ArticlePDF AvailableLiterature Review

Neuroprotective potential of cerium oxide nanoparticles for focal cerebral ischemic stroke

Authors:

Abstract and Figures

During the previous years, with the emerging of nanotechnology, the enormous capabilities of nanoparticles have drawn great attention from researchers in terms of their potentials in various aspects of pharmacology. Cerium oxide nanoparticles (nanoceria), considered as one of the most widely used nanomaterials, due to its tempting catalytic antioxidant properties, show a promising potential in diverse disorders, such as cerebral ischemic stroke (CIS), cancer, neurodegenerative and inflammatory diseases. Overwhelming generation of reactive oxygen species (ROS) and reactive nitrogen species (RNS) during cerebral ischemia and reperfusion periods is known to aggravate brain damage via sophisticated cellular and molecular mechanisms, and therefore exploration of the antioxidant capacities of nanoceria becomes a new approach in reducing cerebral ischemic injury. Furthermore, utilizing nanoceria as a drug carrier might display the propensity to overcome limitations or inefficacy of other conceivable neuroprotectants and exhibit synergistic effects. In this review, we emphasize on the principle features of nanoceria and current researches concerning nanoceria as a potential therapeutic agent or carrier in improving the prognosis of CIS.
Content may be subject to copyright.
480J Huazhong Univ Sci TechnolMed Sci 36(4):2016
Neuroprotective Potential of Cerium Oxide Nanoparticles for Focal
Cerebral Ischemic Stroke
Da ZHOU ( ), Ting FANG ( ), Lin-qing LU (陆林清), Li YI ( )#
Department of Neurology, Peking University Shenzhen Hospital, Shenzhen 518036, China
© Huazhong University of Science and Technology and Springer-Verlag Berlin Heidelberg 2016
Summary: During the previous years, with the emerging of nanotechnology, the enormous capabilities
of nanoparticles have drawn great attention from researchers in terms of their potentials in various as-
pects of pharmacology. Cerium oxide nanoparticles (nanoceria), considered as one of the most widely
used nanomaterials, due to its tempting catalytic antioxidant properties, show a promising potential in
diverse disorders, such as cerebral ischemic stroke (CIS), cancer, neurodegenerative and inflammatory
diseases. Overwhelming generation of reactive oxygen species (ROS) and reactive nitrogen species
(RNS) during cerebral ischemia and reperfusion periods is known to aggravate brain damage via sophis-
ticated cellular and molecular mechanisms, and therefore exploration of the antioxidant capacities of
nanoceria becomes a new approach in reducing cerebral ischemic injury. Furthermore, utilizing
nanoceria as a drug carrier might display the propensity to overcome limitations or inefficacy of other
conceivable neuroprotectants and exhibit synergistic effects. In this review, we emphasize on the princi-
ple features of nanoceria and current researches concerning nanoceria as a potential therapeutic agent or
carrier in improving the prognosis of CIS.
Key words: Cerium oxide nanoparticles; nanotechnology; reactive oxygen species; cerebral ischemic
stroke
Cerebral ischemic stroke (CIS) is considered as one
of the leading causes of severe disability, or even death
in the world. According to the latest statistics, it ranks the
fourth most common cause of death in the United
States[1]. The high incidence of death and disability
among CIS patients poses a significant burden on indi-
vidual patients, their families and society as a whole.
Under conditions of ischemia or reperfusion, the over-
whelming production of free radicals often leads to a
condition known as oxidative stress, which increases the
susceptibility of cerebral tissues to further injury.
Although cerium oxide nanoparticles (nanoceria)
have already been widely used in various fields, includ-
ing solar cells, oxygen pumps and gas sensors[2], their
applications on biomedical aspects still await more re-
search. Nanoceria can reversibly bind oxygen and switch
between Ce3+ (reduced) and Ce4+ (oxidized) states at
surface of particles depending on external stimuli, thus
producing a redox couple that exhibits catalytic antioxi-
dant functions[2, 3]. A large number of diseases have also
been demonstrated to be associated with dysregulation of
reactive oxygen species (ROS) levels. Recent studies
have implicated that naoceria might play an effective role
in anti-cancer[4, 5], anti-inflammation[6], neuro-protec-
tion[7–9], cardio-protection[10] therapies. Therefore, nano-
ceria have drawn increasing attention from researchers
throughout the world as a promising antioxidant agent in
many medical fields. Herein we mainly summarized po-
tential investigations and future prospects of nacoceria in
the protection against CIS.

