spleen,
and the biodegradation and clearance of PEGylated IO nanoparticles
in these tissues were relatively slow
(> 2 weeks)
10 nm PEI-coated IO nanoparticles
showed the lowest uptake in tumor
tissue, whereas 10 nm PEGylated IO
nanoparticles achieved the highest
tumor uptake, followed by 30 nm
PEGylated IO nanoparticles
No obvious toxicity was found for
PEGylated IO nanoparticles in
BALB/c mice, whereas PEI-coated
IO nanoparticles exhibited dosedependent lethal toxicity
Environmental Chemistry Letters
Table 3 (continued)
13
13
Table 3 (continued)
Nanoparticles
Physicochemical characterizations
Pt
Toxicity assays
Results
MTS
Particle size (nm): ≈ 70
DCFH-DA
Composition: citrate
Agglomerate size (nm) in media:
112.4 ± 7.0
Zeta potential (mV) in media:
− 12.2 ± 0.9
Concentration (μg/mL): 5, 25 and 100
HepG2
Si
Particle size (nm): 10, 25, 50, 100
Concentration (μg/mL): 1, 5, 25
HUVECs
QDs
MTT
Shape: Spherical
Flow cytometry
Composition: MPA and PEG
Particle size (nm): ≈ 20
Zeta potential (mV) in buffer: ≈ 20
Concentration (nM): 20, 30, 35, 40,
50, 60, 75, and 100 (PEG) and 80,
90, 100, 110, 125, 150, 175, and 200
(MPA)
Labrador-Rached et al. (2018)
At dosages of 25 μg/mL or less, no
toxicity was identified. However, at
the high exposure concentration an
approximate 25% cytotoxic response
transpired
The Pt nanoparticles induced ROS
production in a dose-dependent
fashion, with a substantial response
associated with the 25 μg/mL condition
Citrate-coated, 70-nm Pt nanoparticles
were able to activate a significant
stress response in HepG2 liver cells,
even in the absence of cytotoxicity
Zhou et al. (2019)
At the administrative concentrations
(1, 5, 25 μg/mL), all the four sizes
of Si nanoparticles could induce
an increase of both DNA damages
and MN frequencies in culture,
with a positive dose- and negative
size-dependent effect relationship
(S100 < S50 < S25 < S10)
Si nanoparticles significantly
enhanced levels of intracellular
ROS, but decreased levels of GSH
The levels of Nrf2 protein were significantly enhanced
Peynshaert et al. (2017)
PEGylated QDs induce cytotoxicity
and oxidative stress, MPA-coated
QDs are non-toxic up to 200 nM and
reduce oxidative stress
PEGylated QDs are easily taken up at
low concentrations though uptake
reaches a maximum around 40 nM.
MPA-coated QDs are taken up proportionally with increasing dosage,
though are only taken up efficiently
at higher concentrations
PEGylated QDs induce lysosomal
impairment, while MPA-coated
cause lysosomal activation
Comet assay
MN
ROS
GSH
GFP-LC3 HeLa
References
Environmental Chemistry Letters
Cells
Nanoparticles
Physicochemical characterizations
Concentration (μg/mL): 0.1, 1, 10, 50
C60
SWCNT Fe3O4 and 100
BET surface area (m2/g): 10.58 (C60),
808.3 (SWCNT), 40.89 (Fe3O4)
Average hydrodynamic diameter (nm):
142.0 (C60), 112.9 (SWCNT), 316.6
(Fe3O4)
Zeta potential (mV): − 37.4 ± 6.45
(C60), 12.9 ± 3.19 (SWCNT),
− 18.4 ± 4.49 (Fe3O4)
Particle size (nm): 36.64 (C60), 9.00
(SWCNT diameter), 28.33 (Fe3O4)
Co3O4 Fe2O3
The hydrodynamic diameter (nm)
in water: 96.4 ± 0.57 (Co3O4),
SiO2
74.6 ± 0.6 (Fe2O3), 41.7 ± 2.6 (SiO2),
Al2O3
and 90.2 ± 2.4 (Al2O3)
Shape: spherical
Particle size (nm): 35.8 ± 0.8 (Co3O4),
43.7 ± 4.7 (Fe2O3), 17.1 ± 2.1 (SiO2),
and 39.4 ± 3.9 (Al2O3)
Concentration (μg/mL): 10, 25, 50, 75
and 100
Toxicity assays
Cells
Results
References
MTT
LDH
Fluorescence
hPDLFs
mDFs
MTT,
LDH,
ROS,
Comet assay
Human lymphocytes
Donmez Gungunes et al. (2017)
All three nanoparticles caused loss
of membrane integrity in time, in a
gradually increasing manner
In the 6 h of exposure, all three nanoparticles triggered the formation of
ROS in both hPDLF and mDF cells
mDF cells were more sensitive to
C60, SWCNT and Fe3O4 nanoparticles exposure compared to hPDLF
cells, which exhibited signs of
cellular adaptation at nanoparticle
exposures of lower concentrations
Co3O4 nanoparticles showed decrease Rajiv et al. (2016)
in cellular viability at doses of 50,
75, and 100 mg/ml and increase in
cell membrane damage followed by
Fe2O3, SiO2, and Al2O3 nanoparticles in a dose-dependent manner
after 24 h of exposure to human
lymphocytes
The oxidative stress was evidenced in
human lymphocytes by the induction
of reactive oxygen species, lipid peroxidation, and depletion of catalase,
reduced glutathione, and superoxide
dismutase
The Al2O3 nanoparticles showed the
least DNA damage when compared
with all the other nanoparticles
Chromosomal aberration was
observed at 100 mg/ml when
exposed to Co3O4 nanoparticles and
Fe2O3 nanoparticles
The cytotoxicity and oxidative stress
lead to DNA damage and chromosomal aberrations in human
lymphocytes
Environmental Chemistry Letters
Table 3 (continued)
13
13
Table 3 (continued)
Nanoparticles
Physicochemical characterizations
Toxicity assays
Cells
Results
References
PLGA-based
Mean diameter (nm) in water:
200–230 (PLGA/PVA, PLGA/CS,
PLGA/PF68),170 (PLGA stabilizerfree)
170–230 (DY700-PLGA, DY700PLGA/PVA, DY700-PLGA/CS,
DY700-PLGA/PF68)
Zeta potential (nm): 200–230 (PLGA/
PVA, PLGA/CS, PLGA/PF68), 170
(PLGA stabilizer-free), 170–230
(DY700-PLGA, DY700-PLGA/
PVA, DY700-PLGA/CS, DY700PLGA/PF68)
Concentration (mg/mL): 0.01, 0.1, 1,
and 10
Shape: spherical and monodisperse
MTT
Apoptosis/necrosis
Cytokine secretion
H2DCFHDA
THP-1
All tested nanoparticles showed no or Grabowski et al. (2015)
scarce signs of toxicity at therapeutically relevant concentrations (up to
0.1 mg/mL)
At high concentrations (above 1 mg/
mL), cytotoxicity was found to be
induced by the presence of stabilizers
While stabilizer-free PLGA nanoparticles exerted no cytotoxicity,
the slightly cytotoxic CS polymer
conferred PLGA nanoparticles
significant cytotoxicity when used
as nanoparticles stabilizer; more
surprisingly, the otherwise innocuous PVA and PF68 polymers also
conferred a significant cytotoxicity
to PLGA nanoparticles
143B: human bone osteosarcoma; Ag: silver; Al2O3: aluminum oxide; Au: gold; BEAS-2B: human bronchial epithelium cell line; CAT: catalase; CHSE-214: chinook salmon cells; Co: cobalt;
Co(OH)2: cobalt hydroxide; Co3(PO4)2: cobalt phosphate; Co3O4: cobalt oxide; GSH: glutathione; GST: glutathione sulfotransferase; HaCaT: human keratinocyte cell; hFOB 1.19: human fetal
osteoblast cell line; LDH: lactate dehydrogenase; LPO: lipid peroxidation; MG-63: human bone osteosarcoma cell line; MTT: mitochondria functioning; NR: Neutral-red uptake; SOD: superoxide dismutase; TiO2: titanium dioxide; WST-1: cell proliferation reagent; DCF-DA: dihydrodichlorofluorescein diacetate; DCFH-DA: dichlorodihydrofluorescein diacetate; GSH: glutathione;
HepG2: human liver cell line; HGPRT: hypoxanthine–guanine phosphor ribosyl transferase; HUVECs: human umbilical vein endothelial cell line; IO: iron oxide; MN: micronucleus; MTS:
CellTiter 96 AQueous One Solution Cell Proliferation; Nu/Nu strain: athymic nude mice; PEG: polyethylene glycol; PEI: polyethylenimine; Pt: platinum; RAW264.7: murine macrophages;
ROS: reactive oxygen species; Si: silica; SKOV-3: human ovarian cancer cell line; V-79: Chinese hamster lung fibroblast cell line; ZnO: zinc oxide; Al2O3: aluminum oxide; C60: fullerene;
Co3O4: cobalt (II, III) oxide; CS: chitosan; Fe2O3: iron (III) oxide; Fe3O4: iron oxide; GFP-LC3 HeLa: cervical epithelial cancer cell line HeLa; hPDLFs: human periodontal ligament fibroblasts; LDH: lactate dehydrogenase; mDFs: mouse dermal fibroblasts; MPA: 3-mercaptopropionic; PF68: poloxamer 188; PLGA: poly(lactide-co-glycolic); PVA: poly(vinyl alcohol); QDs:
quantum dots; SiO2: silicon dioxide; SWCNT: single wall carbon nanotubes; THP-1: human monocyte-derived macrophages
Environmental Chemistry Letters
Environmental Chemistry Letters
Fig. 3 Diseases associated with nanoparticles. Diseases linked to the
inhaled nanoparticles include asthma, bronchitis, lung cancer, Parkinson’s and Alzheimer’s. Nanoparticles in the gastro-intestinal tract
have been associated to Crohn’s disease and colon cancer. Nanoparticles that come into the circulatory system are linked to manifesta-
tion of arteriosclerosis, thrombus and heart diseases, and eventually
cardiac death. Moving to other organs, like liver and spleen, may
cause to some other diseases of those organs. Exposure to some nanoparticles is correlated with the experience of autoimmune diseases.
