Cardiotoxicity of nano-particles Hasan Badie Bostan, Ramin Rezaee, Mahmoud Gorji Valokala, Konstantinos Tsarouhas, Kirill Golokhvast, Aristidis M. Tsatsakis, Gholamreza Karimi PII: DOI: Reference:
S0024-3205(16)30574-4 doi:10.1016/j.lfs.2016.09.017 LFS 15027
To appear in:
Life Sciences
Received date: Revised date: Accepted date:
25 June 2016 14 September 2016 23 September 2016
Please cite this article as: Bostan Hasan Badie, Rezaee Ramin, Valokala Mahmoud Gorji, Tsarouhas Konstantinos, Golokhvast Kirill, Tsatsakis Aristidis M., Karimi Gholamreza, Cardiotoxicity of nano-particles, Life Sciences (2016), doi:10.1016/j.lfs.2016.09.017
This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
ACCEPTED MANUSCRIPT
1
Cardiotoxicity of nano-particles
1
SC R
IP
Kirill Golokhvast4, Aristidis M. Tsatsakis5,*, Gholamreza Karimi1,*
T
Hasan Badie Bostan1, Ramin Rezaee2,4, Mahmoud Gorji Valokala1, Konstantinos Tsarouhas3,
Pharmaceutical Research Center, School of Pharmacy, Mashhad University of Medical
Department of Physiology and Pharmacology, School of Medicine, North Khorasan
MA
2
NU
Sciences, Mashhad, Iran
University of Medical Sciences, Bojnurd, Iran.
D
4
Cardiology Clinic, General University Hospital of Larisa, Larisa, Greece Scientific Educational Center of Nanotechnology, Far Eastern Federal University, 10
TE
3
5
CE P
Pushkinskaya Street, Vladivostok 690950, Russia Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete,
AC
Heraklion 71003, Greece. *Corresponding authors:
Gholamreza Karimi: Pharmaceutical Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran. Tel.: +985138823255-66, Fax: +985138823255, E-mail address:
[email protected]
Aristidis M. Tsatsakis: Head of Departments of Toxicology and Forensic Medicine, Medical School, University of Crete, 71003 Heraklion, Crete, P.O. Box 1393, Greece. E-mail address:
[email protected].
2
ACCEPTED MANUSCRIPT
IP
T
Abstract
SC R
Nano-particles (NPs) are used in industrial and biomedical fields such as cosmetics, food additives and biosensors. Beside their favorable properties, nanoparticles are responsible for toxic effects. Local adverse effects and/or systemic toxicity are described with nanoparticle
NU
delivery to target organs of the human body. Animal studies provide evidence for the
MA
aforementioned toxicity. Cardiac function is a specific target of nanoparticles. Thus, reviewing the current bibliography on cardiotoxicity of nanoparticles and specifically of
TE
D
titanium, zinc, silver, carbon, silica and iron oxide nano-materials is the aim of this study.
AC
CE P
Keywords: Nano-particle, Cardiac toxicity, Inflammatory marker, Reactive oxygen species.
ACCEPTED MANUSCRIPT
3
IP
T
Abbreviations:
SC R
NPs, nano-particles; ROS, reactive oxygen species; MDA, malondialdehyde; SOD; superoxide dismutase; GST, glutathione S-transferases; LDH, lactate dehydrogenase; CK, creatine
kinase;
AST,
aspartate
aminotransferase;
HBHD,
alpha-Hydroxybutyrate
NU
dehydrogenase; NMR, nuclear magnetic resonance; CK-MB, creatine kinase-MB; NO, nitric
MA
oxide; BNP, B-type natriuretic peptide; CRP, c-reactive protein; CPK-MB, creatine phosphokinase-MB; TNF- α, tumor necrosis factor alpha; IL-6, Interleukin 6; FGF2,
D
fibroblast growth factor-2; I/R, Ischemia/Reperfusion; PCR, polymerase chain reaction;
TE
ELISA, enzyme-linked immunosorbent assay; VEGFA, vascular endothelial growth factor A; GSH, Glutathione; CAT, catalase; SWCNTs, single-walled carbon nanotubes; MWCNTs,
CE P
multi-walled carbon nanotubes; BAL, bronchoalveolar lavage; IL-10, Interleukin 10; IL-12, Interleukin 12; MIP1-α, macrophage inflammatory protein 1α; Ccl11, Chemokine (C-C
AC
Motif) Ligand 11; ALT, alanine aminotransferase; ANG II, angiotensin II; ACE, angiotensin converting enzyme ; IV, intravenous; IT, intratracheal; MCP-1, monocyte chemotactic protein-1; hs-CRP, high-sensitivity c-reactive protein; IL-1β, interleukin-1 beta; p-VEGR2, phosphorylated/activated form of VEGF receptor 2; p-ERK1/2, phosphorylated/activated extracellular signal-regulated kinase (ERK)1/2, MEF2C, myocyte enhancer factor 2C; NKX2.5, homeobox protein Nkx-2.5; MRI, magnetic resonance imaging ; APX, ascorbate peroxidase.
ACCEPTED MANUSCRIPT
4
IP
Introduction ............................................................................................................................. 4
SC R
1.
T
Contents
2. Cardiotoxicity and mechanism of nano-materials ...................................................................... 7 2.1. Titanium dioxide NPs........................................................................................................... 7
NU
2.2. Zinc oxide NPs ..................................................................................................................... 8
MA
2.3. Silver NPs........................................................................................................................... 10 2.4. Carbon NPs ........................................................................................................................ 11
D
2.5. Silica NPs ........................................................................................................................... 13
TE
2.6. Iron oxides NPs .................................................................................................................. 13
CE P
3. Conclusion ................................................................................................................................ 14 Conflicts of interest ....................................................................................................................... 14
AC
References ......................................................................................Error! Bookmark not defined.
1. Introduction Nanotechnology, a branch of bionanoscience, is a rapidly growing field which is of great importance to a series of industrial applications. As characterized by the United States Nanotechnology Initiative, nanoparticles (NPs) vary from 1 to 100 nm in dimension. Due to their novel thermal and electrical properties and their small size, engineered NPs seem lately to be the centre of focus in medicinal research as well to a variety of commercial purposes. In recent years, NPs have been used in different industrial fields such as aerospace, electronics, optical devices and computer products[1].
ACCEPTED MANUSCRIPT
5
At the same time, as demand for new medicines is growing, and given the inherent nanoscale size of the biological constituents of living cell, nanotechnology is evaluated in various medical fields like oncology and cardiovascular medicine. In fact, NPs are used to
T
improve drug delivery as well as drug and biomarker research[2].
IP
Delivery of stem cells, inhibition of NOX2 via siRNA and improvement of the mechanical properties of heart valves are examples of novel applications of NPs in heart
SC R
disease [3-5].
However, as a new field with extensive use and growing interest, NPs could exert side effects that are now emerging. Titanium, zinc, silver, silica, iron and carbon-based
NU
nanomaterials are NPs whith potential harmful health effects. Human NPs exposure can unintentionally occur through a series of occupational and non-occupational scenarios.
MA
Industrial procedures such as welding, combustion and electronics production are examples of passive exposure to the said substances[1]. Day by day NPs accumulate and persist in the environment and so is the potential risk
D
of toxicity due to their exposure. Genetic damage, inflammation, inhibition of cell
TE
division and oxidative stress attribute to NPs toxicity. Especially oxidative stress is frequently reported to play a central role in NPs-induced toxicity. Physicochemical
CE P
properties of NPs including surface charge, particle size and chemical composition are thought to be the main factors involved in NPs ability to cause the increased production of reactive oxygen species (ROS) and therefore to lead to tissue oxidative damages[6]. Oxides of transition metals are used in various industrial and medicinal fields as drug
AC
delivery, textile industries and sunscreen preparations. Due to their high surface-tovolume ratio, metal NPs can provoke the generation of free radicals via Fenton reaction. Increased ROS levels and augmented apoptosis were described after exposure to Zn, Ti and Fe NPs[6]. Increased oxidative stress is responsible for the induction of the MAPK signaling pathway and increases the transcription levels of key molecules, such as Nrf2 and NF-κB. These factors in turn could provoke the augmented mRNA expression of a series of proinflammatory mediators who are involved in the pathogenesis of a variety of inflammatory diseases[7]. On the other hand, ROS are thought to play a key role in modulation of the microenvironment of cardiomyocyte. NPs exposure could lead to cardiac toxicity through increased ROS production. As studies in ischemia-reperfusion injury showed increased ROS levels and altered redox homeostasis are responsible for myocardial damage [8].
6
ACCEPTED MANUSCRIPT NPs inhalation is proven to lead to cardiorespiratory diseases in mammalian models. With a freshwater fish as a model, specifically the white sucker (Catostomus commersonii), it was found that when it was exposed to ZnO NPs, parasympathetic input
T
increased and heart rate decreased[9].