Da ZHOU, E-mail: 18588475981@163.com
#Corresponding author, E-mail: yilitj@hotmail.com
1 ISCHEMIC STROKE AND ROLES OF OXIDA-
TIVE STRESS
1.1 Ischemic Stroke
Stroke is one of the biggest killers in the world and
accounts for nearly half of all hospitalized patients in
acute neurological cases. More than 80% of all strokes
are ischemic in origin. Presently, only recombinant tissue
plasminogen activator (rt-PA) has been approved by the
Food and Drug Administration (FDA) in US as the first
option in the clinical practice of acute CIS. The main
mechanism of rt-PA in treating acute CIS is its capability
of converting plasminogen to plasmin, thus breaking
down or dissolving blood clots so as to re-establish blood
flow reperfusion and oxygen supply in the ischemic
cerebral tissues, especially in penumbra areas. The new
protocol guideline suggests intravenous administration of
rt-PA be accomplished within 4.5 h from onset of symp-
toms[11]. However, in real clinical settings, only around
one-quarter of patients with ischemic stroke got admitted
to hospitals within this narrow therapeutic window[12, 13].
Additionally, other adverse effects such as increased risk
of intracerebral hemorrhage, ischemia-reperfusion injury,
and various contraindications have made the clinical ap-
plication of this agent less feasible. Therefore, invention
of alternative neuroprotective drugs that can efficiently
go across blood brain barrier (BBB) and confer protec-
tion to damaged ischemic tissues has become an immi-
nent issue.
Many attempts have been made so far, whereas few
successes have been achieved. Limitation of application
can be summarized as follows: (1) they are unable to
cross BBB efficiently; (2) they have unsolved, lurking
36(4):480-486,2016
J Huazhong Univ Sci TechnolMed Sci
DOI 10.1007/s11596-016-1612-9
J Huazhong Univ Sci TechnolMed Sci 36(4):2016 481
toxicity to biological systems; (3) they possess a short
half-life in blood circulation; (4) they lack the ability of
being selectively taken up by targeted cells or tissues; (5)
they have poor solubility in circulation and rapid elimi-
nation; and (6) some antioxidants such as SOD and cata-
lase (CAT), can scavenge only one type of free radicals,
whereas multiple types are generated during ischemia-
reperfusion periods, thus restricted effects are expected.
1.2 Roles of Oxidative Stress
Various complicated, bizarre, poorly understood
mechanisms such as excitotoxic injury, inflammation,
oxidative stress, nitrative stress, membrane depolariza-
tion are associated with cerebral ischemia[14, 15]. When
ischemia occurs, reactive oxygen species (ROS) and re-
active nitrogen species (RNS), especially the most preva-
lent ones, including hydrogen peroxide (H2O2), superox-
ide anion (O2
), hydroxyl radical (·OH), nitric oxide
(NO), are massively generated and accumulated, result-
ing in oxidative damage to cerebral tissues. Oxidative
damage could further induce even worse consequences
including cell apoptosis via nucleus damage, lipid
peroxidation, protein oxidation and so on[7, 16–18].