Adapted from Buzea et al. (2007)
growth or reproduction, suggesting low toxicity. However,
gold nanoparticles with positively charged coatings, such
as branched polyethylenimine-coated gold nanoparticles or
amine terminated (silicon and polystyrene nanoparticles),
resulted in significant toxic effects in Caenorhabditis elegans.
Additionally, Steckiewicz et al. (2019) showed that gold
nanoparticles stars (≈ 215 nm) with the highest anticancer
potential were the most cytotoxic type of tested nanoparticles, whereas gold nanoparticles spheres (≈ 6.3 nm) which
appear to be the safest one with small anticancer potential.
Additionally, short-term exposure to gold nanoparticles may
not be toxic to cell growth, while long-term exposure may
disturb cellular metabolism as well as energy homeostasis.
Cellular uptake can occur with gold nanoparticles, and they
can stay in endosomes/lysosomes or rely upon their surface
functionalization into nuclei. Gold nanoparticles uptake relies
on functionalization as well as cell type. If the gold nanoparticles are taken up by a healthy cell, then it will be eliminated
eventually, whereas if it is taken up by a cancer cell, then the
result will be cell death (Thota and Crans 2018).
et al. 2015; Guo et al. 2018; Pulit-Prociak et al. 2014). Even
though the increased use of silver nanoparticles provides
many advantages in biomedical applications, their toxicological effects have explored. Silver nanoparticles can enter
the body in different ways and accumulate in different tissues and organs, and passing the blood–brain barrier and
eventually reaching the brain (Ciucă et al. 2017). Compared
with other nanosized metals, silver nanoparticles show
greater toxicity regarding the enhanced synthesis of ROS
and leakage of the enzyme lactate dehydrogenase (Dasgupta
and Ramalingam 2016; Foldbjerg et al. 2011; Haase et al.
2011). The toxicity of silver nanoparticles was reported
to be dependent on many factors, including physical and
chemical properties of silver nanoparticle, environmental
conditions and contact interactions (Guo et al. 2018). Chen
et al. (2015) examined the size-dependent toxicity of silver
nanoparticles using three different characteristic sizes (15,
50 nm and 100 nm) against fish red blood cells. The smallest
sized silver nanoparticle displayed a greater ability to induce
hemolysis and membrane damage than particles of other
sizes. Another toxicological study on silver nanoparticles
revealed polyvinylpyrrolidone-coated silver nanoparticles on
bacteria, viruses, microalgae, fungi and human and animal
cells (including cancer cell lines) via a variety of viability
and toxicological assays. It was discovered that biological
systems from different taxonomies were inhibited with a
concentration of silver nanoparticles at a similar magnitude
(Vazquez-Muñoz et al. 2017).
Silver
Antibacterial, antifungal, antiviral and antifilarial properties of silver nanoparticles lead to use of these particles for
biomedical purposes such as antimicrobial agents, drug
delivery, molecular imaging, biomedical sensing and even
cancer photodynamic therapy (Cameron et al. 2018; Chen
13
Environmental Chemistry Letters
Nonetheless, Mulenos et al. (2020) showed the relatively
low toxicity of silver nanoparticles based on the fact that
only minutes amounts of silver ions, of 6.9 μg/mL, are
released in oxic conditions, and 0.2 μg/mL under anoxic
conditions, both levels being much lower to that of natural conditions. Indeed, toxicity is induced and controlled
by production of ions. They found similar data for copper
and titania.
Cobalt
Cobalt-based nanoparticles are used in pigments, catalysts, sensors, magnetic contrast agents and in energy storage devices (Cappellini et al. 2018). Exposure to cobalt or
cobalt-containing nanoparticles was found to induce oxidative stress, DNA damage, morphological transformation
and inflammatory responses in different cell types (Annangi
et al. 2015; Wan et al. 2012, 2017). In addition, two cobaltcontaining substances, cobalt sulfate and cobalt-tungsten
carbide, are listed as reasonably anticipated to be human
carcinogens in the report on carcinogens based on sufficient
evidence from experimental studies (NTP 2016).
The potential toxicity of cobalt-based nanoparticles has
been studied previously using different in vivo and in vitro
models. Chattopadhyay et al. (2015), for example, evaluated
the intracellular signaling transduction pathways involved
in cobalt oxide nanoparticles mediated oxidative stress.
It was found that bare cobalt oxide nanoparticles induced
cell death significantly generated by ROS, which induced
tumor necrosis factor alpha. This tumor necrosis factor alpha
serves as an important function in cell death by activating
caspase-8 followed by p38 MAPK and caspase-3. This study
also revealed that bare cobalt oxide nanoparticles create a
toxic effect on the human immune cell. Wan et al. (2017)
explored the genotoxic effects of cobalt nanoparticles in vivo
by using guanine phosphoribosyltransferase delta transgenic
mice. They have shown that exposure to cobalt nanoparticles
caused oxidative stress, lung inflammation and injury, and
cell proliferation, which further resulted in DNA damage
and mutation. Cappellini et al. (2018) also showed cellular uptake of three different cobalt-based nanoparticles in
lung cells and induction of DNA strand breaks and oxidative
damage by cobalt metal and cobalt (II) oxide nanoparticles.
Aluminum oxide
The Global Market for aluminum oxide nanoparticles
reported that aluminum-based nanoparticles contribute to
20% of all nanosized chemicals (Future Markets 2013). They
have found a wide range of applications in many sectors as
paints, polymers, coatings, textiles, fuel cells, solar energy,
airbag propellants and energetic materials because of its
high hardness and mechanical strength (Kim et al. 2010;
13
Poborilova et al. 2013). There is evidence that exposure
of aluminum oxide nanoparticles may lead to reduce cell
viability, increase oxidative stress, mitochondrial dysfunction and alter proteins expression of blood–brain barrier
(Alshatwi et al. 2012; Kim et al. 2010; Pakrashi et al. 2011;
Yu et al. 2011; Zhang et al. 2011).
Di Virgilio et al. (2010) observed dose-dependent cytotoxicity at lower concentrations (5 µg/mL) in Chinese hamster ovary cells treated with titanium oxide and aluminum
oxide nanoparticles after 24 h by changes in lysosomal and
mitochondrial dehydrogenase activity. They also reported
genotoxic effects by micronucleus frequencies, which significantly increased at 0.5 and 1 µg/mL titanium oxide and
0.5–10 µg/mL aluminum oxide nanoparticles. Moreover,
Srikanth et al. (2015) revealed that aluminum oxide nanoparticles induce cytotoxicity and membrane damage in Chinook
salmon cells that may be mediated through morphological
abnormalities, lipid peroxidation and oxidative stress.
Titanium dioxide
Titanium dioxide is one of the most increasingly manufactured nanoparticles and is listed in the top five nanoparticles
in consumer products (Gea et al. 2019). They have been
widely used in consumer products (sunscreens, cosmetics,
toothpaste, pharmaceuticals, paints, plastics, self-cleaning
devices and food additives) and industrial and medical applications because of their stronger catalytic activity (Shi et al.
2013; Ursini et al. 2014; Weir et al. 2012; Yin et al. 2012).
The cytotoxicity and genotoxicity potential of titanium oxide
nanoparticles has been investigated both in vitro and in vivo
studies; however, the toxicological data are conflicting (Ranjan and Ramalingam 2016). Some studies in the literature
observed no or low toxic effects (Ammendolia et al. 2017;
Di Bucchianico et al. 2017); on the contrary, other studies
have reported that titanium dioxide nanoparticles induce,
cytotoxic, genotoxic and oxidative effects through oxidants
generation, inflammation and apoptosis (Proquin et al. 2017;
Rizk et al. 2017; Shi et al. 2010; Ursini et al. 2014; Yin et al.
2012). According to the International Agency for Research
on Cancer, titanium dioxide nanoparticles are categorized as
a possible carcinogen to humans (IARC 2010). The National
Institute of Occupational Safety and Health also recommends exposure limits of 2.4 mg/m3 for fine titanium dioxide
and 0.3 mg/m3 for ultrafine titanium dioxide (NIOSH 2011).
The phototoxicity of titanium dioxide nanoparticles with
four different sizes (< 25 nm, 31 nm, < 100 nm and 325 nm)
and two crystal forms (anatase and rutile) toward human
skin keratinocytes under ultraviolet radiation was evaluated
by Yin et al. (2012). According to their results, ROS are
generated by the ultraviolet radiation of all titanium dioxide
nanoparticles (anatase and/or mixed anatase/rutile). Moreover, smaller titanium dioxide nanoparticles resulted in higher
Environmental Chemistry Letters
phototoxicity than larger particles. In another study, Proquin
et al. (2017) showed food-grade titanium dioxide is cytotoxic
in human colon carcinoma cell at a lower concentration than
the nano-sized titanium dioxide particles and micro-sized
titanium dioxide particles. However, food-grade titanium
dioxide has no toxic effects in human colon adenocarcinoma
cells cultures at concentrations up to 100 μg/cm2. Gea et al.
(2019) evaluated the cytotoxicity, lactate dehydrogenase
release and genotoxicity of three engineered titanium dioxide nanoparticles with different shapes (bipyramids, rods,
platelets) in comparison with two commercial titanium
dioxide nanoparticles (P25, food grade). Results showed that
the cytotoxicity was overall low and was influenced by the
nanoparticles shape as well as by light exposure. Besides,
no significant lactate dehydrogenase release was detected.
Instead, genotoxicity seemed to be influenced by the cellular
uptake and the aggregation tendency of titanium dioxide
nanoparticles. Moreover, the presence of light enhanced the
genotoxic effect of some nanoparticles, primarily increasing
the oxidative stress. Petersen et al. (2014) demonstrated that
DNA samples incubated in the dark for 24 h with titanium
dioxide nanoparticles (0.5–50 μg/mL) do not lead to the formation of oxidatively induced DNA damage. However, when
the same samples are exposed to either visible light from 400
to 800 nm (energy dose of ~ 14.5 kJ/m) for 24 h or UVA light
at 370 nm for 30 min (energy dose of~ 10 kJ/m2), there is a
significant increase in oxidatively induced DNA damage at
the 50 μg/mL dose for the visible light exposure and a significant increase in oxidatively induced DNA damage at the
5 μg/mL and 50 μg/mL doses for the UVA light exposure.