IP
At the same time, cytotoxic and genotoxic effects were observed when SICH cell line was exposed to Ag NPs. Altered oxidative status and decreased oxidative defenses were
SC R
observed as GSH, SOD and CAT levels decreased.
NPs use has a lot to offer especially in the field of cardiovascular medicine. However, the aim of this study is to delineate the adverse effects of NPs use and their related
AC
CE P
TE
D
MA
different NPs are presented in figure 1.
NU
cardiotoxicity. Adverse cardiovascular effects and mechanisms of cardiotoxicity of
Fig. 1: Adverse cardiovascular effects and mechanisms of cardiotoxicity of six highly used nanoparticles. Increase; Decrease; Reg: regulation; Inh: inhibition.
ACCEPTED MANUSCRIPT
7
T
2. Cardiotoxicity of nano-materials and the underlying mechanisms
IP
2.1. Titanium dioxide NPs
SC R
In comparison with other metal-based NPs, titanium derived NPs are used in large quantities. TiO2 NPs, the most common form of titanium NPs, are water-insoluble and odorless materials. Biocompatibility, optical, electrical and whitening properties of TiO2 are
NU
some of its interesting features which make it a valuable substance for a number of applications. It can be found in abundance in products like white pigments, sunscreens, and
MA
food colorants [1, 10].
Despite its widespread use, nano-sized TiO2 toxicity has not yet been completely understood. Recent toxicological studies have indicated harmful effects of TiO2 NPs in
D
different biological systems and organs[11]. For instance, accumulation of TiO2 NPs in the
TE
brain induces the release and metabolism of neurotransmitters such as norepinephrine and 5hydroxytryptamine[1]. Thus, understanding the risks and hazards including cardiotoxicity
CE P
associated with TiO2 NPs exposure and evaluation of its dose-dependent effects are of great importance. In the zebrafish, as a model of chronic toxicity, it was shown that Titanium NPs can translocate between organs, are capable of passing through the blood–heart barrier and accumulate in the heart [12].
AC
The exact mechanisms via these particles act in mammalian species and cause their toxic effects are not understood yet. Despite the effective function of blood–heart barrier, particular NPs can accumulate in heart tissues and exert their toxic effects [12]. Chronic exposure to TiO2 NPs resulted in sparse cardiac muscle fibers, inflammatory responses in tissue level, cell necrosis and cardiac biochemical derangement. Increased ROS levels (including superoxide radicals and hydrogen peroxide), reduced malondialdehyde (MDA) content and augmented DNA peroxidation in the cardiac muscle was observed following exposure to TiO2. Additionally, TiO2 attenuated the antioxidant enzymes activity, especially of superoxide dismutase (SOD) and glutathione S-transferases (GST) [10]. Spectroscopic assays demonstrated that TiO2 NPs directly bind to lactate dehydrogenase (LDH) and result in alteration of the secondary structure of LDH. Thus, it is
ACCEPTED MANUSCRIPT
8
believed that the nano-anatase TiO2 is responsible for a new metal ion-active site in LDH and subsequently for a LDH activity enhancement [13]. Bu and colleagues showed that administration of nano-anatase TiO2 into the
T
abdominal cavity could increase the activity of creatine kinase (CK), LDH, AST, and HBDH.
IP
Elevated levels of these enzymes are indicators of myocardial injury. Also, NMR-based metabonomic approach showed that AST, CK and LDH activities were elevated in the heart
SC R
tissues of TiO2 NPs-treated rats. Mitochondrial swelling was another feature of toxicity observed in those animals [14]. Increased troponin T, myoglobin, CK-MB and nitric oxide were detected. Also, caspase-3 was elevated in the cardiac tissue of TiO2-intoxicated rats.
NU
TiO2 NPs induced apoptosis is thought to occur via mitochondria mediated pathways. This effect may be due to increased mitochondrial permeability transition followed by the release
MA
of apoptogenic factors such as cytochorome c and activation of caspase-9 and caspase-3. With the use of the Comet assay it was demonstrated also that the said NPs induced DNA damage, a fact that was evident by an increase in DNA tail length[15].
D
However, in order to understand better the cardiovascular impact of TiO2 NPs, further studies
TE
on specific effects of TiO2 NPs on action potential of cardiac cells, ion channels function, coronary blood flow, electrocardiogram changes and biomarker levels referring to myocardial
CE P
derangement such as BNP and CRP. A summary of reports on titanium dioxide NPs are
AC
presented in Table 1.
2.2. Zinc oxide NPs
Zinc (Zn) favorable characteristics are vastly used in the production of alloys, paints and brass[16]. Recently, quantum effect, large surface area and reactive surface sites of nanoscale metal powders have made Zn an exciting choice[17]. The special properties of zinc oxide made it the fifth most applicable engineering-based nanomaterial in consumer products as it is used in large quantities in sunscreens, food additives and pigments [18].Zn NPs are used in semiconductors, paints and coatings. Exposure to these particles occurs through oral (main route), inhalation, intravenous and dermal contact. Wide use of these elements leads to human exposure intentionally or unintentionally[19].
ACCEPTED MANUSCRIPT
9
After oral administration of ZnO NPs, tissue absorption and accumulation occurs through interactions between zinc and sulfur-containing sites of the proteins. Kinetic profile studies on ZnO NPs showed that the majority of them are excreted via feces and a small
T
amount is excreted via urine[20]. In vivo and in vitro studies showed that the adverse effects
IP
of ZnO NPs also involve their dissolved fractions such as zinc ions. In vitro exposure to ZnO NPs can also reduce mitochondrial membrane potential, increase ROS production, and
SC R
activate apoptotic signaling pathways [21]. In vivo studies showed that the liver, spleen, heart, pancreas and bone are target organs following oral exposure to NPs of 20 to 120-nm diameter [22]. Rats exposed to occupational levels of air-borne ZnO NPs developed
NU
inflammation in lung tissue and exhibited myocardial damage following long-term exposure [23]. Additionally, ZnO NPs cause cardiorespiratory toxicity in fish model as the Catostomus
MA
commersnii. Heart rate declined in ZnO NPs-treated fish, possibly as a result of damage to gill neuroepithelial cells and a consequent increase in parasympathetic input to the cardiac pacemaker[9]. DNA damage, inflammation and apoptosis in rat’s heart have been observed
D
when ZnO NPs were administrated orally. Elevations of troponin T, CPK-MB and myoglobin
TE
levels which are indicative of cardiac damage were also reported [24]. Remarkable increase in serum pro-inflammatory biomarkers including TNF-α, IL-6
CE P
and CRP in rats intoxicated with ZnO NPs, were reported. The induction of such biomarkers may play a principle role in cardiac toxicity induced by ZnO NPs. It has been demonstrated that, in cultured cardiac myocytes, TNF-α increased the production of reactive oxygen species (ROS) resulting in DNA damage [25]. Also, up-regulation of TNF-α can lead to
AC
increase in other cytokines such as IL-6 which is known as the chief stimulator of CRP production[26].
These particles may cause cardiac toxicity via disturbing calcium homeostasis. ZnO NPs can affect calcium permeability and may result in accumulation of Ca2+ in cytosol. This phenomenon is associated with the pathogenesis of myocardial injury[27]. Baky and colleagues showed that ZnO NPs induced cardiac DNA fragmentation evident by a significant increase in the DNA tail length and of % DNA bases in the tail in cardiac samples of rats intoxicated with ZnO NPs. DNA damaging potential of ZnO NPs was confirmed by several experimental studies[28, 29]. Damages to DNA backbone and bases were reported following ROS generation[30]. One of the most important events after DNA damage is apoptosis [25]. Elevated levels of caspase-3 were observed in rats exposed to ZnO NPs, proposing that apoptosis is related to DNA damages [24]. Unfortunately, there were just a couple of studies that showed
ACCEPTED MANUSCRIPT
10
adverse effects of ZnO NPs on cardiac tissue. Thus, complementary research is necessary to provide a better understanding of the toxic aspects of these substances. Table 2 depicts studies on zinc oxide NPs.
IP
T
2.3. Silver NPs
SC R
Silver (Ag), is a soft, white bright metal. Silver coins and jewelry are used in large quantities. Industrially, silver is used in mirrors, photography and conductor tools[1]. Silver NPs have been developed as potent antimicrobial agents and have a variety of applications in toothpastes, bedding, water purification and nursing bottles [31, 32]. After oral exposure, it is
NU
shown that about 18% of silver could be absorbed in humans. Animal studies showed that it is distributed to all body organs and the majority of it is excreted in bile [33].