Free radicals, as intermediates or byproducts gener-
ated from sophisticated reactions in living cells, causing
the production of reactive oxygen and nitrogen species,
normally remain in a homeostatic state[19]. Within a nor-
mal range, ROS/RNS can not only help eliminate bio-
logical intermediate metabolic wastes, but also act as
essential mediators in various cell signal transduction
pathways. However, ROS/RNS can also potentially pose
deleterious effects on cells and generally be scavenged
by endogenous antioxidant systems, like enzymes includ-
ing CAT, superoxide dismutase (SOD), and glutathione
peroxidase (GSH-Px)[20, 21]. Under pathological circum-
stances, excessive accumulation of ROS and insufficient
supply of antioxidants lead to a condition, namely oxida-
tive stress. Oxidative stress has been proven to be in-
volved in a great deal of human disorders, including neu-
rodegenerative diseases, stroke, tumors, acute or chronic
inflammatory diseases. Consequently, antioxidants have
become a prevalent issue in the field of neurological sci-
ence due to their potent and promising application poten-
tial in CIS therapy[18, 22, 23].
2 INTRODUCTION OF ANTIOXIDANT ACTIVI-
TIES OF NANOCERIA
2.1 Antioxidant Activities of Nanoceria
Nanoceria, as one of the most noticeable catalysts,
offer the possibility to be investigated as a novel ap-
proach for treatment of CIS and other diseases involved
with oxidative stress. Cerium oxide, with specific fluorite
crystalline lattice structure, exhibits powerful antioxidant
capacity by reversibly binding oxygen and switching
between Ce3+ (reduced) and Ce4+ (oxidized) states (fig.
1). The valence shift creates a redox couple with a highly
reactive surface for scavenging free radicals, and this
characteristic allows nanoceria to participate in various
biological reactions[3, 24, 25]. It was reported that nanoceria
could react with various types of ROS, in particular with
H2O2 and O2
, and possessed so called SOD-mimetic and
CAT-mimetic activities. Generally, SOD transforms the
superoxide into H2O2 and oxygen under the catalytic
effect of Cu2+ and Zn2+ while H2O2 is further eliminated
by CAT into water and oxygen.
Fig. 1 The antioxidant mechanism of nanoceria
A very classic research carried out by Self. et al[26]
showed a dramatically increased generation of H2O2 lev-
els with the addition of nanoceria and the level of H2O2
was even greater with higher ratio of Ce3+/Ce4+ in the
nanoparticles. In order to test the SOD-mimetic activity
of nanoceria, they also used ceria nanopaticles with dif-
ferent concentrations and Ce3+/Ce4+ ratios to compete
with ferricytochrome C, and found that a higher ratio
displayed more efficacy in reducing superoxides, sug-
gesting nanoceria might be a more efficient catalyst than
SOD. Moreover, a recent study by Ganesana et al[27]
found that 1 μg/mL of nanoceria was equivalent to 527 U
of SOD in terms of superoxide scavenging activity. The
same team led by Self[28] also confirmed nanoceria ex-
hibited CAT-mimetic activity in a redox-state dependent
manner, and a lower ratio of Ce3+/Ce4+ was found to be
more powerful in the presence of hydrogen peroxide.
Besides, they proved that molecular oxygen was one of
the products of the catalytic reactions. Other than the
scavenging capabilities of H2O2 and O2
, several other
catalytic potentials were also recently confirmed. NO can
react with O2
to form a more dangerous and lethal free
radical, peroxynitrite (ONOO), under the facilitation of
SOD. According to the study, nanoceria might inhibit the
formation of NO or even directly act on ONOO[7]. Inter-
estingly, Self supported this theory later by obtaining the
proof that naoceria was able to exhibit their scavenging
feature for nitric oxide radical, which was especially
prominent in the presence of nanoparticles with a lower
ratio of Ce3+/Ce4+[29]. Hydroxyl radical, if exceeds ho-
meostatic threshold, could transform into one of the most
deleterious free radicals, causing unpredictable injuries
in the biological systems. Another study drew a conclu-
sion that nanceria exhibited hydroxyl radical scavenging
activity, which was likely influenced by the size and
Ce3+/Ce4+ ratio of the particles[30]. Asati et al[31] demon-
strated that polymer-coated nanoceria possessed a unique
intrinsic oxidase ability in a slightly acidic environment
without assistance from any oxidizing agents like hydro-
gen peroxide or enzymes such as peroxidase and oxi-
dases. Furthermore, although the precise mechanisms are
not fully understandable, the possible self-regenerating
activities make this nanoparticle an astonishingly pre-
cious and useful antioxidant to be potentially applied in
biological systems. Therefore, taken the above features
together, nanoceria has been studied in various disease
models both in vivo and vitro.