Moreover, Xin et al. (2019) explored the interactive impacts
of nano-TiO2 and triclosan on green alga Eremosphaera viridis under visible light using Lake Erie water.
Platinum
Even if platinum nanoparticles have proven to be excellent
therapeutic agents in medicine for the treatment of cancer
cells, they seem to produce undesirable side effects (Azmi
and Shad 2017). In one in vitro study, Bendale et al. (2017)
examined the cytotoxic effect of platinum nanoparticles
on human lung adenocarcinoma, ovarian teratocarcinoma,
pancreatic cancer cells and normal peripheral blood mononucleocyte cells and evaluated anticancer potential through
induction of apoptosis on ovarian teratocarcinoma cells.
They found that platinum nanoparticles exerted a cytotoxic
effect on cancer cell lines, whereas no cytotoxic effect was
observed at the highest dose on normal cells. The results
showed that platinum nanoparticles had potent anticancer
activities against ovarian teratocarcinoma cell line via induction of apoptosis and cell cycle arrest. A dose-dependent
cytotoxicity of platinum nanoparticles was also investigated
by Labrador-Rached et al. (2018). Findings revealed that a
high platinum nanoparticles dosage induced cytotoxicity of
human liver cell. However, low-level exposure to platinum
nanoparticles was able to elicit multiple stress responses,
secretion of proinflammatory cytokines and modulation of
insulin-like growth factor-1-dependent signal transduction.
Nevertheless, Gatto et al. (2018) have shown that citratecoated platinum nanoparticles are non-toxic and immunecompatible with monocytes cells. They also observed platinum nanoparticle-mediated ROS reduction and discovered
their ability to modulate gene transcription without alteration of inflammatory cytokine release. Konieczny et al.
(2013) found platinum nanoparticles trigger toxic effects
on primary keratinocytes, decreasing cell metabolism, but
these changes have no effects on cell viability or migration.
Moreover, smaller nanoparticles exhibited more deleterious
effect on DNA stability and higher caspases activation than
the big ones. Samadi et al. (2018) exposed human ovarian
cancer cells to platinum nanoparticles for 24 h at varying
concentrations (0–50 μg/ml) to assess the toxicity of platinum nanoparticles on living cells. They observed no changes
in neither cellular oxidative stress nor apoptotic or necrotic
cell death.
Zinc oxide
Higher chemical activity, extremely strong oxidation and
corrosion resistance, photocatalysis, unique stronger absorption and shielding ability to the ultraviolet rays properties
of zinc oxide nanoparticles enable to the use of these nanoparticles in a broad range of application including catalysis,
paints, wave filters, UV detectors, transparent conductive
films, varistors, gas sensors, solar cells, sunscreens and cosmetic products (Guan et al. 2012; Huang et al. 2010; Subramaniam et al. 2019). Because of zinc oxide nanoparticle’s
wide application in cosmetics and daily care products, both
its exposure and toxicity to humans have increased (Senapati and Kumar 2018). De Berardis et al. (2010) evaluated
the physical properties and the toxicological effects of zinc
oxide nanoparticles. They showed a significant decrease in
cell viability, remarkable morphological changes, apoptosis
induction via ROS production and IL-8 release after treatment with zinc oxide nanoparticles (5 μg/cm2) for 24 h on
human colon carcinoma cells. Besides, higher concentrations (10, 20 and 40 μg/cm2) induced about 98% cytotoxicity, with a cell survival lower than 5%, already after 24 h of
treatment. Guan et al. (2012) also demonstrated that zinc
oxide nanoparticles cause morphological changes, cytotoxicity and oxidative stress to human hepatocyte and human
embryonic kidney cells. They have also observed the DNAdamaging effects for which lipid peroxidation and oxidative
stress may be attributed as the probable causes. Similarly,
Jain et al. (2019) revealed that zinc oxide nanoparticles
induce oxidative stress, which leads to genotoxic insult, and
13
Environmental Chemistry Letters
hypoxanthine-guanine phosphor ribosyl transferase gene
mutation and later on cellular apoptosis in Chinese hamster
lung fibroblast cells.
Ng et al. (2017) investigated the toxicological profiles of
zinc oxide nanoparticles in human lung fibroblasts in vitro
and in vivo models employing the fruit fly Drosophila melanogaster. For in vitro toxicity, the results showed that there
was a significant release of extracellular lactate dehydrogenase and decreased cell viability in zinc oxide nanoparticlestreated lung cells, indicating cellular damage and cytotoxicity. Production of ROS was seen to be identified with the
critical articulation of DNA damage-inducible transcript
and endoplasmic reticulum to nucleus signaling one gene,
which is stress-associated. Oxidative stress-induced DNA
damage was defined by the release of a DNA oxidation product and 8-hydroxydeoxyguanosine. Considering the in vivo
study of the fruit fly D. melanogaster, substantial toxicity
was detected in F1 progenies based on the intake of zinc
oxide nanoparticles, which actuated noteworthy diminishing in the egg-to-adult suitability of the flies. This study
recommended that zinc oxide nanoparticles induce a significant oxidative stress associated with cytotoxicity and
genotoxicity in human lung fibroblasts in vitro and in D.
melanogaster in vivo. Xiao et al. (2016) indicated that zinc
oxide nanoparticles induced kidney toxicity via oxidative
stress, which accompanied with the elevated level of ROS,
malondialdehyde and decreased the level of superoxide dismutase in vitro and in vivo.
Iron oxide
Iron oxide nanoparticles have been widely used for biomedical applications including drug delivery, magnetic resonance
imaging, thermal ablation therapy, in vivo cell tracking and
magnetic separation of cells or molecules fields due to their
superparamagnetic properties (Feng et al. 2018; Naqvi et al.
2010). In addition, high specific surface area and high reactivity of nanoscale zero-valent iron (nZVI) particles lead
to the use of these particles in environmental remediation
(Schmid et al. 2015). As an environmental remediation
using nanofluid prepared by iron(II, III) oxide significantly
increased CO2 absorption from the water (Arshadi et al.
2019). CO2 absorption mechanism was explained under the
grazing (Fig. 4a), and hydrodynamic effect (Fig. 4b). The
grazing effect is related to the absorption of gas molecules
with methods of the nanoparticle surfaces at the bubble
interface and afterward removing the adsorbed gas components from the nanoparticles surface into the fluid.
Hydrodynamic effect of the nanoparticles encompassing the bubbles break the diffusion boundary layer which
prompts a thinner viable layer. Gas diffusion into the liquid
film increments by the presence of the particles near the
interface between the bubble and the liquid which enhances
13
Fig. 4 Absorption mechanism in nanofluidic systems: a grazing effect
or shuttle where the shuttle effect arises through the transport process
of gas by nanoparticles in the small-scale liquid and b hydrodynamic
effect in which the nanoparticles encircling the bubbles break the diffusion boundary layer and then make it thinner. Adapted from Zhang
et al. (2018)
the turbulence of the flow and the mass transfer coefficient.
CO2 absorption by nanofluids was found to be dependent on
many factors including type, size, particle and gas concentration, temperature, flow rates of gas and liquid. Besides
the iron oxide nanoparticles, copper oxide, aluminum oxide,
magnesium oxide, silicon dioxide, titanium dioxide, carbon
nanotube and multiwalled carbon nanotube were used for
CO2 absorption (Zhang et al. 2018).
Additionally, nano zero-valent iron (Fe0) has been used
effectively for remediation of contaminants in soil and
ground water. Zero-valent iron is a reducing agent that
reacts with dissolved oxygen, water and contaminants such
as chlorinated ethenes, chlorinated methanes, brominated
methanes, trihalomethanes, other polychlorinated hydrocarbon pesticides, as well as dyes (Petersen et al. 2012; Sarathy
et al. 2008).
Inhalation of iron oxide nanoparticles was found to
cause oxidative stress by accumulating in the liver, spleen,
lungs and brain with high ROS, which resulted in inflammation, low cell viability, cell lysis and disturbance of the
blood coagulation system (Tan et al. 2018). The most of
the studies investigated size and surface coating-dependent
effects of iron oxide nanoparticles; for example, Naqvi et al.
(2010) analyzed the dose- and time-dependent toxicity of
Environmental Chemistry Letters
superparamagnetic iron oxide nanoparticles with a mean
size of 30 nm coated with Tween 80 surfactant on murine
macrophage cells. Low concentrations of iron oxide nanoparticles (25–200 µg/mL for a two-hour exposure) have
found to exhibit greater cell toxicity than higher concentrations (300–500 µg/mL for a six-hour exposure). Nevertheless, after seven days of incubation, dextran-coated iron
oxide nanoparticles (100–150 nm, 0.1 mg/mL) reduced the
cell viability in human macrophages, about 20%. Other
researchers, Magdolenova et al. (2015) investigated the
potential cytotoxicity and genotoxicity of uncoated and
oleate-coated iron oxide nanoparticles on human lymphoblastoid cells and primary peripheral lymphocytes in vitro.
Uncoated iron oxide nanoparticles were found not to be
cytotoxic or genotoxic, while oleate-coated iron oxide nanoparticles were found to be cytotoxic in a dose-dependent
manner and also induce DNA damage, indicating genotoxic
potential. Moreover, Sadeghi et al. (2015) have studied the
toxicity of iron oxide nanoparticle on human hepatoma cells.
Results showed an increase of the oxidative damage leads
cells to the apoptosis and therefore reduced cell viability
after 12-h and 24-h exposure to 25, 50, 75 and 100 µg/ml
of iron oxide nanoparticle. Feng et al. (2018) investigated
the influence of particle size and surface coating on the biological distribution of iron oxide nanoparticles and their
biological effects in vitro and in vivo. They demonstrated
that both size and coating have a remarkable impact on the
cellular uptake, cytotoxicity, distribution and clearance of
iron oxide nanoparticles. Polyethylenimine-coated iron oxide
nanoparticles have found to exhibit higher cellular uptake
in both macrophages and cancer cells, severe cytotoxicity,
faster clearance and less tumor distribution than polyethylene glycol-coated iron oxide nanoparticles. Besides, smallersized polyethylene glycol-coated iron oxide nanoparticles
(10 nm) showed relatively higher cellular uptake and tumor
accumulation than larger ones (30 nm).