MA
Recently, numerous studies have reported silver nano-materials adverse effects including cytotoxicity and ROS generation [34, 35]. Noticeable decrease in function and membrane integrity of mitochondria, increased levels of inflammatory cytokines and several
D
other toxic effects were observed in alveolar macrophages [31]. In PC12 cell line, silver NPs
TE
induced irregular membrane borders and cell shrinkage [36]. Silver NPs exposure enhanced superoxide anion production and caused deleterious effect in heart tissue and led to oxidative
CE P
stress and inflammation in rats [37]. Heart rate retardation in medaka embryos (Oryzias latipes) was reported by Cho et al.[37]. After sub-chronic (dermal) application of Ag NPs in guinea pigs, it was found that the said nanoparticles accumulate in different organs such as
AC
the heart and cardiomyocytes deformity is observed[38]. Fibroblast growth factor-2 (FGF2) in the heart contributes to pathological states such as hypertrophy, atherosclerosis and ischemia/reperfusion (I/R) injury [39, 40]. By means of PCR and ELISA methods, it was shown that FGF2 mRNA expression level was downregulated in chickens that received 20 ppm of Ag NPs. On the other hand, vascular endothelial growth factor A (VEGFA) which is an angiogenesis modulator in the heart was up-regulated at the same dose [40-42]. Measurement of GSH content, SOD and catalase confirmed the noxious effect of silver NPs due to oxidative stress in A431 and HT-1080 cell lines. Another study showed that Ag NPs significantly diminished GSH content, decreased SOD and catalase activities and increased lipid peroxidation in SICH cell line in a concentration-dependent manner. Also, condensation and fragmentation of nuclei was shown by comet assay [31]. A summary of studies on silver NPs is given in Table 3.
ACCEPTED MANUSCRIPT
11
2.4. Carbon NPs
T
Carbon (C) can be found in a wide range of substances. Depending on binding sites,
IP
this atom can form different allotropes such as graphite and diamond. As an allotrope of carbon, single-walled carbon nanotubes (SWCNTs) are composed of a sheet of graphene with
SC R
a diameter of 1–4 nm and a length ranging from 100 nm to 1.5 mm. Because of versatile chemistry, SWCNTs exhibit a wide range of application in cancer therapy, drug delivery, gene therapy, sensors, conductive plastics, paints and technical textiles [43, 44].
NU
Small particle size, large surface area and ornamentation with catalytic metals are the factors which make SWCNTs toxic agents. These conditions result in ROS generation and
deleterious effect on cells [44-46].
MA
oxidative damages by Fenton reaction, a recognized pathway via which SWCNTs yield their
Graphite sheets with a nano-scale diameter can produce another form of carbon nanotubes
D
called multi-walled carbon nanotubes (MWCNTs) [47]. Because of special physical
TE
compositions and inherent designs, these particles have been used in biomedical, electronic, computer, and aerospace products[48].
CE P
Inhalation is the main route of exposure to carbon NPs. Thus, lung is the first organ exposed to toxic effect of these particles. Consequently, these NPs are transported to secondary organs [49, 50]. After the pulmonary contact with carbon NPs, liver and kidney necrosis, inflammation, microvascular abnormality, depletion of serum antioxidant and other
AC
toxic effect may occur [43]. Mercer and colleagues showed that MWCNTs settled in the lungs were also distributed to the heart, brain, liver and kidney [51]. After exposure to SWCNTs, levels of carbon-centered lipid-derived radicals were elevated in the lung, heart and liver. Pulmonary exposure to SWCNTs leads to oxidative damage and recruitment of inflammatory factors in the bronchoalveolar lavage (BAL) fluids. Systematic toxicity may occur after a week of exposure which is characterized by a decrease in thiol contents, an elevation in lipid-peroxidation products and an increase in inflammatory indices in the heart and liver [43]. I/R play an important role in the pathophysiology of myocardial damage [52]. Also, a direct correlation between inflammatory cytokines (i.e. IL-6, IL-10, and IL-12) and I/R injury has been reported. MIP1-α is another factor which is implicated in I/R injury in myocardial tissue [52].
ACCEPTED MANUSCRIPT
12
The effects of MWCNTs on myocardial ischemia-reperfusion (I/R) injury model were investigated by Urankar et al. [52]. Post I/R myocardial infarction worsened in a dose and time-dependent manner following intratracheal (IT) instillation of multi-walled carbon
T
nanotubes. MWCNTs caused elevated levels of inflammatory mediators. Eotaxin levels were
IP
increased after exposure to MWCNTs and a correlation between serum eotaxin (eosinophil chemotactic proteins) levels and cardiac injury, heart failure and regulation of
SC R
inflammatory cell was observed. Eotaxin is mainly secreted by smooth muscle cells and fibroblasts and its production in generally related to involvement of eosinophils and basophils in certain inflammatory conditions [53-55].
NU
Pulmonary exposure to carbon NPs can lead to liver and myocardial damage and result in mild alteration in ALT, AST, LDH and CK levels in serum. SWCNT phagocytosis
MA
in lung epithelium may induce production of pro-inflammatory factors, which play a key role in local and systemic inflammatory responses. When SWCNTs were instilled into the lung of hypertensive rats, alterations in cardiovascular and pulmonary system occurred 24 and 72 h
D
post instillation. Significant increases of ET-1 and ACE levels in plasma were observed in
TE
treated animals [44]. In another animal study, it was found that MWCNT inhalation could lead to impairments of endothelium-dependent dilation in the coronary microcirculation
CE P
within 24 h, a condition from which full recovery was not achieved within 168 h [56]. Animal studies on embyogenesis showed that chicken embryos heart vascularization was significantly reduced following exposure to diamond and graphite NPs. At the same time, mRNA and protein expression of the proangiogenic basic fibroblast growth factor (b
AC
FGF) was decreased in heart tissue[57]. Similarly using the chick chorioallantoic membrane (CAM) model, carbon-based nanomaterials as carbon nanotubes and fullerene as well as graphite proved to be anti-angiogenic in the presence of 2 pro-angiogenic factors, the vascular endothelial growth factor (VEGF) and the basic fibroblast growth factor (FGF2), with a relatively greater potency in inhibiting VEGF versus FGF2 by the said carbon materials [58]. Intravenous (IV) and intra-tracheal (IT) administration of C60 caused myocardial infarction in rats. Coronary artery contraction was indicated, after IT exposure to C60. Thompson et al. demonstrated that IV and IT administration of C60 fullerene resulted in expansion of myocardial infarction in Sprague-Dawley rats following I/R injury. In animals that received C60 IV, elevated concentration of IL-6 and MCP-1 were observed [59]. It was observed that IT instillation of MWCNT caused arrhythmogenic effect, increased ET-1 release and depressed coronary flow in Sprague-Dawley rat. These events
ACCEPTED MANUSCRIPT
13
may contribute to post-I/R myocardial infarct expansion [60]. Some of studies on carbon NPs are mentioned in Table 4.
IP
T
2.5. Silica NPs
SC R
Silica is an oxide form of silicon [61]. Silica NPs are ranked among top five of mostly used nano-materials in commercial products and are used in drug delivery, gene therapy, diagnosis, papermaking and cosmetics [62]. In addition, due to its higher biocompatibility than other imaging NPs, silica NPs were introduced as ideal tools for medical
NU
photography[1].
Because of low density, these substances can readily mix with air. Inhalation is the main
MA
route of exposure in the work places during production and storage [63, 64]. Lung is the first organ which is adversely affected and this may lead to systematic toxicity (e.g. cardiovascular inflammation) [65].
D
Recently, silica NPs were used for delivery of adenosine, a prototype cardioprotective agent,
TE
into I/R heart tissue [66]. Silica NPs are capable of producing ROS which may lead to cytokine release and apoptosis [67].
CE P
In rats IT administration of silica NPs cause hs-CRP level elevation in a sizedependent manner and TNF-a, IL-6, and IL-1β levels increase. These particles cause inflammation in cardiovascular system. ET-1, D-dimer, LDH and CK-MB increased after
AC
silica NPs exposure [64].
MEF2C is a key factor in cardiac morphogenesis and myogenesis before the formation heart tube [68, 69]. NKX2.5 is another agent which regulates heart development in humans. Genetic insufficiency in NKX2.5 contributes to atrioventricular defects [70]. Histamine can be used as a marker of myocardial ischemia and its release usually results in coronary vasoconstriction. Elevated levels of histamine were observed by Chen et al. Increased levels of cTnT which is indicative of acute myocardial injury were observed in old rats which were exposed to aerosol of silica NPs [63]. Table 5 gives a summary of reports on silica NPs.