2.2 Application of Nanoceria in Cerebral Ischemic
482J Huazhong Univ Sci TechnolMed Sci 36(4):2016
Models
Due to the multipotent enzyme-like activities, as we
mentioned in the previous context, nanoceria displays the
potential to scavenge almost all the uncontrolled ROS.
Efforts aimed at eliminating ROS for the treatment of
various disorders associated with oxidative stress injury
are being explored. Given its biological tolerability and
minimal systemic toxicity[32], nanoceria is receiving
much more attention nowadays for the purpose of being
studied as therapeutic antioxidants. Free oxygen species
such as ROS and RNS play vital roles in the ischemia
and post-ischemia processes of CIS. Therefore, nanoceria,
with the ability of targeting these free radicals, has been
proposed as a potential neuroprotective reagent in cere-
bral ischemic or other injury models in vivo and vitro.
Das and his colleagues[33] presented that nanoceria
could successfully protect cells of adult rat spinal cords
from H2O2 induced oxidative damage. Administration of
nanoceria, prepared by microemulsion methods, to a se-
rum-free cell culture model of the adult rat spinal cord
showed good functional biocompatibility, obvious neu-
roprotection and retention of neuronal function, which is
basically in agreement with a previous study, reporting
that nanoceria had a protective effect against exogenous
natural pro-oxidant, glutamate for rodent nervous system
derived HT22 cells[34]. In conclusion, nanoceria may be
proven to be a potential new therapeutic strategy for
ischemic insults and other oxidative injuries in other neu-
rodegenerative diseases.
A very classic experiment conducted by Estevez et
al[7] demonstrated that nanoceria reduced the ischemic
cell death in a hippocampal brain slice of mouse model
of cerebral ischemia. They introduced nanoceria with
doses ranging from 0.1 to 2 μg/mL into the slices at the
beginning of ischemia, and a dose-dependent reduction
in cell death was observed. In order to investigate the
appropriate timing of nanoceria administration, 1 μg/mL
nanoceria were added to the ischemic brain slices 0, 2
and 4 h after ischemia, and neuroprotective effects were
decreased in a time-dependent manner. Moreover, they
also proposed that these protective effects were due to
the scavenging activities of ROS/NOS and reduction in
peroxynitrite (formed from the reaction of NO and su-
peroxide) might be a pivotal mechanism through which
the nanoceria exert functions during the ischemic and
post-ischemic damage periods.
Interestingly, a recent review[35] concluded that tar-
geting RNS, especially peroxynitrite (ONOO) and NO
might be a potent approach to ameliorate cerebral ische-
mia-reperfusion damage. Excessive generation of
ONOO from the reaction of superoxide and NO, were
discovered in both middle cerebral artery occlusion
(MCAO) animal models and blood samples obtained
from CIS patients. In terms of the complicated and bi-
zarre mechanisms, ONOO not only exhibited cytotoxic
effects through lipid peroxidation, tyrosine nitration,
DNA modification, but induced disruption of BBB via
MMPs modulation and FeTMPyP decomposition. Thus,
development of therapeutic agents with ONOO scav-
enging ability might be a promising and novel strategy
for CIS. Actually, several peroxynitrite scavengers, in-
cluding uric acid, resveratrol, curcumin, green tea cate-
chins and caffeic acid, have been demonstrated to exert
protective effects on ischemic stroke models either in
vivo or vitro. However, there is still lack of convincing
evidence to support the neuoprotective effect of
nanoceria in cerebral ischemia-reperfusion injury, and
therefore further studies are required.