Non-metallic nanoparticles
Silica
Silica nanoparticles, also known as silicone dioxide, have
attracted great attention because of their unique properties
including large surface area-to-volume ratios, tunable pore
size and connectivity, biocompatibility and ease of surface
modification (Geranio et al. 2010; Kim et al. 2015).These
properties make them appealing tools for biomedical and
biotechnological fields, such as medical diagnostics, drug
delivery, gene therapy, biomolecules detection, photodynamic therapy and bioimaging (Duan et al. 2013; Yu et al.
2014). Even though silica nanoparticle was thought to be
vastly biocompatible material for use in biomedical and
biotechnological applications, they could be accumulated
and retained in the heart, liver, spleen, kidney and brain
after ingestion, inhalation or skin application (Zhou et al.
2019). Recent studies have been also indicated that exposure to silica nanoparticles could induce ROS generation and
cause oxidative stress (Athinarayanan et al. 2014; Duan et al.
2013; Zhou et al. 2019). Yu et al. (2014) investigated the
effects and interaction mechanisms of ROS and autophagy
triggered by silica nanoparticles. Autophagy and autophagic
cell death were induced as a result of ROS generation in
human heptacellular carcinoma cells after exposed to the
silica nanoparticles. Wang et al. (2018) have reported that
four sizes (10, 25, 50, 100 nm) of silica nanoparticles can
cause apoptosis to human umbilical vein endothelial cells
in culture. Genotoxic potentials of four sizes (10, 25, 50,
100 nm) of silica nanoparticle to human umbilical vein
endothelial cells in culture have determined by Zhou et al.
(2019). The results showed that exposure to silica nanoparticles could induce a significant increase of both DNA damage
and cytokinesis-block micronucleus frequencies in human
umbilical vein endothelial cells. Additionally, increased
levels of ROS decreased levels of reduced glutathione, and
enhanced levels of erythroid 2-related factor 2 protein were
observed in the silica nanoparticles-treated human umbilical
vein endothelial cells.
Quantum dots
The optical fluorescent property of quantum dots can be conjugated by bioactive moieties to target specific biological
events and cellular structures, such as labeling neoplastic
cells, DNA and cell membrane receptors (Liu et al. 2015).
The toxicity of quantum dots relies on many factors, mainly
the result of individual quantum dots physicochemical
properties and environmental settings: size, concentration,
charge, outer coating bioactivity. Also, oxidative, photolytic and mechanical stability appear to be decisive factors
for quantum dots toxicity. According to Havrdova et al.
(2016), significant differences were found in the toxicity
of sulfide quantum dots depending on their surface charge.
Polyethyleneglycol-modified dots with neutral charge did not
induce any abnormalities in cell morphology, intracellular
trafficking, and cell cycle up to concentrations of 300 μg/
mL. Pristine carbon dots with negative charge arrested the
G2/M phase of the cell cycle, stimulated proliferation and
led to higher oxidative stress; however, they did not enter
the cell nucleus. In contrast, positively charged polyethylenimine-coated dots were found to be the most cytotoxic,
entered into the nucleus and induced the largest changes
in the G0/G1 phase of the cell cycle, even at concentrations of around 100 μg/mL. In another research, exposure
to cadmium sulfide quantum dots at low concentrations was
induced only minor damage to nuclear DNA and none to
mitochondrial DNA. However, the stress caused an increase
13
Environmental Chemistry Letters
in the production of ROS, which triggered the mitochondriamediated intrinsic apoptotic pathway involving a cascade
of transcriptomic events, finally prompting the activation
of a rescue pathway (Bali et al. 2020; Paesano et al. 2016).
Peynshaert et al. (2017) reported that 3-mercaptopropioniccoated quantum dots are highly biocompatible, where the
lysosomal activation and ROS reductions were induced by
these quantum dots, and likely rescue the cell from potentially nanomaterial-induced toxic effects. However, polyethylene glycol-coated quantum dots exhibited significant
toxicity owing to their capacity to induce ROS production
and autophagy malfunction through lysosomal impairment.
Fullerene
Non-functionalized fullerenes can be highly distributed in
all tissues, and indicated a long-lasting buildup, which has
been observed in bones, spleen, liver and kidney (Bahadar
et al. 2016; Sergio et al. 2013). An in vitro and in vivo study
of fullerene nanoparticle toxicity was explored by several
researchers. As demonstrated by in vitro study, exposure to
C70 fullerene for 24 h at a concentration of 25.2 µg/mL led to
the induction of intracellular ROS level in human keratinocyte and lung carcinoma cells (Horie et al. 2013). Shipelin
et al. (2015) have investigated the effect of fullerene dispersion on iliac mucosa and liver at doses of 0.1, 1.0 and 10 mg/
kg body weight over 92 days in vivo. Fullerene found to be
toxic to the hepatic tissue at the highest examined doses
of 10 mg/kg body weight. However, in another study, no
significant cytotoxicity was observed upon 24 h exposure to
fullerene derivatives at less than 50 µg/mL. Higher concentration (> 100 µg/mL) of fullerene derivatives with different surface modification found to decrease the proliferation
of macrophage cell and induced cytotoxicity through the
apoptosis pathway (Xiang et al. 2012). Zhang et al. (2015)
also reported that fullerene aggregates have no toxic effect
at concentrations lower than 1.65 mg/L. However, fullerene
aggregates induce cytotoxicity with the increase in concentration through the depletion of the mitochondrial membrane
potential and the increase of intracellular ROS, triggering
apoptosis of macrophage by activation of the mitochondrial
pathway. Additionally, the benthic organism Chironomus
riparius was exposed to C60 fullerene using the environmentally realistic method of allowing the fullerenes to settle
down on the sediment surface (Waissi-Leinonen et al. 2012).
According to light microscopic images, fullerene agglomerates were observed in the gut, but no absorption into the gut
epithelial cells was detected. In the organisms exposed to
fullerenes, microvilli were damaged and were significantly
shorter. The potential toxicity of fullerene to Chironomus
riparius appears to be caused by morphological changes,
inhibiting larval growth.
13
Polymeric material
Polymeric nanoparticles have been considered as excellent
drug carriers in cancer theraphy due to their excellent pharmacokinetic properties such as drug loading, drug release,
structure stability and nanoparticles degradation (Li et al.
2017). These nanoparticles can be also conjugated to or
encapsulated in polymers to improve nanomedicine that
offers sustained release and decent biocompatibility with
cells and tissues (Bahadar et al. 2016). Additionally, most
biodegradable polymeric nanoparticles are known to be nontoxic, non-inflammatory and non-immunologic, and they do
not activate neutrophils (Kumari et al. 2010). Voigt et al.
(2014) nanoparticles injected rhodamine-labeled polybutylcyanoacrylate nanoparticles variations into rats and monitored the survival and morphology of retrogradely labeled
neurons by in vivo confocal neuroimaging for 5 weeks. They
demonstrated that polybutylcyanoacrylate nanoparticles do
not induce neuronal death in pharmacologically effective
concentrations, even when coated with surfactants that are
adverse to viability in cell cultures. They concluded that
polybutylcyanoacrylate nanoparticles are a non-toxic tool
for drug delivery into the central nervous system. On the
contrary, it has been known that positively charged nanoparticles have been shown to be toxic to many different cell
types (Elliott et al. 2017). Grabowski et al. (2015) evaluated the toxicity of poly(lactide-co-glycolic)-based nanoparticles (stabilizer-free or associated with stabilizers) on
human-like THP-1 macrophages. At concentrations below
0.1 mg/mL, poly(lactide-co-glycolic)-based nanoparticles
showed few signs of toxicity, either in mitochondrial activity, induction of apoptosis/necrosis or production of intracellular ROS and secretions of pro-inflammatory cytokines.
At high concentrations (above 1 mg/mL), cytotoxicity was
found to be higher with the presence of stabilizers, whereas
the stabilizer-free poly(lactide-co-glycolic)-based nanoparticles showed no cytotoxicity. Therefore, surface coating
can significantly affect the cytotoxicity of nanoparticles by
changing their physicochemical properties and can drastically alter the pharmacokinetics, distribution, accumulation
and toxicity of nanoparticles (Gatoo et al. 2014).
Conclusion
Improvement in nanotechnology has been viewed as one of
the original bearings in current technological advancements
in many industrial fields. Nevertheless, the danger of contaminating the earth and the conceivable adverse effects on
human health must be taken into consideration. In summary,
the discussion here has focused on the toxicity and behavior
of various classes of nanoparticles in the environment. Based
on previous studies, these nanoparticles were found to be
Environmental Chemistry Letters
toxic; however, the reason for their toxicity is undetermined.
There is still an unclear relationship between nanoparticles
and their surroundings. As a future work, more investigations are expected to assess the safety of complex multi-part
and functional nanomaterials, and researchers should create validated models equipped of release, transport, transformation, accumulation, as well as uptake of engineered
nanomaterials in the environment. These models ought to
relate physical and chemical attributes of nanomaterials to
their manner, enable a coordinated way to deal with potential
effect of engineered nanomaterials and nanoproducts, and
predict impacts in subjected population. Also, carrying out
research on nanotechnology risks and advantages outside the
scientific community is difficult and, however, is fundamental for dialogs dependent on the sound science. This implies
creating communication exercises that empower fundamental data to be summarized, investigated and eventually incorporated for different invested individuals, including decision makers and buyers. At last, a worldwide understanding
of nanotechnology-explicit risks is basic if big and small
ventures are to work on a level playing field and creating
economies on structuring safe nanotechnologies. Regardless of evolvement reviewed here, many research obstacles
within the area must be addressed. Training in nanotechnology and sharing continuous knowledge to a wider audience
are extremely important future endeavors.