2.6. Iron oxides NPs Superparamagnetic and physicochemical properties of iron oxide NPs are important features for medical and industrial applications. Iron oxide NPs act as a contrast agent in
ACCEPTED MANUSCRIPT
14
positron electron tomography, magnetic resonance imaging and ultrasound. In addition, they are used in drug and gene delivery, cancer therapy and catalysis[1]. After subcutaneous injection, superparamagnetic iron oxide NPs were distributed in heart,
T
lung, kidney, liver and spleen [71]. Due to generation of ROS, iron oxides NPs result in
IP
oxidative damage. Subsequently, cytoskeletal disruption, decrease in proliferation and cell death occurred [72]. On the contrary, using confocal microscopy, it was also reported that
SC R
negatively charged superparamagnetic iron oxide NPs did not affect the actin cytoskeleton of heart cells significantly while they markedly disrupted the actin cytoskeleton in kidney and brain cells (similar results were reported from in vivo experiments). Increased vascular
NU
permeability was reported after exposure to these particles [73]. In another study slight change of cell viability and genetic content was observed [74, 75]. Acute heart rate reduction
MA
was observed when BALB/c mice received 10 mg/kg polyacrylic acid coated γ-Fe2O3 NPs [75]. Effect of Fe2O3 and Fe3O4 NPs (0.001– 100 μg/ml) on endothelial cells of heart microvascular were assessed by Sun et al. Result of their study revealed that these particles
D
did not change plasma membrane permeability and inflammatory factors, significantly [73].
3. Conclusion
CE P
TE
A summary of investigations performed on iron oxide NPs is given in Table 6.
Because of the unique properties of nano-materials, these substances are widely used
AC
in biomedical and industrials fields. Understanding all aspects of nano-material safety is of great importance. In recent years various studies showed specific organ toxicity of these substances especially of the heart, brain and lung. Although discrepancies are observed among published studies, nano-scale materials seem to affect heart tissue and vigilance is needed in their broad and augmented application. Further research and heart functional studies especially with the use of echocardiography are needed for the better understanding of the possible heart toxicity of nanoparticles.
Conflicts of interest We declare no conflict of interest.
ACCEPTED MANUSCRIPT
15
Table 1. Cardiotoxic properties of TiO2 NPs. Experimental
Treatment
model
protocol
Main results
Proposed
Referen
mechanisms
ce [76]
Male
and
A single dose
No apparent damage was seen in the
Probably due to late
Female
ICR
of TiO2 NPs (0,
heart
onset
and
1387
mg/kg
BW)
was
T
damages.
IP
140, 300, 645,
injected
and evaluation
14 days later. Animals
were
Increased
rats
treated
with
myoglobin, TNF-ɑ, Il-6, NO, IgG,
TiO2 (600 and
VEGF, serum glucose and calcium, C-
directly
1000 mg/ kg
reactive
and
phosphate
/day, orally) for
increased
and
and/or
5
augmented DNA damage.
protein,
activity
Nanoanatase
TiO2
NPs
lead
(via
DNA
indirectly stress)
damage DNA. Higher
TiO2 (5, 10, 50,
Even at high doses, heart weight /body
toxicity
100,
and
weight values were not significantly
inflammation.
150mg/kg/day)
different from controls. Titanium heart
were
injected
tissue levels were lower than those of
the
other evaluated organs (namely, liver,
AC
[77]
moiety)
An LD50 of 150 mg/kg was found.
in
to
genotoxicity as they
(oxidative
D
caspase-3
CK-MB
T,
CE P
mice
troponin
TE
days ICR
of
MA
Wistar albino
Female
levels
NU
was performed
consecutive
myocardial
SC R
mice
of
abdominal
kidney, spleen, lung and brain).
cavity for 14
Although AST, CK, LDH, and alpha-
days.
HBDH activities at 5 and 10 mg/kg
doses
cause
[78]
and
was not different from the control, at 50,
100,
and
parameters
150 were
mg/kg
these
significantly
increased implying that myocardium damage occurs at higher doses.
ICR mice
female
Intragastric
Heart parameters and titanium heart
TiO2
administration
tissue levels were significantly and
oxidative
of
nano-TiO2
dose-dependently increased. Evidence
excessive
(2.5, 5, and 10
of myocardial pathology increased in a
production and lipid
mg/kg/day) for
dose-dependent manner (e.g. severe
peroxidation and
90 days.
inflammatory
weakening
cell
infiltration
on
NPs
induce
stress
by ROS
the
by anti-
[79]
ACCEPTED MANUSCRIPT
16
tunica externa at 5mg/kg and cell
oxidant defense.
necrosis at 10 mg/kg). The levels of O2–,
H2O2,
(MDA),
and
lipid
peroxidation
protein peroxidation
T
(protein carbonyl), as well as DNA
IP
damage were increased. The activities of SOD, catalase (CAT), ascorbic acid (APx),
glutathione-S-
SC R
peroxidase
transferase (GST), and glutathione
reductase (GR) were decreased with
increasing doses of TiO2. Increased
NU
levels of creatine kinase reflected myocardial damage. Fish
were
Titanium heart tissue levels were
Despite the presence of
zebrafish
exposed to 1.0,
comparable with brain levels and both
blood–brain barrier and
(Danio rerio)
2.0, 4.0, 5.0,
were dose and time-dependant.
blood–heart
MA
wild-type
7.0 mg/l TiO2 and
TE
months
D
NPs for up to 6
randomly
[80]
barrier,
nanomaterials are able to pass (due to their very small size) and induce toxic effects.
sampled every
Rat
Lactate
In
CE P
2 months.
vitro
While at lower TiO2 concentrations
TiO2
to
LDH activity was increased, it was
secondary
decreased at higher concentrations.
and
exposure
e
TiO2 (5 nm)
Rats
AC
dehydrogenas
A single
Decreased action potential period,
intratracheal
impairment of sarcomere
dose of TiO2
shortening and diminished stability
NPs (2
of resting membrane potential
mg/kg).
were observed, in vitro. Increment
Also, isolated
of cardiac conduction velocity and
cardiomyocyt
tissue excitability (increased
es were also
chance of arrhythmias).
exposed to TiO2 NPs, in vitro
changes
LDH
[81]
structure
increases
its
activity.
ROS overproduction
[82]
ACCEPTED MANUSCRIPT
17
Table 2. Cardiotoxic properties of ZnO NPs. Treatment
model
protocol
Main results
and
Nanoscale zinc
Only fatty degeneration was seen in
female
CD-
5
Zn/kg
the cardiovascular cells. An apparent
(samples taken
rise was seen in lactate dehydrogenase
14 days after
(LDH), aspartate aminotransferase
the
(AST), creatine kinase (CK) and
gastrointestinal
hydroxybutyrate
administration)
(HBD) levels (conventional cardiac
mechanisms
ce [83]
SC R
ICR mice
g
Referen
IP
Male
Proposed
T
Experimental
dehydrogenase
NU
enzymes). Male Sprague-
Intratracheal
After 3, 6 and 12 hr, Zn heart tissue
Systemic inflammation
Dawley rats
administration
levels were not significantly different
and
of 10 mg/ml
from controls but 24 hr post-exposure
disturbance.
ZnO NPs
they reduced significantly.
MA
oxidative
Exposure
to
Rat’s heart samples showed focal
Dawley rats
ZnO NPs for 2
fibrosis, myocardial degeneration and
weeks (5 h/day
necrosis 7 days post exposure.
TE
D
Male Sprague-
and
5
CE P
days/week) white suckers
[84]
Fish
exposed
were
Fish treated with ZnO showed a
Gill
to
significant 35% reduction in heart rate
cells injury possibly
following 15 and 20 h treatment
resulted
ZnO NPs (1.0
in
parasympathetic input to
the
cardiac
pacemaker
Wistar albino
Oral
Serum
rats
administration
(TNF-α, Il-6 and CRP), troponin-T,
of
and
CK-MB, myoglobin, serum VEGF
1000 mg/kg for
(angiogenic factor), NO (oxidative
5 days
stress marker), and cardiac calcium
600
[85]
enhancement of
AC
mg/L) for 25 hr
neuroepithelial
pro-inflammatory
markers
level, caspase-3 activity and DNA damage were increased at both doses of ZnO NPs as compared to control animals.
[86]
ACCEPTED MANUSCRIPT
18
Table 3. Cardiotoxic properties of Silver NPs. Experimental
Treatment
model
protocol
mechanisms
ce [87]
Oral
A non-significant increase in CPK-
Heart injury was not
administration
MB as compared to control animals.
accompanied
of silver NPs (3
Histopathological
increase in CPK-MB
mg/kg/day, 10–
showed mild edema and separation of
25 nm particle
myofibrils.
size)
for
examination
21
days
Superoxide anion production was
Inflammatory
Dawley rats
administration
increased by 41% in heart tissue.
responses
NU
Oral
500
mg
and
India
24-hr exposure
At concentrations >2 μg/mL, NPs
Ag
Catla
catla
to 2, 4, 8, 16,
were toxic for SICH. A concentration-
oxidative
heart cell line
32
64
dependent increase of tail DNA (%)
cytotoxicity
(SICH)
μg/mL of Ag
was detected. While not significant at
genotoxicity.