Kim and his coworkers[8] proposed the protective
effect of nanoceria against CIS in a rat model. They syn-
thesized phospholipid-polyethylene glycol (PEG) encap-
sulated nanoceria to get better colloidal stability and
lower nonspecific uptake. They found that intravenously
introduced nanoceria with optimal doses (0.5 and 0.7
mg/kg) could significantly mitigate the brain damage in
vivo, as represented by reduced infarct volumes and
apoptotic cells, and penetrate ischemic cerebral tissues,
accumulate in the peri-infarct area. In vitro, they also
demonstrated nanoceria, when added into CHO-K1 cells
which had been pre-processed with tert-butyl hydroper-
oxide (tBHP), exhibited protective effect against ROS-
induced cell death. Taken together, this study elucidated
the neuroprotective effect of nanoceria after ischemic
injury and provided possibility of the application in fu-
ture clinical practice.
3 THE POTENTIAL OF NANOCERIA: PROS AND
CONS
In recent years, application of nanoparticles them-
selves or as therapeutic carriers in the treatment of CIS
has provided endless possibilities for researchers. It was
presented that pharmacokinetics of nanoparticles in bio-
logical circumstances were greatly influenced by their
size, dosage, solubility, surface chemical modification[36].
Surface modifications with biocompatible and biode-
gradable polymers such as polyethylene glycol (PEG),
polybutylcyanoacrylate (PBCA) and poly lactic acid-co-
glycolic acid (PLGA) could remit nanoparticles from
elimination by the reticuloendothelial system (RES),
prolong systemic circulation time, increase permeability
to BBB and maintain satisfactory, sustained drug re-
lease[37, 38].
Several studies suggested that nanoparticles with
surface modifications could assist delivery of exogenous
agents such as SOD to exert biological functions. For
example, Reddy et al[39] demonstrated that PLGA
nanoparticles encapsulated SOD, when introduced into
rats of MCAO via the intracarotid route, dramatically
ameliorated ischemic insult. On the contrary, the simple
use of SOD did not show significant neuroprotective
effects. In another literature[40], N-methyl-D-aspartate
receptor 1 (NR1) targeted antibody was decorated on the
surface of SOD-loaded PBCA nanoparticles and this
composite, not only showed better permeability across
BBB and localization into cerebral tissue, but also sug-
gested efficient neuro-protection with evidence of obvi-
ously less infarct volume, reduced inflammatory media-
tors, decreased free radicals and improved neurological
behavior.
Nanoceria, as a vital component of the nanoparticle
family, has emerged as a novel therapeutic agent or vec-
tor for various intractable diseases, including cancer,
neurodegenerative disorders, ischemic stroke, radiation
associated injury[18], cardiovascular disorders, retinal
disorders by itself or as a drug carrier (table 1). However,
to date, nanoceria targeted delivery of antioxidants or
other neuroprotectives to the brain in the context of
J Huazhong Univ Sci TechnolMed Sci 36(4):2016 483
ischemia-reperfusion has not been elucidated yet.
Table 1 Examples of potential applications of nanoceria in other oxidative stress-associated diseases
Subjects Protocol of syn-
thesis
Doses Size Method of
entry
Duration of ob-
servation
Outcome References
1 Retinal
diseases
(1) SD rats Simple wet
chemistry
methods
1 µmol/L
or 0.344
ng to 1
mmol/L
or 344 ng
3–5 nm IV injec-
tion
1 h to 120 days
post-injection
Preferentially
taken up; non-toxic
effects on retinal
functions
[41, 42]
(2) Vldlr
knockout mice
Simple wet
chemistry
methods
172 ng 3–5 nmIV injec-
tion
1 to 4 weeks Inhibit the rise in
ROS/VEGF/the
formation of ne-
ovascular tufts
[43]
2 Neurode-
generative
disease
(1) Aβ (25–35)
incubated human
neuroblastoma-
SH-SY5Y cells
(ATCC)
PEG-CNPs-Aβ
Ab (CNPs-Ab)
200
nmol/L
3–5 nm / 20 h Non-toxic to neu-
ron cells; selec-
tively delivered to
Aβ plaques
[44]
(2) Murine
model of EAE
Citrate/EDTA
coated CeNP
10, 20