References
Ahamed M, Siddiqui M, Akhtar M, Ahmad I, Pant A, Alhadlaq H
(2010) Genotoxic potential of copper oxide nanoparticles in
human lung epithelial cells. Biochem Biophys Res Commun
396(2):578–583. https://doi.org/10.1016/j.bbrc.2010.04.156
Akter M, Sikder MT, Rahman MM, Ullah AA, Hossain KFB, Banik S,
Hosokawa T, Saito T et al (2018) A systematic review on silver
nanoparticles-induced cytotoxicity: physicochemical properties
and perspectives. J Adv Res 9:1–16. https://doi.org/10.1016/j.
jare.2017.10.008
Alshatwi AA, Vaiyapuri Subbarayan P, Ramesh E, Al-Hazzani AA,
Alsaif MA, Alwarthan AA (2012) Al2O3 nanoparticles induce
mitochondria-mediated cell death and upregulate the expression
of signaling genes in human mesenchymal stem cells. J Biochem
Mol Toxicol 26(11):469–476. https://doi.org/10.1002/jbt.21448
Ammendolia MG, Iosi F, Maranghi F, Tassinari R, Cubadda F, Aureli
F, Raggi A, Superti F et al (2017) Short-term oral exposure to
low doses of nano-sized TiO2 and potential modulatory effects
on intestinal cells. Food Chem Toxicol 102:63–75. https://doi.
org/10.1016/j.fct.2017.01.031
Annangi B, Bach J, Vales G, Rubio L, Marcos R, Hernandez A (2015)
Long-term exposures to low doses of cobalt nanoparticles induce
cell transformation enhanced by oxidative damage. Nanotoxicology 9(2):138–147. https://doi.org/10.3109/17435390.2014.90058
2
Ansari SM, Bhor RD, Pai KR, Sen D, Mazumder S, Ghosh K, Kolekar
YD, Ramana CV (2017) Cobalt nanoparticles for biomedical
applications: facile synthesis, physiochemical characterization,
cytotoxicity behavior and biocompatibility. Appl Surf Sci
414:171–187. https://doi.org/10.1016/j.apsusc.2017.03.002
Anwar A, Numan A, Siddiqui R, Khalid M, Khan NA (2019) Cobalt
nanoparticles as novel nanotherapeutics against Acanthamoeba
castellanii. Parasit Vectors 12(1):280. https://doi.org/10.1186/
s13071-019-3528-2
Arshadi M, Taghvaei H, Abdolmaleki M, Lee M, Eskandarloo H,
Abbaspourrad A (2019) Carbon dioxide absorption in water/
nanofluid by a symmetric amine-based nanodendritic adsorbent.
Appl Energy 242:1562–1572
Aschberger K, Johnston H, Stone V, Aitken R, Tran C, Hankin S,
Peters S, Christensen F (2010) Review of fullerene toxicity and
exposure-appraisal of a human health risk assessment, based on
open literature. Regul Toxicol Pharmacol 58(3):455–473. https
://doi.org/10.1016/j.yrtph.2010.08.017
Asmatulu RE (2013) Nanotechnology safety, 1st edn. Elsevier,
Amsterdam
Asmatulu R, Nguyen P, Asmatulu E (2013) Nanotechnology safety in
the automotive industry. In: Khan WS, Asmatulu R (eds) Nanotechnology safety. Elsevier, Amsterdam, pp 57–72
Athinarayanan J, Periasamy VS, Alsaif MA, Al-Warthan AA, Alshatwi
AA (2014) Presence of nanosilica (E551) in commercial food
products: Tnf-mediated oxidative stress and altered cell cycle
progression in human lung fibroblast cells. Cell Biol Toxicol
30(2):89–100. https://doi.org/10.1007/s10565-014-9271-8
Azmi M, Shad K (2017) Role of nanostructure molecules in enhancing the bioavailability of oral drugs. In: Grumezescu DFA (ed)
Nanostructures for novel therapy, 1st edn. Elsevier, Amsterdam,
pp 375–407
Bahadar H, Maqbool F, Niaz K, Abdollahi M (2016) Toxicity of nanoparticles and an overview of current experimental models. Iran
Biomed J. https://doi.org/10.7508/ibj.2016.01.001
Bali AS, Sidhu GPS, Kumar V (2020) Root exudates ameliorate cadmium tolerance in plants: a review. Environ Chem Lett. https://
doi.org/10.1007/s10311-020-01012-x
Bendale Y, Bendale V, Paul S (2017) Evaluation of cytotoxic activity
of platinum nanoparticles against normal and cancer cells and its
anticancer potential through induction of apoptosis. Integr Med
Res 6(2):141–148. https://doi.org/10.1016/j.imr.2017.01.006
Buzea C, Pacheco I, Robbie K (2007) Nanomaterials and nanoparticles: sources and toxicity. Biointerphases 2: Mr17–Mr71. arXiv
preprint arXiv:0801.3280
Cameron SJ, Hosseinian F, Willmore WG (2018) A current overview
of the biological and cellular effects of nanosilver. Int J Mol Sci.
https://doi.org/10.3390/ijms19072030
Cappellini F, Hedberg Y, McCarrick S, Hedberg J, Derr R, Hendriks G,
Odnevall Wallinder I, Karlsson HL (2018) Mechanistic insight
into reactivity and (geno) toxicity of well-characterized nanoparticles of cobalt metal and oxides. Nanotoxicology 12(6):602–620.
https://doi.org/10.1080/17435390.2018.1470694
Chattopadhyay S, Dash SK, Tripathy S, Das B, Mandal D, Pramanik
P, Roy S (2015) Toxicity of cobalt oxide nanoparticles to normal
cells; an in vitro and in vivo study. Chem Biol Interact 226:58–
71. https://doi.org/10.1016/j.cbi.2014.11.016
Chen LQ, Fang L, Ling J, Ding CZ, Kang B, Huang CZ (2015) Nanotoxicity of silver nanoparticles to red blood cells: size dependent
adsorption, uptake, and hemolytic activity. Chem Res Toxicol
28(3):501–509. https://doi.org/10.1021/tx500479m
Ciucă AG, Grecu CI, Rotărescu P, Gheorghe I, Bolocan A, Grumezescu AM, Holban AM, Andronescu E (2017) Nanostructures for
drug delivery: pharmacokinetic and toxicological aspects. In:
Andronescu E, Grumezescu AM (eds) Nanostructures for drug
delivery, 1st edn. Elsevier, Amsterdam, pp 941–957
Dasgupta N, Ramalingam C (2016) Silver nanoparticle antimicrobial
activity explained by membrane rupture and reactive oxygen
13
Environmental Chemistry Letters
generation. Environ Chem Lett 14(4):477–485. https ://doi.
org/10.1007/s10311-016-0583-1
De Berardis B, Civitelli G, Condello M, Lista P, Pozzi R, Arancia G,
Meschini S (2010) Exposure to Zno nanoparticles induces oxidative stress and cytotoxicity in human colon carcinoma cells. Toxicol Appl Pharmacol 246(3):116–127. https://doi.org/10.1016/j.
taap.2010.04.012
Di Bucchianico S, Cappellini F, Le Bihanic F, Zhang Y, Dreij K, Karlsson HL (2017) Genotoxicity of TiO2 nanoparticles assessed by
mini-gel comet assay and micronucleus scoring with flow cytometry. Mutagenesis 32(1):127–137. https://doi.org/10.1093/mutag
e/gew030
Di Virgilio AL, Reigosa M, Arnal PM, Lorenzo F, de Mele M (2010)
Comparative study of the cytotoxic and genotoxic effects of titanium oxide and aluminium oxide nanoparticles in chinese hamster ovary (Cho-K1) cells. J Hazard Mater 177(1–3):711–718.
https://doi.org/10.1016/j.jhazmat.2009.12.089
Donmez Gungunes C, Seker S, Elcin AE, Elcin YM (2017) A comparative study on the in vitro cytotoxic responses of two mammalian cell types to fullerenes, carbon nanotubes and iron oxide
nanoparticles. Drug Chem Toxicol 40(2):215–227. https://doi.
org/10.1080/01480545.2016.1199563
Duan J, Yu Y, Li Y, Yu Y, Li Y, Zhou X, Huang P, Sun Z (2013) Toxic
effect of silica nanoparticles on endothelial cells through DNA
damage response via Chk1-dependent G2/M checkpoint. PLoS
ONE 8(4):e62087. https://doi.org/10.1371/journal.pone.0062087
Elliott JT, Rosslein M, Song NW, Toman B, Kinsner-Ovaskainen A,
Maniratanachote R, Salit ML, Petersen EJ et al (2017) Toward
achieving harmonization in a nano-cytotoxicity assay measurement through an interlaboratory comparison study. Altex
34(2):201–218. https://doi.org/10.14573/altex.1605021
Ema M, Hougaard KS, Kishimoto A, Honda K (2016) Reproductive
and developmental toxicity of carbon-based nanomaterials: a
literature review. Nanotoxicology 10(4):391–412. https://doi.
org/10.3109/17435390.2015.1073811
EPA U (2017) Technical fact sheet: nanomaterials
Fard J, Jafari S, Eghbal M (2015) A review of molecular mechanisms
involved in toxicity of nanoparticles. Adv Pharm Bull 5(4):447–
454. https://doi.org/10.15171/apb.2015.061
Farré M, Barceló D (2012) Introduction to the analysis and risk of
nanomaterials in environmental and food samples. In: Barcelo
D, Farre M (eds) Comprehensive analytical chemistry, vol 59,
1st edn. Elsevier, Amsterdam, pp 1–32
Feng Q, Liu Y, Huang J, Chen K, Huang J, Xiao K (2018) Uptake,
distribution, clearance, and toxicity of iron oxide nanoparticles
with different sizes and coatings. Sci Rep 8(1):2082. https://doi.
org/10.1038/s41598-018-19628-z
Foldbjerg R, Dang DA, Autrup H (2011) Cytotoxicity and genotoxicity of silver nanoparticles in the human lung cancer cell line,
A549. Arch Toxicol 85(7):743–750. https ://doi.org/10.1007/
s00204-010-0545-5
Fu P, Xia Q, Hwang H, Ray P, Yu H (2014) Mechanisms of nanotoxicity: generation of reactive oxygen species. J Food Drug Anal
22(1):64–75. https://doi.org/10.1016/j.jfda.2014.01.005
Future Markets I (2013) The global market for aluminium oxide nanoparticles (76). Retrieved from http://www.nanotechmag.com/
wp-content/uploads/2014/09/ALUMINIUM-OXIDE-NANOP
ARTICLES-2013-.pdf
Gatoo MA, Naseem S, Arfat MY, Mahmood Dar A, Qasim K, Zubair S
(2014) Physicochemical properties of nanomaterials: implication
in associated toxic manifestations. Biomed Res Int 2014:498420.