NPs
2 and 4 μg/mL, a significant decrease
TE
D
Sahul
and
[88]
oxidative stress.
MA
silver NPs/kg
an
levels.
Male Sprague
of
by
T
rats
Referen
IP
Wistar
Proposed
SC R
Male
Main results
NPs
induce
[89]
stress, and
in CAT, SOD and GSH was observed at 16, 32 and 64 μg/mL.
Intratracheal
Although IT administration of Ag NPs
Probably
Dawley rats
(IT) instillation
did
of
sensitize the immune
of
citrate
proinflammatory markers (IL -1b, IL-
system resulting in a
AgNP
2, IL-5, IL-6, IL-10, IL-13, IL-17a,
more marked response
(200 µg). After
IL-18, MCP-1, IFN-γ, RANTES, and
in
24 and 168 hr,
TNF-α), heart rate and QT interval,
myocardial
myocardial
the levels of IL-2, IL-6, and IL-18
reperfusion injury.
CE P
male Sprague
AC
capped
ischemia
(by
not
significantly
affect
the
increased
level
after
the
Ag
setting
NPs
[90]
of
ischemia
I/R.
ligation of left
Treatment with Ag NPs worsened the
anterior
outcomes of I/R without inducing
descending
systemic inflammatory reactions or
coronary artery
electrical disturbances.
for 20 min) and subsequently reperfusion (for 2hr)
were
induced. Male Sprague-
Five-hour
Dawley rats
exposure
by
cardiovascular parameters remained
Short-term exposure to
unchanged on day 1 and 7 d post
Ag NPs does not cause
[91]
ACCEPTED MANUSCRIPT
19
inhalation
to
exposure.
acute
100 (equal to
cardiovascular
toxicity.
20 mg/L total silver)
μg/m3
T
1000
and
total
silver)
SC R
mg/L
IP
(equal to 200
Broiler
Birds
were
At 20 ppm, Ag NPs augmented the
chickens
treated
with
expression of VEGFA gene, and
water
reduced FGF2 gene expression at
solution containing
mRNA and protein level.
NU
oral
10
or 20 ppm of
MA
Ag NPs Oryzias
Embryos were
Following a 7-day exposure to Ag
latipes
exposed
NPs (0.8 mg/l), a significant decrease
(medaka)
0.1−1 mg/l of
embryos
Ag NPs for 14 days Brain, heart
male Wistar
and skeletal
rats
muscle
D Creatine kinase was inhibited in brain
Silver NPs potentially
and
interact with
CE P
Adult and
[93]
in heart beat was recorded.
TE
to
[92]
skeletal
muscle;
however
it
remained unaffected in the heart.
homogenates
[94]
sulphydryl moieties of CK.
were treated
AC
with silver NPs (10, 25 and 50 mg/l) for 1 hr
Albino zebrafish
Si NPs (1 to 12
Pericardial edema and bradycardia while no
SiNPs inhibited calcium
mg/mL) via
effect on atrioventricular conduction. Cardiac
signaling pathway and
intravenous
output dose-dependently decreased.
decreased cardiac
microinjection for
Oxidative stress and neutrophil-mediated
contraction via down-
24 hr.
cardiac inflammation were observed.
regulation of related genes, such as ATPase-related genes (atp2a1l, atp1b2b, atp1a3b), calcium channelrelated genes (cacna1ab,cacna1da) and the regulatory gene tnnc1a for cardiac troponin C.
[95]
ACCEPTED MANUSCRIPT
20
Table 4. Cardiotoxic properties of carbon NPs. Experimental
Treatment
model
protocol
Male
Rats
spontaneously
Main results
Referen
mechanisms
ce
Probably
intratracheally
in the left ventricle and myofiber
inflammatory
hypertensive
exposed to Fe-
degeneration. Fe-rich NPs post 72-h of
responses
rats
poor SWCNTs
exposure caused more marked heart
oxidative stress.
and
tissue derangement.
SWCNTs. Adult female
A
single
C57BL/6mice
pharyngeal
to
7 days post-exposure, only at 80 μg
Increased
treated animals, LDH levels and
inflammation
and and
[97]
of
myeloperoxidase activity in heart
oxidative
stress
SWCNTs
(40
tissue homogenates increased by 22%
depleted
antioxidant
80
and 15-34%, respectively. SWCNTs
defense.
exposure at 40 and 80 μg/mouse caused
MA
μg/mouse)
NU
aspiration
and
[96]
and
IP
Fe-rich
due
T
SWCNTs caused histological damages
SC R
were
Proposed
heart
tissue
thiols
levels
reduction by 7 and 10%, respectively
D
and elevated carbonyls levels by 24
TE
and 44%, respectively. Male
A single dose
Administration of MWCNT dose and
C57BL/6J
of
time-dependently
mice
(0.01, 0.1, 1, 10
myocardial infarction and the severity
and 100 μg) in
changes with different forms of the
3
particles.
CE P
MWCNT
forms
AC
(unmodified, commercial grade,
and
functionalized)
worsened
[98]
ACCEPTED MANUSCRIPT
21
Table 5. Cardiotoxic properties of silica NPs. Proposed
Referen
protocol
mechanisms
ce
male Sprague-
Animals
myocardial
Ischemia may be due
[99]
Dawley
inhaled
ischemia was reported (more marked
to an increase in whole
nanoparticles
with
blood viscosity which
weeks (65 g),
for 40 min per
Troponin-T (as a marker of acute
8 weeks (265
day
myocardial
g),
weeks.
rats
of
3
and
months
20
An SiO2
for
4
age-dependent
increasing
significantly
(670
age).
Cardiac
ischemia)
T
model
Main results
IP
Treatment
may in turn increase
was
peripheral
vascular
Cardiac
resistance,
decrease
SC R
Experimental
increased.
rhythm was altered only in older rats.
MA
NU
g) old.
perfusion
of
the
coronary
artery
and
facilitate
thrombosis
resulting in increased risk
of
myocardial infarction.
Myocytes were
After 10 min of incubation, MCM41-
MCM41-cal
myocytes
exposed to 200
cal and SBA15-cal either attached to
SBA15-cal
isolated from
μg/mL
the plasma membrane or entered
reasonable
male
MCM41-cal or
ventricular myocytes.
bioavailability
Male
SBA15-cal
Wistar
(two types of
min following intravenous injection
calcined
(2.0 mg/mL), MCM41-cal passed in
rats for in vivo
Mesoporous
the
experiments
silica
ventricular myocytes and localized
microparticles)
around the mitochondrial membranes.
AC
[100]
show
In vivo results showed that within 10-
CE P
rats
and
TE
Wistar
D
Ventricular
of
acute
perivascular
space,
entered
for 10 min
Human
Cells
were
According to TEM images, NPs
Cell death was induced
umbilical vein
treated
with
entered the cells (mostly found in
by oxidative stress and
endothelial
silica
perinuclear region). Cell viability
apoptosis.
cells
nanoparticles
decreased and LDH activity (as a
(HUVECs)
(25, 50, 75 and
marker
100 μg/mL) for
increased with increasing dose and
24 hr.
time. NPs induced early and late
of
membrane
damage)
apoptosis and the latter was much more marked. NPs increased ROS production and reduced SOD activity in a dose-dependent manner.
[101]
ACCEPTED MANUSCRIPT
22 of
silica
In vivo study, showed that treatment
the AB strain
nanoparticles
with
(wild-type) for
(25,50, 100 and
malformations such as pericardial
in
200 μg/mL) for
edema.
vivo
experiments
4-96
hr
NPs
caused
embryonic
post
T
Zebrafish
For all diameters, NPs heart tissue
with diameters
concentration
of 30, 60, 90,
increasing
and
concentrations were seen for 30 nm
600
nm
Higher
and lower concentrations for 600 nm
administered
NPs. While GSH content remained
(intratracheally
unchanged, SOD decreased and ROS
16 times every
and MDA levels increased with the
other day) at
increase of dose and diameter. LDH
three doses of
and
2, 5 and 10
significantly in all treated groups.
CK-MB
particles
(50–
200 nm with
dominant
diameter) was injected.
[102]
oxidative stress, and ROS generation
D
increased
Particles
groups were not different from the
accumulate
control group.
tissue.