and 30
mg/kg
2.9 nm IV via tail
vein
/ Able to penetrate
brain, reduce ROS,
alleviate clinical
symptoms
[9]
3 Cardio-
myopathy
(1) MCP-1 trans-
genic mice
NanoActive™
cerium oxide
nanoparticles
(NanoScale mate-
rials, Inc, USA)
15 nmol 7 nm IV via tail
vein
Twice a week for
2 weeks
Protect against the
progression of car-
diac dysfunction
and remodeling
[10]
(2) CPC / 10, 25,
and 50
μg/mL
5–8 nm / 1, 3 and 7 days Protect CPC from
H2O2 induced
cytotoxicity
[45]
4 Cancer (1) Human bron-
choalveolar carci-
noma-derived cell
line (A549)
Homogenous
nucleation
method
3.5, 10.5,
and 23.3
μg/mL
20 nm / 24, 48 and 72 h Decrease cell vi-
ability significantly
[46]
(2) Squamous
tumor cells SCL-
1/ human dermal
fibroblasts/ myo-
fibroblastic cells
Dextran-coated
nanoparticles
50, 150,
300
μmol/L
5 nm / 24/48 h Increase ROS lev-
els; lower the inva-
sive capacity
of tumor cells
[47, 48]
5 Chronic
inflamma-
tion
J774A.1 murine
macrophages
Simple wet chem-
istry methods
0 to 10
μmol/L
5 nm / 24 h nanoceria
pretreated; vari-
ous time sets for
observation
Protect against
ROS damage; sup-
press iNOS protein
levels
[6]
IV: intravitreal; SD: Sprague-Dawley; EAE: experimental autoimmune encephalomyelitis; CPC: cardiac progenitor cells; MCP:
monocyte chemoattractant protein
Many studies reported that nanoceria were biocom-
patible and able to penetrate brain tissues. For example,
to investigate whether nanoceria could cross the BBB
and further exhibit the free radicals scavenging abilities,
Portioli et al[49] injected in-house synthesized nanoceria
into living animal models and 24 h later, they found the
presence of fluorescein isothiocyanate conjugated
nanoceria, also in limited amount, in the cerebral tissues
under confocal microscopy and electron microscopy.
Heckman et al[9] demonstrated that custom-synthesized
nanoceria, when introduced intravenously to mice with
autoimmune encephalomyelitis of multiple sclerosis, was
able to pass through the brain to lower the level of ROS
and ameliorate clinical symptoms. Another report pre-
sented by Kim[8] also analyzed the biodistribution of
nanoceria in a rat ischemic stroke model and confirmed
that intravitreal administered PEG-encapsulated particles
were increased markedly in the penumbra area of the
ischemic hemisphere.
Many pilot studies have proven that nanoceria is
non-toxic. Hirst and colleagues[6] introduced nanoceria
into mice and tracked them for one month; the result
revealed that nanoceria were well tolerated and no obvi-
ous toxicity was found. Additionally, a recent published
paper also suggested that nanoceria only selectively tar-
geted cancerous cells, but normal structures in the organ-
ism were exempted from them[32]. However, in contrast
to the above positive evidence, some researchers re-
484J Huazhong Univ Sci TechnolMed Sci 36(4):2016
vealed relatively unsatisfactory aspects of nanoceria. Ma
et al[50] concluded that exposure of nanceria to rats in-
duced pulmonary inflammation and damage, possibly via
alveolar macrophages involved cellular signaling path-
ways. Later, the same laboratory conducted another ex-
periment aiming at clarifying the mechanisms underlying
the pulmonary injury caused by intra-tracheal instillation
of nanoceria; the results showed that nanoceria contrib-
uted to the over-expression of a variety of pulmonary
fibrosis-associated mediators, including hydroxyproline,
TGF-β1, OPN, MMPs, some of which were increased in
a time- and/or dose-dependent manner[51]. Furthermore, it
is worth noticing that numerous parameters, such as sur-
face charge, pH, particle size, surface coating, and ratio
of Ce3+/Ce4+ might endow naoceria with distinct toxicity,
stability and even catalytic abilities[4, 52–55]. Given the
possible application of nanoceria in central nervous sys-
tem (CNS), concerns regarding neurotoxicity have arisen,
although no overt adverse effect on neuronal cells or
cerebral tissues in neurodegenerative or ischemic models
has been reported so far.