https://doi.org/10.1155/2014/498420
Gatto F, Moglianetti M, Pompa PP, Bardi G (2018) Platinum nanoparticles decrease reactive oxygen species and modulate gene
expression without alteration of immune responses in THP-1
13
monocytes. Nanomaterials 8(6):392. https ://doi.org/10.3390/
nano8060392
Gea M, Bonetta S, Iannarelli L, Giovannozzi AM, Maurino V, Bonetta
S, Hodoroaba VD, Armato C et al (2019) Shape-engineered
titanium dioxide nanoparticles (TiO2-Nps): cytotoxicity and
genotoxicity in bronchial epithelial cells. Food Chem Toxicol
127:89–100. https://doi.org/10.1016/j.fct.2019.02.043
Geranio L, Hommes G, Shahgaldian P, Wirth-Heller A, Pieles U, Corvini PF-X (2010) Radio (14C)-and fluorescent-doubly labeled
silica nanoparticles for biological and environmental toxicity
assessment. Environ Chem Lett 8(3):247–251
Gliga A, Skoglund S, Wallinder I, Fadeel B, Karlsson H (2014)
Size-dependent cytotoxicity of silver nanoparticles in
human lung cells: the role of cellular uptake, agglomeration and Ag release. Part Fibre Toxicol 11(1):11. https ://doi.
org/10.1186/1743-8977-11-11
Grabowski N, Hillaireau H, Vergnaud J, Tsapis N, Pallardy M, KerdineRomer S, Fattal E (2015) Surface coating mediates the toxicity
of polymeric nanoparticles towards human-like macrophages.
Int J Pharm 482(1–2):75–83. https ://doi.org/10.1016/j.ijpha
rm.2014.11.042
Guan R, Kang T, Lu F, Zhang Z, Shen H, Liu M (2012) Cytotoxicity, oxidative stress, and genotoxicity in human hepatocyte and
embryonic kidney cells exposed to Zno nanoparticles. Nanoscale
Res Lett 7(1):602. https://doi.org/10.1186/1556-276X-7-602
Guerra FD, Attia MF, Whitehead DC, Alexis F (2018) Nanotechnology for environmental remediation: materials and applications.
Molecules 23(7):1760
Guo Z, Zeng G, Cui K, Chen A (2018) Toxicity of environmental
nanosilver: mechanism and assessment. Environ Chem Lett
17(1):319–333. https://doi.org/10.1007/s10311-018-0800-1
Gurunathan S, Kim J (2016) Synthesis, toxicity, biocompatibility,
and biomedical applications of graphene and graphene-related
materials. Int J Nanomed 11:1927. https://doi.org/10.2147/IJN.
S105264
Haase A, Tentschert J, Jungnickel H, Graf P, Mantion A, Draude F,
Plendl J, Goetz M, et al (2011) Toxicity of silver nanoparticles
in human macrophages: uptake, intracellular distribution and
cellular responses. In: Paper presented at the Journal of Physics:
Conference Series
Hanna SK, Montoro Bustos AR, Peterson AW, Reipa V, Scanlan LD,
Hosbas Coskun S, Cho TJ, Johnson ME et al (2018) Agglomeration of Escherichia Coli with positively charged nanoparticles
can lead to artifacts in a standard caenorhabditis elegans toxicity assay. Environ Sci Technol 52(10):5968–5978. https://doi.
org/10.1021/acs.est.7b06099
Havrdova M, Hola K, Skopalik J, Tomankova K, Petr M, Cepe K,
Polakova K, Tucek J et al (2016) Toxicity of carbon dots: effect
of surface functionalization on the cell viability, reactive oxygen
species generation and cell cycle. Carbon 99:238–248. https://
doi.org/10.1016/j.carbon.2015.12.027
He X, Aker WG, Fu PP, Hwang H-M (2015) Toxicity of engineered
metal oxide nanomaterials mediated by Nano–Bio–Eco–interactions: a review and perspective. Environ Sci Nano 2(6):564–582.
https://doi.org/10.1039/c5en00094g
Horie M, Nishio K, Kato H, Shinohara N, Nakamura A, Fujita K, Kinugasa S, Endoh S et al (2013) In vitro evaluation of cellular influences induced by stable fullerene C(7)(0) medium dispersion:
induction of cellular oxidative stress. Chemosphere 93(6):1182–
1188. https://doi.org/10.1016/j.chemosphere.2013.06.067
Huang C, Aronstam R, Chen D, Huang Y (2010) Oxidative stress,
calcium homeostasis, and altered gene expression in human lung
epithelial cells exposed to Zno nanoparticles. Toxicol In Vitro
24(1):45–55. https://doi.org/10.1016/j.tiv.2009.09.007
IARC (2010) Carbon black, titanium dioxide, and talc, vol 93. IARC
Press, International Agency for Research on Cancer, Lyon
Environmental Chemistry Letters
ISO (2015) Vocabulary: part 1—core terms. In: International Standardisation Organisation (ISO), Technical Specification ISO/TS
Jain AK, Singh D, Dubey K, Maurya R, Pandey AK (2019) Zinc oxide
nanoparticles induced gene mutation at the HGPRT locus and
cell cycle arrest associated with apoptosis in V-79 cells. J Appl
Toxicol 39(5):735–750. https://doi.org/10.1002/jat.3763
Jeevanandam J, Barhoum A, Chan Y, Dufresne A, Danquah M (2018)
Review on nanoparticles and nanostructured materials: history, sources, toxicity and regulations. Beilstein J Nanotechnol
9:1050–1074. https://doi.org/10.3762/bjnano.9.98
Jia Y-P, Ma B-Y, Wei X-W, Qian Z-Y (2017) The in vitro and in vivo
toxicity of gold nanoparticles. Chin Chem Lett 28(4):691–702.
https://doi.org/10.1016/j.cclet.2017.01.021
Kahraman E, Güngör S, Özsoy Y (2017) Potential enhancement and
targeting strategies of polymeric and lipid-based nanocarriers in
dermal drug delivery. Ther Deliv 8(11):967–985
Kaphle A, Navya PN, Umapathi A, Daima HK (2018) Nanomaterials for agriculture, food and environment: applications. Toxicity and regulation. Environ Chem Lett 16(1):43–58. https://doi.
org/10.1007/s10311-017-0662-y
Kim I-S, Baek M, Choi S-J (2010) Comparative cytotoxicity of Al2O3,
CeO2, TiO2 and ZnO nanoparticles to human lung cells. J
Nanosci Nanotechnol 10(5):3453–3458. https://doi.org/10.1166/
jnn.2010.2340
Kim IY, Joachim E, Choi H, Kim K (2015) Toxicity of silica nanoparticles depends on size, dose, and cell type. Nanomedicine
11(6):1407–1416. https://doi.org/10.1016/j.nano.2015.03.004
Konieczny P, Goralczyk AG, Szmyd R, Skalniak L, Koziel J, Filon FL,
Crosera M, Cierniak A et al (2013) Effects triggered by platinum
nanoparticles on primary keratinocytes. Int J Nanomed 8:3963–
3975. https://doi.org/10.2147/IJN.S49612
Kumar H, Venkatesh N, Bhowmik H, Kuila A (2018) Metallic nanoparticle: a review. Biomed J Sci Techn Res 4(2):3765–3775
Kumari A, Yadav SK, Yadav SC (2010) Biodegradable polymeric nanoparticles based drug delivery systems. Colloids Surf B Biointerfaces 75(1):1–18. https://doi.org/10.1016/j.colsurfb.2009.09.001
Labrador-Rached CJ, Browning RT, Braydich-Stolle LK, Comfort
KK (2018) Toxicological implications of platinum nanoparticle exposure: stimulation of intracellular stress, inflammatory
response, and akt signaling in vitro. J Toxicol 2018:1367801.
https://doi.org/10.1155/2018/1367801
Lamberti M, Zappavigna S, Sannolo N, Porto S, Caraglia M (2014)
Advantages and risks of nanotechnologies in cancer patients
and occupationally exposed workers. Expert Opin Drug Deliv
11(7):1087–1101
Li JJ, Hartono D, Ong C-N, Bay B-H, Yung L-YL (2010) Autophagy
and oxidative stress associated with gold nanoparticles. Biomaterials 31(23):5996–6003. https://doi.org/10.1016/j.biomateria
ls.2010.04.014
Li B, Li Q, Mo J, Dai H (2017) Drug-loaded polymeric nanoparticles
for cancer stem cell targeting. Front Pharmacol 8:51. https://doi.
org/10.3389/fphar.2017.00051
Liu G, Gao J, Ai H, Chen X (2013) Applications and potential toxicity
of magnetic iron oxide nanoparticles. Small 9(9–10):1533–1545.
https://doi.org/10.1002/smll.201201531
Liu Q, Li H, Xia Q, Liu Y, Xiao K (2015) Role of surface charge in
determining the biological effects of Cdse/Zns quantum dots. Int
J Nanomed 10:7073–7088. https://doi.org/10.2147/IJN.S94543
Lopez-Chaves C, Soto-Alvaredo J, Montes-Bayon M, Bettmer J, Llopis
J, Sanchez-Gonzalez C (2018) Gold nanoparticles: distribution,
bioaccumulation and toxicity. In vitro and in vivo studies. Nanomedicine 14(1):1–12. https://doi.org/10.1016/j.nano.2017.08.011
Magdolenova Z, Drlickova M, Henjum K, Runden-Pran E, Tulinska
J, Bilanicova D, Pojana G, Kazimirova A et al (2015) Coatingdependent induction of cytotoxicity and genotoxicity of iron
oxide nanoparticles. Nanotoxicology 9(Suppl 1):44–56. https://
doi.org/10.3109/17435390.2013.847505
Ma-Hock L, Strauss V, Treumann S, Küttler K, Wohlleben W, Hofmann T, Gröters S, Wiench K et al (2013) Comparative inhalation toxicity of multi-wall carbon nanotubes, graphene, graphite
nanoplatelets and low surface carbon black. Part Fibre Toxicol
10(1):23. https://doi.org/10.1186/1743-8977-10-23
Manke A, Wang L, Rojanasakul Y (2013) Mechanisms of nanoparticle-induced oxidative stress and toxicity. Biomed Res Int
2013:942916. https://doi.org/10.1155/2013/942916
Matea CT, Mocan T, Tabaran F, Pop T, Mosteanu O, Puia C, Iancu C,
Mocan L (2017) Quantum dots in imaging, drug delivery and
sensor applications. Int J Nanomed 12:5421–5431. https://doi.
org/10.2147/IJN.S138624
McWilliams A (2016) The maturing nanotechnology market: products
and applications. BCC Research, Wellesley
Mengesha AE, Youan BBC (2013) Nanodiamonds for drug delivery
systems. In: Narayan R (ed) Diamond-based materials for biomedical applications. Woodhead Publishing, Cambridge, pp
186–205
Ming Z, Feng S, Yilihamu A, Ma Q, Yang S, Yang ST (2018) Toxicity
of pristine and chemically functionalized fullerenes to white rot
fungus phanerochaete chrysosporium. Nanomaterials (Basel).
https://doi.org/10.3390/nano8020120
Mukherjee S, Sau S, Madhuri D, Bollu VS, Madhusudana K, Sreedhar
B, Banerjee R, Patra CR (2016) Green synthesis and characterization of monodispersed gold nanoparticles: toxicity study,
delivery of doxorubicin and its bio-distribution in mouse model.