CE P
150 nm as the
levels
NPs heart tissue levels of experimental
TE
0.5 mg of SiO2
AC
rats
Wistar
doses.
with
were
mg/Kg male
increased
Inflammatory reaction,
SC R
rats
Silica particles
NU
Wistar
MA
Male
IP
fertilization
do
not
in
heart
[103]
ACCEPTED MANUSCRIPT
23
Table 6. Cardiotoxic properties of iron oxide NPs. Proposed
Referen
model
protocol
mechanisms
ce
human cardiac
Fe2O3
Following 12 and 24 h treatments, no
Probably due to their
[104]
microvascular
Fe3O4 NPs
marked decrease in cell viability was
large specific surface
endothelial
0.001-100
seen. A non-significant increase in
area
cells
μg/ml)
LDH leakage (as a marker of cell
and (
(HCMECs)
membrane
integrity)
and
ROS
T
Main results
IP
Treatment
SC R
Experimental
production was recorded. Also, this
treatment did not affect endothelial permeability
nor
the
mRNA
NU
expression of inflammatory markers
(Vascular cell adhesion molecule-1,
MA
intercellular adhesion molecule 1, macrophage cationic peptide-1 and IL-8).
mice
Either a single
In macrophages of multiple organs, no
The
dose
of
iron-positive pigment was reported
accumulation
100mg/kg or a
from histopathological studies and the
marked.
10-day repeated
of
level
of is
[105]
not
level of NPs between single and multiple dose treatment was not significantly different.
CE P
administration
D
KM
TE
Male
Superparamagn
AC
etic iron oxide (SPIO)
(subcutaneousl y)
Sprague-
Fe2O3@DMSA
Infarction size in NPs-treated groups
NPs protected
Dawley rats
(0.1, 0.25, 0.5
was significantly lower compared to
myocardium from
mg Fe /kg/day)
saline-treated animals. SOD activity in
ischemia and this
for 7 days.
NPs-treated animals (0.5 and 0.25
effect depends on core
mg/kg) was significantly higher but
sizes but not on
MDA,
molecules used to coat
LDH,
activities and
CK,
and
CK-MB
MDA content were
[106]
NPs.
significantly lower. Mice
Intravenous
NPs enhanced plasma plasminogen
Oxidative stress
administration
activator inhibitor-1 (PAI-1) levels.
evident by augmented
of ultrasmall
Moreover, they increased creatine
lipid peroxidation
superparamagn
phosphokinase-MB isoenzyme (CK-
markers, reactive
[107]
ACCEPTED MANUSCRIPT
24
etic iron oxide
MB), lactate dehydrogenase (LDH)
oxygen species and
nanoparticles
and troponin-I levels in plasma.
superoxide dismutase
(0.4, 2 and 10
activity in the heart.
μg/kg) Mitochondrial respiratory chain
Fe3O4 exposure had no
mitochondria
complexes activities remained
toxic effects on
from brain,
unchanged in all tissues and at all
heart, liver,
concentrations of iron oxide
lung and
nanoparticles.
IP
T
Isolated
mitochondrial coupling
SC R
and respiratory chain complexes I, II, III,
kidneys were
and IV activities,
exposed to
probably due to
NU
Fe3O4 (0, 100, 200, 300 and 500 µg/ml) for
CE P
TE
D
MA
30 min at 25oC.
AC
Wistar rats
insufficient duration of exposure.
[108]
ACCEPTED MANUSCRIPT
25 References 1.
Karmakar, A., Q. Zhang, and Y. Zhang, Neurotoxicity of nanoscale materials. J Food Drug Anal, 2014. 22(1): p. 147-60.
2.
Medina, C., et al., Nanoparticles: pharmacological and toxicological significance. Br J Pharmacol,
Perin, E.C. and J. Lopez, Methods of stem cell delivery in cardiac diseases. Nat Clin Pract Cardiovasc
IP
3.
T
2007. 150(5): p. 552-8.
4.
SC R
Med, 2006. 3 Suppl 1: p. S110-3.
Somasuntharam, I., et al., Delivery of Nox2-NADPH oxidase siRNA with polyketal nanoparticles for improving cardiac function following myocardial infarction. Biomaterials, 2013. 34(31): p. 7790-8.
5.
Ghanbari, H., A. de Mel, and A.M. Seifalian, Cardiovascular application of polyhedral oligomeric
NU
silsesquioxane nanomaterials: a glimpse into prospective horizons. Int J Nanomedicine, 2011. 6: p. 775-86. 6.
Manke, A., L. Wang, and Y. Rojanasakul, Mechanisms of nanoparticle-induced oxidative stress and
7.
MA
toxicity. Biomed Res Int, 2013. 2013: p. 942916.
Guo, C., et al., Silica nanoparticles induce oxidative stress, inflammation, and endothelial dysfunction in vitro via activation of the MAPK/Nrf2 pathway and nuclear factor-kappaB signaling. Int J Nanomedicine, 2015. 10: p. 1463-77.
Zhou, T., C.C. Chuang, and L. Zuo, Molecular Characterization of Reactive Oxygen Species in
D
8.
9.
TE
Myocardial Ischemia-Reperfusion Injury. Biomed Res Int, 2015. 2015: p. 864946. Bessemer, R.A., et al., Cardiorespiratory toxicity of environmentally relevant zinc oxide nanoparticles
10.
CE P
in the freshwater fish Catostomus commersonii. Nanotoxicology, 2015. 9(7): p. 861-70. Sheng, L., et al., Cardiac oxidative damage in mice following exposure to nanoparticulate titanium dioxide. J Biomed Mater Res A, 2013. 101(11): p. 3238-46. Nel, A., et al., Toxic potential of materials at the nanolevel. Science, 2006. 311(5761): p. 622-7.
12.
Chen, J., et al., Effects of titanium dioxide nano-particles on growth and some histological parameters
AC
11.
of zebrafish (Danio rerio) after a long-term exposure. Aquat Toxicol, 2011. 101(3-4): p. 493-9. 13.
Duan, Y., et al., The effects of nano-anatase TiO(2) on the activation of lactate dehydrogenase from rat heart. Biol Trace Elem Res, 2009. 130(2): p. 162-71.
14.
Bu, Q., et al., NMR-based metabonomic study of the sub-acute toxicity of titanium dioxide nanoparticles in rats after oral administration. Nanotechnology, 2010. 21(12): p. 125105.
15.
Faddah, L.M., et al., Biochemical responses of nanosize titanium dioxide in the heart of rats following administration of idepenone and quercetin. African Journal of Pharmacy and Pharmacology, 2013. 7(38): p. 2639-2651.
16.
Chandra, R.K., Excessive intake of zinc impairs immune responses. JAMA, 1984. 252(11): p. 1443-6.
17.
Colvin, V.L., The potential environmental impact of engineered nanomaterials. Nat Biotechnol, 2003. 21(10): p. 1166-70.
18.
Sharma, V., et al., Induction of oxidative stress, DNA damage and apoptosis in mouse liver after subacute oral exposure to zinc oxide nanoparticles. Mutat Res, 2012. 745(1-2): p. 84-91.
19.
Hillyer, J.F. and R.M. Albrecht, Gastrointestinal persorption and tissue distribution of differently sized colloidal gold nanoparticles. J Pharm Sci, 2001. 90(12): p. 1927-36.
ACCEPTED MANUSCRIPT
26 20.
Vandebriel, R.J. and W.H. De Jong, A review of mammalian toxicity of ZnO nanoparticles. Nanotechnol Sci Appl, 2012. 5: p. 61-71.
21.
Guo, D., et al., Reactive oxygen species-induced cytotoxic effects of zinc oxide nanoparticles in rat retinal ganglion cells. Toxicol In Vitro, 2013. 27(2): p. 731-8. Pasupuleti, S., et al., Toxicity of zinc oxide nanoparticles through oral route. Toxicol Ind Health, 2012.
T
22.
23.
Chuang, H.C., et al., Cardiopulmonary toxicity of pulmonary exposure to occupationally relevant zinc
SC R
oxide nanoparticles. Nanotoxicology, 2014. 8(6): p. 593-604. 24.
IP
28(8): p. 675-86.
Baky, N.A., et al., Induction of inflammation, DNA damage and apoptosis in rat heart after oral exposure to zinc oxide nanoparticles and the cardioprotective role of alpha-lipoic acid and vitamin E. Drug Res (Stuttg), 2013. 63(5): p. 228-36.
Suematsu, N., et al., Oxidative stress mediates tumor necrosis factor-alpha-induced mitochondrial
NU
25.
DNA damage and dysfunction in cardiac myocytes. Circulation, 2003. 107(10): p. 1418-23. 26.
de Ferranti, S.D. and N. Rifai, C-reactive protein: a nontraditional serum marker of cardiovascular
27.
MA
risk. Cardiovasc Pathol, 2007. 16(1): p. 14-21.
Hasenfuss, G., Alterations of calcium-regulatory proteins in heart failure. Cardiovasc Res, 1998. 37(2): p. 279-89.