Das et al[33] reported the possible autoregenerative
mechanism of nanoceria. As has been stated before,
nanoceria exhibited both SOD-mimetic and CAT-
mimetic activities. Therefore, when we combine them
together, a possible non-stop recycle reaction might oc-
cur, which may render it to become a valuable antioxi-
dant with self-cycling potential. This new antioxidant
candidate would be different from other antioxidants in
that no repetitive dose is needed. Furthermore, nanoceria
could also scavenge other free radicals, such as nitric
oxide radical, peroxynitrite anion and even hydroxyl
radical, all of which are well-known to exacerbate neu-
ronal insult and cytotoxicity in cerebral ischemia-
reperfusion period (fig. 2).
Fig. 2 Potential advantages for application of nanoceria in
cerebral ischemic stroke
Similar to other nanoparticles, novel development of
nanoceria-based drug delivery or targeting systems are
also appearing in recent years. A research group pro-
posed to use nanoceria as a carrier for carboxybenzene-
sulfonamide, a type of human carbonic anhydrase, to
investigate the potential application in glaucoma therapy;
successful derivatization and therapeutic effects were
later observed both in vivo and vitro[56]. In another study,
transferrin-conjugated nanoceria were demonstrated to
improve selective uptake by cancer cells[57]. Furthermore,
in terms of neurodegenerative disorders, Li et al[58] de-
scribed the synergistic protective effects were obtained
when nanoceria capped H2O2-responsive controlled drug
release system was applied for the treatment of Alz-
heimer’s disease. Taken together, it is easy to get
nanoceria modified due to the ultra-small particle size
and unique chemical structure. Nanoceria exhibit better
biocompatibility, longer survival time in systemic circu-
lation and better permeability across microcirculation
after surface coating/modification with hydrophilic or
amphiphilic biodegradable polymers such as PEG and
PLGA. Additionally, when combined or incorporated
with other targeting agents that pose affinity to specific
protein, cell or tissue, nanoceria might exert more strik-
ing synergistic effects on enormous disease models. Un-
fortunately, there are only a handful studies concerning
nanoceria application in ischemic stroke, and more in-
vestigations are anticipated.
4 CONCLUSIONS AND PROSPECTS
CIS has been demonstrated to be associated with
multiple bizarre, poorly understandable pathological
mechanisms such as excitotoxic injury, inflammation,
oxidative stress, nitrative stress and apoptosis. Many of
these mechanisms are influenced by free radicals includ-
ing H2O2, O2
, HO, NO and ONOO. Overwhelming
generation and accumulation of these free radicals during
the ischemia-reperfusion periods may further exacerbate
ischemic damage to cerebral tissues. Given that, target-
ing the deleterious free radicals may provide promising
advances in ameliorating insults and improving out-
comes in clinical research and therapy of CIS. Nanoceria
possess unique antioxidant features owing to coexistence
of Ce3+ (reduced) and Ce4+ (oxidized) ions, rapid shifting
between these two states and oxygen-vacancy defects.
Previous studies have also confirmed that nanoceria have
multiple free racial scavenging enzyme-like properties.
Furthermore, good biocompatibility, longer systemic
circulation, better BBB permeability are all vital factors
for any administered therapeutic agents to enter brain
and exert efficient functions in cerebral ischemia, which
could be easily achieved through surface modification
with biodegradable and non-toxic polymers, such as PEG,
PLGA. Utilizing polymermodified nanoparticles as drug
carriers has indicated enormous potential in the treatment
of CIS, whereas evidence concerning nanoceria is still
lacking.