J Biomed Nanotechnol 12(1):165–181
Mulenos MR, Liu J, Lujan H, Guo B, Lichtfouse E, Sharma VK,
Sayes CM (2020) Copper, silver, and titania nanoparticles do not
release ions under anoxic conditions and release only minute ion
levels under oxic conditions in water: evidence for the low toxicity of nanoparticles. Environ Chem Lett. https://doi.org/10.1007/
s10311-020-00985-z
Naqvi S, Samim M, Abdin M, Ahmed FJ, Maitra A, Prashant C, Dinda
AK (2010) Concentration-dependent toxicity of iron oxide nanoparticles mediated by increased oxidative stress. Int J Nanomed
5:983–989. https://doi.org/10.2147/IJN.S13244
Negahdary M, Chelongar R, Zadeh SK, Ajdary M (2015) The antioxidant effects of silver, gold, and zinc oxide nanoparticles on
male mice in in vivo condition. Adv Biomed Res. https://doi.
org/10.4103/2277-9175.153893
Nelson BC, Petersen EJ, Marquis BJ, Atha DH, Elliott JT, Cleveland
D, Watson SS, Tseng I-H et al (2013) Nist gold nanoparticle
reference materials do not induce oxidative DNA damage.
Nanotoxicology 7(1):21–29. https ://doi.org/10.3109/17435
390.2011.626537
Ng CT, Yong LQ, Hande MP, Ong CN, Yu LE, Bay BH, Baeg GH
(2017) Zinc oxide nanoparticles exhibit cytotoxicity and genotoxicity through oxidative stress responses in human lung fibroblasts and drosophila melanogaster. Int J Nanomed 12:1621.
https://doi.org/10.2147/IJN.S124403
NIOSH (2011) Occupational exposure to titanium dioxide. In: Current intelligence bulletin, 63. National Institute for Occupational
Safety and Health, Cincinnati. Retrieved from
NTP (2016) Report on carcinogens monograph on cobalt and cobalt
compounds that release cobalt ions in vivo. Retrieved from
Paesano L, Perotti A, Buschini A, Carubbi C, Marmiroli M, Maestri E, Iannotta S, Marmiroli N (2016) Markers for toxicity to
Hepg2 exposed to cadmium sulphide quantum dots; damage to
mitochondria. Toxicology 374:18–28. https://doi.org/10.1016/j.
tox.2016.11.012
Pakrashi S, Dalai S, Sabat D, Singh S, Chandrasekaran N, Mukherjee
A (2011) Cytotoxicity of Al2O3 nanoparticles at low exposure
13
Environmental Chemistry Letters
levels to a freshwater bacterial isolate. Chem Res Toxicol
24(11):1899–1904. https://doi.org/10.1021/tx200244g
Patel KD, Singh RK, Kim H-W (2019) Carbon-based nanomaterials
as an emerging platform for theranostics. Mater Horiz 6(3):434–
469. https://doi.org/10.1039/c8mh00966j
Petersen EJ, Pinto RA, Shi X, Huang Q (2012) Impact of size and
sorption on degradation of trichloroethylene and polychlorinated biphenyls by nano-scale zerovalent iron. J Hazard Mater
243:73–79
Petersen EJ, Reipa V, Watson SS, Stanley DL, Rabb SA, Nelson BC
(2014) DNA damaging potential of photoactivated P25 titanium
dioxide nanoparticles. Chem Res Toxicol 27(10):1877–1884.
https://doi.org/10.1021/tx500340v
Peynshaert K, Soenen SJ, Manshian BB, Doak SH, Braeckmans K, De
Smedt SC, Remaut K (2017) Coating of quantum dots strongly
defines their effect on lysosomal health and autophagy. Acta Biomater 48:195–205. https://doi.org/10.1016/j.actbio.2016.10.022
Poborilova Z, Opatrilova R, Babula P (2013) Toxicity of aluminium
oxide nanoparticles demonstrated using a by-2 plant cell suspension culture model. Environ Exp Bot 91:1–11. https://doi.
org/10.1016/j.envexpbot.2013.03.002
Proquin H, Rodriguez-Ibarra C, Moonen CG, Urrutia Ortega IM,
Briede JJ, de Kok TM, van Loveren H, Chirino YI (2017) Titanium dioxide food additive (E171) induces ros formation and
genotoxicity: contribution of micro and nano-sized fractions.
Mutagenesis 32(1):139–149. https ://doi.org/10.1093/mutage/
gew051
Pulit-Prociak J, Stokłosa K, Banach M (2014) Nanosilver products
and toxicity. Environ Chem Lett 13(1):59–68. https ://doi.
org/10.1007/s10311-014-0490-2
Rajiv S, Jerobin J, Saranya V, Nainawat M, Sharma A, Makwana P,
Gayathri C, Bharath L et al (2016) Comparative cytotoxicity
and genotoxicity of cobalt (Ii, Iii) oxide, iron (Iii) oxide, silicon dioxide, and aluminum oxide nanoparticles on human lymphocytes in vitro. Hum Exp Toxicol 35(2):170–183. https://doi.
org/10.1177/0960327115579208
Ranjan S, Ramalingam C (2016) Titanium dioxide nanoparticles induce
bacterial membrane rupture by reactive oxygen species generation. Environ Chem Lett 14(4):487–494. https://doi.org/10.1007/
s10311-016-0586-y
Rizk MZ, Ali SA, Hamed MA, El-Rigal NS, Aly HF, Salah HH (2017)
Toxicity of titanium dioxide nanoparticles: effect of dose and
time on biochemical disturbance, oxidative stress and genotoxicity in mice. Biomed Pharmacother 90:466–472. https://doi.
org/10.1016/j.biopha.2017.03.089
Roy A, Bhattacharya J (2015) Nanotechnology in industrial wastewater
treatment. IWA Publishing, London
Sadeghi L, Tanwir F, Yousefi Babadi V (2015) In vitro toxicity of
iron oxide nanoparticle: oxidative damages on Hep G2 cells.
Exp Toxicol Pathol 67(2):197–203. https://doi.org/10.1016/j.
etp.2014.11.010
Sahu D, Kannan G, Vijayaraghavan R (2014) Carbon black particle
exhibits size dependent toxicity in human monocytes. Int J
Inflamm. https://doi.org/10.1155/2014/827019
Sajid M, Ilyas M, Basheer C, Tariq M, Daud M, Baig N, Shehzad
F (2015) Impact of nanoparticles on human and environment:
review of toxicity factors, exposures, control strategies, and
future prospects. Environ Sci Pollut Res Int 22(6):4122–4143.
https://doi.org/10.1007/s11356-014-3994-1
Samadi A, Klingberg H, Jauffred L, Kjaer A, Bendix PM, Oddershede
LB (2018) Platinum nanoparticles: a non-toxic, effective and
thermally stable alternative plasmonic material for cancer therapy and bioengineering. Nanoscale 10(19):9097–9107. https://
doi.org/10.1039/c8nr02275e
13
Sandeep KV (2013) Nanomaterials-based health care and bioanalytical
applications: trend and prospects. J Nanomater Mol Nanotechnol
2(2):2–6. https://doi.org/10.4172/2324-8777.1000109
Sarathy V, Tratnyek PG, Nurmi JT, Baer DR, Amonette JE, Chun CL,
Penn RL, Reardon EJ (2008) Aging of iron nanoparticles in aqueous solution: effects on structure and reactivity. J Phys Chem C
112(7):2286–2293
Sardoiwala MN, Kaundal B, Roy Choudhury S (2018) Development of
engineered nanoparticles expediting diagnostic and therapeutic
applications across blood–brain barrier. In: Hussain CM (ed)
Handbook of nanomaterials for industrial applications, 1st edn.
Elsevier, Amsterdam, pp 696–709
Scherer MD, Sposito JCV, Falco WF, Grisolia AB, Andrade LHC,
Lima SM, Machado G, Nascimento VA et al (2019) Cytotoxic and genotoxic effects of silver nanoparticles on meristematic cells of Allium cepa roots: a close analysis of particle
size dependence. Sci Total Environ 660:459–467. https://doi.
org/10.1016/j.scitotenv.2018.12.444
Schmid D, Micić V, Laumann S, Hofmann T (2015) Measuring the
reactivity of commercially available zero-valent iron nanoparticles used for environmental remediation with iopromide. J Contam Hydrol 181:36–45
Senapati VA, Kumar A (2018) Zno nanoparticles dissolution, penetration and toxicity in human epidermal cells. Influence of pH.