Huang, T.H., H. Lee, and C.T. Kao, Evaluation of the genotoxicity of zinc oxide eugenol-based,
D
28.
27(12): p. 744-8. 29.
TE
calcium hydroxide-based, and epoxy resin-based root canal sealers by comet assay. J Endod, 2001.
Sharma, V., et al., DNA damaging potential of zinc oxide nanoparticles in human epidermal cells.
CE P
Toxicol Lett, 2009. 185(3): p. 211-8. 30.
Martinez, G.R., et al., Oxidative and alkylating damage in DNA. Mutat Res, 2003. 544(2-3): p. 115-27.
31.
Taju, G., et al., In vitro assay for the toxicity of silver nanoparticles using heart and gill cell lines of Catla catla and gill cell line of Labeo rohita. Comp Biochem Physiol C Toxicol Pharmacol, 2014. 161:
AC
p. 41-52. 32.
Kim, Y.S., et al., Subchronic oral toxicity of silver nanoparticles. Part Fibre Toxicol, 2010. 7: p. 20.
33.
Hadrup, N. and H.R. Lam, Oral toxicity of silver ions, silver nanoparticles and colloidal silver--a review. Regul Toxicol Pharmacol, 2014. 68(1): p. 1-7.
34.
Carlson, C., et al., Unique cellular interaction of silver nanoparticles: size-dependent generation of reactive oxygen species. J Phys Chem B, 2008. 112(43): p. 13608-19.
35.
AshaRani, P.V., et al., Cytotoxicity and genotoxicity of silver nanoparticles in human cells. ACS Nano, 2009. 3(2): p. 279-90.
36.
Hussain, S.M., et al., The interaction of manganese nanoparticles with PC-12 cells induces dopamine depletion. Toxicol Sci, 2006. 92(2): p. 456-63.
37.
Ebabe Elle, R., et al., Dietary exposure to silver nanoparticles in Sprague-Dawley rats: effects on oxidative stress and inflammation. Food Chem Toxicol, 2013. 60: p. 297-301.
38.
Korani, M., S.M. Rezayat, and S. Arbabi Bidgoli, Sub-chronic Dermal Toxicity of Silver Nanoparticles in Guinea Pig: Special Emphasis to Heart, Bone and Kidney Toxicities. Iran J Pharm Res, 2013. 12(3): p. 511-9.
27 39.
ACCEPTED MANUSCRIPT Joseph-Silverstein, J., et al., Basic fibroblast growth factor in the chick embryo: immunolocalization to striated muscle cells and their precursors. J Cell Biol, 1989. 108(6): p. 2459-66.
40.
Liao, S., et al., Biological functions of the low and high molecular weight protein isoforms of fibroblast growth factor-2 in cardiovascular development and disease. Dev Dyn, 2009. 238(2): p. 249-64. Tomanek, R.J., et al., VEGF and bFGF stimulate myocardial vascularization in embryonic chick. Am J
T
41.
Zheng, W., et al., Bradycardia-induced coronary angiogenesis is dependent on vascular endothelial growth factor. Circ Res, 1999. 85(2): p. 192-8.
43.
SC R
42.
IP
Physiol, 1998. 274(5 Pt 2): p. H1620-6.
Shvedova, A.A., et al., ESR evidence for in vivo formation of free radicals in tissue of mice exposed to single-walled carbon nanotubes. Free Radic Biol Med, 2014. 73: p. 154-65.
44.
Ge, C., et al., Acute pulmonary and moderate cardiovascular responses of spontaneously hypertensive
45.
NU
rats after exposure to single-wall carbon nanotubes. Nanotoxicology, 2012. 6(5): p. 526-42. Shvedova, A.A., et al., Antioxidant balance and free radical generation in vitamin e-deficient mice after dermal exposure to cumene hydroperoxide. Chem Res Toxicol, 2002. 15(11): p. 1451-9. Shvedova, A.A., et al., Mechanisms of carbon nanotube-induced toxicity: focus on oxidative stress.
MA
46.
Toxicol Appl Pharmacol, 2012. 261(2): p. 121-33. 47.
Lam, C.W., et al., A review of carbon nanotube toxicity and assessment of potential occupational and
Akbar, S. and A.A. Taimoor, Functionalization of carbon nanotubes: manufacturing techniques and
TE
48.
D
environmental health risks. Crit Rev Toxicol, 2006. 36(3): p. 189-217.
properties of customized nanocomponents for molecular-level technology. Recent Pat Nanotechnol, 2009. 3(2): p. 154-61.
Kreyling, W.G., et al., Size dependence of the translocation of inhaled iridium and carbon nanoparticle
CE P
49.
aggregates from the lung of rats to the blood and secondary target organs. Inhal Toxicol, 2009. 21 Suppl 1: p. 55-60. 50.
Reddy, A.R., et al., Evaluation of oxidative stress and anti-oxidant status in rat serum following
51.
AC
exposure of carbon nanotubes. Regul Toxicol Pharmacol, 2011. 59(2): p. 251-7. Matthews, I.P., et al., Maximal extent of translocation of single-walled carbon nanotubes from lung airways of the rat. Environ Toxicol Pharmacol, 2013. 35(3): p. 461-4. 52.
Urankar, R.N., et al., Expansion of cardiac ischemia/reperfusion injury after instillation of three forms of multi-walled carbon nanotubes. Part Fibre Toxicol, 2012. 9: p. 38.
53.
Vistnes, M., et al., Circulating cytokine levels in mice with heart failure are etiology dependent. J Appl Physiol (1985), 2010. 108(5): p. 1357-64.
54.
Bonaros, N., et al., CCR3- and CXCR4-mediated interactions regulate migration of CD34+ human bone marrow progenitors to ischemic myocardium and subsequent tissue repair. J Thorac Cardiovasc Surg, 2008. 136(4): p. 1044-53.
55.
Falcone, C., et al., Markers of eosinophilic inflammation and risk prediction in patients with coronary artery disease. Eur J Clin Invest, 2006. 36(4): p. 211-7.
56.
Stapleton, P.A., et al., Impairment of coronary arteriolar endothelium-dependent dilation after multiwalled carbon nanotube inhalation: a time-course study. Int J Mol Sci, 2012. 13(11): p. 13781-803.
ACCEPTED MANUSCRIPT
28 57.
Wierzbicki, M., et al., Carbon nanoparticles downregulate expression of basic fibroblast growth factor in the heart during embryogenesis. Int J Nanomedicine, 2013. 8: p. 3427-35.
58.
Murugesan, S., et al., Carbon inhibits vascular endothelial growth factor- and fibroblast growth factorpromoted angiogenesis. FEBS Lett, 2007. 581(6): p. 1157-60. Thompson, L.C., et al., C(6)(0) exposure augments cardiac ischemia/reperfusion injury and coronary
T
59.
60.
IP
artery contraction in Sprague Dawley rats. Toxicol Sci, 2014. 138(2): p. 365-78. Thompson, L.C., et al., Pulmonary instillation of multi-walled carbon nanotubes promotes coronary
61.
SC R
vasoconstriction and exacerbates injury in isolated hearts. Nanotoxicology, 2014. 8(1): p. 38-49. Martin, K.R., The chemistry of silica and its potential health benefits. J Nutr Health Aging, 2007. 11(2): p. 94-7. 62.
Duan, J., et al., Cardiovascular toxicity evaluation of silica nanoparticles in endothelial cells and
63.
NU
zebrafish model. Biomaterials, 2013. 34(23): p. 5853-62.
Chen, Z., et al., Age-related differences in pulmonary and cardiovascular responses to SiO2 nanoparticle inhalation: nanotoxicity has susceptible population. Environ Sci Technol, 2008. 42(23):
64.
MA
p. 8985-92.
Du, Z., et al., Cardiovascular toxicity of different sizes amorphous silica nanoparticles in rats after intratracheal instillation. Cardiovasc Toxicol, 2013. 13(3): p. 194-207. Gauderman, W.J., et al., The effect of air pollution on lung development from 10 to 18 years of age. N
D
65.
66.
TE
Engl J Med, 2004. 351(11): p. 1057-67.
Galagudza, M., et al., Passive targeting of ischemic-reperfused myocardium with adenosine-loaded silica nanoparticles. Int J Nanomedicine, 2012. 7: p. 1671-8. Hamilton, J.M., et al., Visuospatial deficits predict rate of cognitive decline in autopsy-verified
CE P
67.
dementia with Lewy bodies. Neuropsychology, 2008. 22(6): p. 729-37. 68.
Lin, Q., et al., Control of mouse cardiac morphogenesis and myogenesis by transcription factor MEF2C. Science, 1997. 276(5317): p. 1404-7. Illi, B., et al., Epigenetic histone modification and cardiovascular lineage programming in mouse
AC
69.
embryonic stem cells exposed to laminar shear stress. Circ Res, 2005. 96(5): p. 501-8. 70.