Environ Chem Lett 16(3):1129–1135
Senut MC, Zhang Y, Liu F, Sen A, Ruden DM, Mao G (2016) Sizedependent toxicity of gold nanoparticles on human embryonic
stem cells and their neural derivatives. Small 12(5):631–646.
https://doi.org/10.1002/smll.201502346
Sergio M, Behzadi H, Otto A, van der Spoel D (2013) Fullerenes toxicity and electronic properties. Environ Chem Lett 11(2):105–118
Shi Y, Wang F, He J, Yadav S, Wang H (2010) Titanium dioxide nanoparticles cause apoptosis in BEAS-2b cells through the caspase 8/T-Bid-independent mitochondrial pathway. Toxicol Lett
196(1):21–27. https://doi.org/10.1016/j.toxlet.2010.03.014
Shi H, Magaye R, Castranova V, Zhao J (2013) Titanium dioxide nanoparticles: a review of current toxicological data. Part Fibre Toxicol. https://doi.org/10.1186/1743-8977-10-15
Shin SW, Song IH, Um SH (2015) Role of physicochemical properties
in nanoparticle toxicity. Nanomaterials (Basel) 5(3):1351–1365.
https://doi.org/10.3390/nano5031351
Shipelin VA, Smirnova TA, Gmoshinskii IV, Tutelyan VA (2015)
Analysis of toxicity biomarkers of fullerene C(6)(0) nanoparticles by confocal fluorescent microscopy. Bull Exp Biol Med
158(4):443–449. https://doi.org/10.1007/s10517-015-2781-4
Shvedova A, Pietroiusti A, Fadeel B, Kagan V (2012) Mechanisms of
carbon nanotube-induced toxicity: focus on oxidative stress. Toxicol Appl Pharmacol 261(2):121–133. https://doi.org/10.1016/j.
taap.2012.03.023
Srikanth K, Mahajan A, Pereira E, Duarte AC, Venkateswara Rao J
(2015) Aluminium oxide nanoparticles induced morphological
changes, cytotoxicity and oxidative stress in chinook salmon
(Chse-214) cells. J Appl Toxicol 35(10):1133–1140. https://doi.
org/10.1002/jat.3142
Stark W, Stoessel P, Wohlleben W, Hafner A (2015) Industrial applications of nanoparticles. Chem Soc Rev 44(16):5793–5805. https
://doi.org/10.1039/C4CS00362D
Steckiewicz KP, Barcinska E, Malankowska A, Zauszkiewicz-Pawlak
A, Nowaczyk G, Zaleska-Medynska A, Inkielewicz-Stepniak I
(2019) Impact of gold nanoparticles shape on their cytotoxicity
against human osteoblast and osteosarcoma in in vitro model.
Evaluation of the safety of use and anti-cancer potential. J
Mater Sci Mater Med 30(2):22. https://doi.org/10.1007/s1085
6-019-6221-2
Subramaniam VD, Prasad SV, Banerjee A, Gopinath M, Murugesan R, Marotta F, Sun XF, Pathak S (2019) Health hazards of
Environmental Chemistry Letters
nanoparticles: understanding the toxicity mechanism of nanosized Zno in cosmetic products. Drug Chem Toxicol 42(1):84–
93. https://doi.org/10.1080/01480545.2018.1491987
Sung JH, Ji JH, Park JD, Song MY, Song KS, Ryu HR, Yoon JU,
Jeon KS et al (2011) Subchronic inhalation toxicity of
gold nanoparticles. Part Fibre Toxicol 8(1):16. https ://doi.
org/10.1186/1743-8977-8-16
Tan KX, Barhoum A, Pan S, Danquah MK (2018) Risks and toxicity
of nanoparticles and nanostructured materials. In: Barhoum A,
Makhlouf ASH (eds) Emerging applications of nanoparticles and
architecture nanostructures. Elsevier, Amsterdam, pp 121–139
Thakor AS, Paulmurugan R, Kempen P, Zavaleta C, Sinclair R, Massoud TF, Gambhir SS (2011) Oxidative stress mediates the
effects of Raman-active gold nanoparticles in human cells. Small
7(1):126–136. https://doi.org/10.1002/smll.201001466
Thota S, Crans DC (2018) Metal nanoparticles: synthesis and applications in pharmaceutical sciences. Wiley, New York
Ursini CL, Cavallo D, Fresegna AM, Ciervo A, Maiello R, Tassone P,
Buresti G, Casciardi S et al (2014) Evaluation of cytotoxic, genotoxic and inflammatory response in human alveolar and bronchial
epithelial cells exposed to titanium dioxide nanoparticles. J Appl
Toxicol 34(11):1209–1219. https://doi.org/10.1002/jat.3038
Vazquez-Muñoz R, Borrego B, Juárez-Moreno K, García-García M,
Morales JDM, Bogdanchikova N, Huerta-Saquero A (2017) Toxicity of Silver nanoparticles in biological systems: does the complexity of biological systems matter? Toxicol Lett 276:11–20.
https://doi.org/10.1016/j.toxlet.2017.05.007
Voigt N, Henrich-Noack P, Kockentiedt S, Hintz W, Tomas J, Sabel BA
(2014) Toxicity of polymeric nanoparticles in vivo and in vitro. J
Nanopart Res. https://doi.org/10.1007/s11051-014-2379-1
Waissi-Leinonen GC, Petersen EJ, Pakarinen K, Akkanen J, Leppanen MT, Kukkonen JV (2012) Toxicity of fullerene (C60) to
sediment-dwelling invertebrate chironomus riparius larvae. Environ Toxicol Chem 31(9):2108–2116. https://doi.org/10.1002/
etc.1926
Wan R, Mo Y, Feng L, Chien S, Tollerud DJ, Zhang Q (2012) DNA
damage caused by metal nanoparticles: involvement of oxidative
stress and activation of ATM. Chem Res Toxicol 25(7):1402–
1411. https://doi.org/10.1021/tx200513t
Wan R, Mo Y, Zhang Z, Jiang M, Tang S, Zhang Q (2017) Cobalt
nanoparticles induce lung injury, DNA damage and mutations
in mice. Part Fibre Toxicol 14(1):38. https://doi.org/10.1186/
s12989-017-0219-z
Wang W, Zeng C, Feng Y, Zhou F, Liao F, Liu Y, Feng S, Wang X
(2018) The size-dependent effects of silica nanoparticles on
endothelial cell apoptosis through activating the P53-caspase
pathway. Environ Pollut 233:218–225. https://doi.org/10.1016/j.
envpol.2017.10.053
Weir A, Westerhoff P, Fabricius L, Hristovski K, Von Goetz N (2012)
Titanium dioxide nanoparticles in food and personal care
products. Environ Sci Technol 46(4):2242–2250. https ://doi.
org/10.1021/es204168d
Wu T, Tang M (2018) Review of the effects of manufactured nanoparticles on mammalian target organs. J Appl Toxicol 38(1):25–40.
https://doi.org/10.1002/jat.3499
Xiang K, Dou Z, Li Y, Xu Y, Zhu J, Yang S, Sun H, Liu Y (2012)
Cytotoxicity and Tnf-Α secretion in Raw264.7 macrophages
exposed to different fullerene derivatives. J Nanosci Nanotechnol
12(3):2169–2178. https://doi.org/10.1166/jnn.2012.5681
Xiao L, Liu C, Chen X, Yang Z (2016) Zinc oxide nanoparticles induce
renal toxicity through reactive oxygen species. Food Chem Toxicol 90:76–83. https://doi.org/10.1016/j.fct.2016.02.002
Xin X, Huang G, An C, Feng R (2019) Interactive toxicity of triclosan
and nano-TiO2 to green alga Eremosphaera viridis in Lake Erie:
a new perspective based on fourier transform infrared spectromicroscopy and synchrotron-based X-Ray fluorescence imaging.
Environ Sci Technol 53(16):9884–9894
Yadav HKS, Almokdad AA, Shaluf SIM, Debe MS (2019) Polymerbased nanomaterials for drug-delivery carriers. In: Mohapatra
Shyam, Ranjan Shivendu, Dasgupta Nandita, Kumar Raghvendra, Thomas Sabu (eds) Nanocarriers for drug delivery. Elsevier,
Amsterdam, pp 531–556
Yin JJ, Liu J, Ehrenshaft M, Roberts JE, Fu PP, Mason RP, Zhao B
(2012) Phototoxicity of nano titanium dioxides in hacat keratinocytes-generation of reactive oxygen species and cell damage.
Toxicol Appl Pharmacol 263(1):81–88. https://doi.org/10.1016/j.
taap.2012.06.001
Yu S, Rui Q, Cai T, Wu Q, Li Y, Wang D (2011) Close association of
intestinal autofluorescence with the formation of severe oxidative damage in intestine of nematodes chronically exposed to
Al(2)O(3)-nanoparticle. Environ Toxicol Pharmacol 32(2):233–
241. https://doi.org/10.1016/j.etap.2011.05.008
Yu Y, Duan J, Yu Y, Li Y, Liu X, Zhou X, Ho K-f, Tian L et al (2014)
Silica nanoparticles induce autophagy and autophagic cell
death in Hepg2 cells triggered by reactive oxygen species. J
Hazard Mater 270:176–186. https ://doi.org/10.1016/j.jhazm
at.2014.01.028
Zhang X-D, Wu H-Y, Wu D, Wang Y-Y, Chang J-H, Zhai Z-B, Meng
A-M, Liu P-X et al (2010) Toxicologic effects of gold nanoparticles in vivo by different administration routes. Int J Nanomed
5:771. https://doi.org/10.2147/IJN.S8428
Zhang XQ, Yin LH, Tang M, Pu YP (2011) Zno, TiO(2), Sio(2), and
Al(2)O(3) Nanoparticles-induced toxic effects on human fetal
lung fibroblasts. Biomed Environ Sci 24(6):661–669. https://
doi.org/10.3967/0895-3988.2011.06.011
Zhang B, Bian W, Pal A, He Y (2015) Macrophage apoptosis induced
by aqueous C60 aggregates changing the mitochondrial membrane potential. Environ Toxicol Pharmacol 39(1):237–246. https
://doi.org/10.1016/j.etap.2014.11.013
Zhang Z, Cai J, Chen F, Li H, Zhang W, Qi W (2018) Progress in
enhancement of Co2 absorption by nanofluids: a mini review
of mechanisms and current status. Renew Energy 118:527–535
Zhou F, Liao F, Chen L, Liu Y, Wang W, Feng S (2019) The sizedependent genotoxicity and oxidative stress of silica nanoparticles on endothelial cells. Environ Sci Pollut Res Int 26(2):1911–
1920. https://doi.org/10.1007/s11356-018-3695-2
Publisher’s Note Springer Nature remains neutral with regard to
jurisdictional claims in published maps and institutional affiliations.
13