Pierpont, M.E., R.R. Markwald, and A.E. Lin, Genetic aspects of atrioventricular septal defects. Am J Med Genet, 2000. 97(4): p. 289-96.
71.
Liu, S.Y., et al., Toxicology Studies of a Superparamagnetic Iron Oxide Nanoparticle In Vivo. Advanced Materials Research, 2008. 47-50: p. 1097-1100.
72.
Mahmoudi, M., et al., Assessing the in vitro and in vivo toxicity of superparamagnetic iron oxide nanoparticles. Chem Rev, 2012. 112(4): p. 2323-38.
73.
Sun, J., et al., Cytotoxicity, permeability, and inflammation of metal oxide nanoparticles in human cardiac microvascular endothelial cells: cytotoxicity, permeability, and inflammation of metal oxide nanoparticles. Cell Biol Toxicol, 2011. 27(5): p. 333-42.
74.
Hong, S.C., et al., Subtle cytotoxicity and genotoxicity differences in superparamagnetic iron oxide nanoparticles coated with various functional groups. Int J Nanomedicine, 2011. 6: p. 3219-31.
ACCEPTED MANUSCRIPT
29 75.
Iversen, N.K., et al., Superparamagnetic iron oxide polyacrylic acid coated gamma-Fe2O3 nanoparticles do not affect kidney function but cause acute effect on the cardiovascular function in healthy mice. Toxicol Appl Pharmacol, 2013. 266(2): p. 276-88.
76.
Xu, J., et al., Acute toxicity of intravenously administered titanium dioxide nanoparticles in mice. PloS
Faddah, L., et al., Biochemical responses of nanosize titanium dioxide in the heart of rats following
IP
77.
T
one, 2013. 8(8): p. e70618.
administration of idepenone and quercetin LM Faddah*, Nayira A. Abdel Baky, Nouf M. Al-Rasheed
SC R
and Nawal M. Al-Rasheed. African Journal of Pharmacy and Pharmacology, 2013. 7(38): p. 26392651. 78.
Liu, H., et al., Biochemical toxicity of nano-anatase TiO2 particles in mice. Biological trace element research, 2009. 129(1-3): p. 170-180.
Sheng, L., et al., Cardiac oxidative damage in mice following exposure to nanoparticulate titanium
NU
79.
dioxide. Journal of Biomedical Materials Research Part A, 2013. 101(11): p. 3238-3246. 80.
Chen, J., et al., Effects of titanium dioxide nano-particles on growth and some histological parameters
81.
MA
of zebrafish (Danio rerio) after a long-term exposure. Aquatic Toxicology, 2011. 101(3): p. 493-499. Duan, Y., et al., The effects of nano-anatase TiO2 on the activation of lactate dehydrogenase from rat heart. Biological trace element research, 2009. 130(2): p. 162-171. Savi, M., et al., Titanium dioxide nanoparticles promote arrhythmias via a direct interaction with rat
D
82.
83.
TE
cardiac tissue. Part Fibre Toxicol, 2014. 11: p. 63. Wang, B., et al., Acute toxicity of nano-and micro-scale zinc powder in healthy adult mice. Toxicology letters, 2006. 161(2): p. 115-123.
Chuang, H.-C., et al., Cardiopulmonary toxicity of pulmonary exposure to occupationally relevant zinc
CE P
84.
oxide nanoparticles. Nanotoxicology, 2014. 8(6): p. 593-604. 85.
Bessemer, R.A., et al., Cardiorespiratory toxicity of environmentally relevant zinc oxide nanoparticles in the freshwater fish Catostomus commersonii. Nanotoxicology, 2015. 9(7): p. 861-870. Baky, N., et al., Induction of inflammation, DNA damage and apoptosis in rat heart after oral exposure
AC
86.
to zinc oxide nanoparticles and the cardioprotective role of α-lipoic acid and vitamin E. Drug research, 2013. 63(05): p. 228-236. 87.
Rathore, M., et al., Comparative in vivo assessment of the subacute toxicity of gold and silver nanoparticles. Journal of Nanoparticle Research, 2014. 16(4): p. 1-12.
88.
Elle, R.E., et al., Dietary exposure to silver nanoparticles in Sprague–Dawley rats: effects on oxidative stress and inflammation. Food and chemical toxicology, 2013. 60: p. 297-301.
89.
Taju, G., et al., In vitro assay for the toxicity of silver nanoparticles using heart and gill cell lines of Catla catla and gill cell line of Labeo rohita. Comparative Biochemistry and Physiology Part C: Toxicology & Pharmacology, 2014. 161: p. 41-52.
90.
Holland,
N.A.,
Intratracheal
instillation
of
silver
nanoparticles
exacerbates
cardiac
ischemia/reperfusion injury in male sprague-dawley rats. 2014. 91.
Roberts, J.R., et al., Pulmonary and cardiovascular responses of rats to inhalation of silver nanoparticles. Journal of Toxicology and Environmental Health, Part A, 2013. 76(11): p. 651-668.
ACCEPTED MANUSCRIPT
30 92.
Hotowy, A., et al., Silver nanoparticles administered to chicken affect VEGFA and FGF2 gene expression in breast muscle and heart. Nanoscale research letters, 2012. 7(1): p. 1-8.
93.
Cho, J.-G., et al., Stepwise embryonic toxicity of silver nanoparticles on Oryzias latipes. BioMed research international, 2013. 2013. Paula, M.M.d.S., et al., In vitro effect of silver nanoparticles on creatine kinase activity. Journal of the
T
94.
95.
IP
Brazilian Chemical Society, 2009. 20: p. 1556-1560.
Duan, J., et al., Low-dose exposure of silica nanoparticles induces cardiac dysfunction via neutrophil-
SC R
mediated inflammation and cardiac contraction in zebrafish embryos. Nanotoxicology, 2016. 10(5): p. 575-85. 96.
Ge, C., et al., Acute pulmonary and moderate cardiovascular responses of spontaneously hypertensive rats after exposure to single-wall carbon nanotubes. Nanotoxicology, 2012. 6(5): p. 526-542. Shvedova, A., et al., ESR evidence for in vivo formation of free radicals in tissue of mice exposed to
NU
97.
single-walled carbon nanotubes. Free Radical Biology and Medicine, 2014. 73: p. 154-165. 98.
Urankar, R.N., et al., Expansion of cardiac ischemia/reperfusion injury after instillation of three forms
99.
MA
of multi-walled carbon nanotubes. Particle and fibre toxicology, 2012. 9(1): p. 1. Chen, Z., et al., Age-related differences in pulmonary and cardiovascular responses to SiO2 nanoparticle inhalation: nanotoxicity has susceptible population. Environmental science & technology,
Aburawi, E.H., et al., Biocompatibility of Calcined Mesoporous Silica Particles with Ventricular
TE
100.
D
2008. 42(23): p. 8985-8992.
Myocyte Structure and Function. Chemical research in toxicology, 2012. 26(1): p. 26-36. 101.
Duan, J., et al., Cardiovascular toxicity evaluation of silica nanoparticles in endothelial cells and
102.
CE P
zebrafish model. Biomaterials, 2013. 34(23): p. 5853-5862. Du, Z., et al., Cardiovascular toxicity of different sizes amorphous silica nanoparticles in rats after intratracheal instillation. Cardiovascular toxicology, 2013. 13(3): p. 194-207. 103.
Borak, B., et al., In vivo study on the biodistribution of silica particles in the bodies of rats. Adv Clin
104.
AC
Exp Med, 2012. 21(1): p. 13-18. Sun, J., et al., Cytotoxicity, permeability, and inflammation of metal oxide nanoparticles in human cardiac microvascular endothelial cells. Cell biology and toxicology, 2011. 27(5): p. 333-342. 105.
Liu, S.Y., et al. Toxicology studies of a superparamagnetic iron oxide nanoparticle in vivo. in Advanced Materials Research. 2008. Trans Tech Publ.
106.
Xiong, F., et al., Cardioprotective activity of iron oxide nanoparticles. Sci Rep, 2015. 5: p. 8579.
107.
Nemmar, A., et al., Ultrasmall superparamagnetic iron oxide nanoparticles acutely promote thrombosis and cardiac oxidative stress and DNA damage in mice. Part Fibre Toxicol, 2016. 13(1): p. 22.
108.
Baratli, Y., et al., Impact of iron oxide nanoparticles on brain, heart, lung, liver and kidneys mitochondrial respiratory chain complexes activities and coupling. Toxicol In Vitro, 2013. 27(8): p. 2142-8.
ACCEPTED MANUSCRIPT
31
AC
CE P
TE
D
MA
NU
SC R
IP
T
Graphical abstract