Notwithstanding, a broad range of issues concerning
toxicological, biological and pharmacological profiles
still needs to be addressed. Contrary to the majority of
published literature, some presented toxicity and poor
biocompatibility of nanoceria in vivo or vitro, which
could possibly be explained by different synthetic proto-
cols, thus with distinct particle sizes, chemical or physi-
cal structures, surface modifications; all of these features
might exert undefined, equivocal effects on its pharma-
cokinetics including absorption, distribution, metabolism,
excretion and catalytic properties in biological systems.
In addition, methodological differences in administered
doses, routes or even individual skills might alter its be-
havior and therapeutic effect. It has been suggested that
nanoceria could exert pro-oxidant effects, thus increasing
the generation of ROS, or even inducing apoptosis. Al-
though explanation for this discrepancy from well-
known antioxidant features has not been fully elucidated
yet, environmental pH, especially an acidic milieu, is
likely to transform nanoceria into oxidants[4, 26]. More-
over, pro-oxidants might be provoked due to a series of
signal transduction cascades induced by possible interac-
tion between nanoceria and different cells. Therefore,
more detailed and systematic investigations based on
J Huazhong Univ Sci TechnolMed Sci 36(4):2016 485
standardized protocols of synthesis and utility, toxicol-
ogy and biocompatibility in experimental models which
mimic biological internal environment are required, es-
pecially if our definitive purpose is to put nanoceria onto
clinical application, such as in treatment of ischemic
stroke and other neurodegenerative diseases.
Preclinical evidence has clarified that some peptides
possess pivotal roles in the pathological cascades during
ischemia-reperfusion periods; and therefore inhibition or
promotion of one or more peptides might exert notable
efficacy in the therapy of cerebral ischemia, particularly
in the context of inflammation, edema and apoptosis.
Up-regulation of integrin alpha v beta 3 has been re-
ported in the context of cerebral ischemia, and selective
inhibition of this integrin showed significant improve-
ment after ischemic damage in vivo[59, 60]. Kim et al[8]
also concluded that PEG-coated nanoceria conferred
protection to the cerebral tissues. Therefore, continued
efforts will be needed in future research, intending to test
and analyze the neuroprotective effects of alpha v beta 3
inhibitor and nanoceria separately in acute ischemic
stroke models. Furthermore, we propose utilizing possi-
ble biological composite carrier alpha v beta 3 inhibitor-
CeO2-NPs to investigate its biocompatibility, bio-
distribution, stability, and the synergistic effect against
ischemic stroke in vivo, hoping that it could serve as a
potential new type of highly efficient, targeted control-
released carrier to deliver neural protective drugs for the
treatment of CIS.
Conflict of Interest Statement
The authors declare that there is no conflict of interest
with any financial organization or corporation or individual that
can inappropriately influence this work
REFERENCES:
1 Towfighi A, Saver JL. Stroke declines from third to fourth
leading cause of death in the United States: historical
perspective and challenges ahead. Stroke, 2011,42(8):
2351-2355
2 Celardo I, Traversa E, Ghibelli L. Cerium oxide nanopa-
rticles: a promise for applications in therapy. J Exp Ther
Oncol, 2011,9(1):47-51
3 CelardoI, Pedersen JZ, Traversa E, et al. Pharmacological
potential of cerium oxide nanoparticles. Nanoscale,
2011,3(4):1411-1420
4 Wason MS, Colon J, Das S, et al.Sensitization of
pancreatic cancer cells to radiation by cerium oxide
nanoparticle-induced ROS production. Nanomedicine,
2013,9(4):558-569
5 Gao Y, Chen K, Ma JL, et al. Cerium oxide nanoparticles
in cancer. Onco Targets Ther, 2014,7:835-840
6 Hirst SM, Karakoti AS, Tyler RD, et al. Anti-
inflammatory properties of cerium oxide nanoparticles.
Small, 2009,5(24):2848-2856
7 Estevez AY, Pritchard S, Harper K, et al. Neuroprotective
mechanisms of cerium oxide nanoparticles in a mouse
hippocampal brain slice model of ischemia. Free Radic
Biol Med, 2011,51(6):1155-1163
8 Kim CK, Kim T, Choi IY, et al. Ceria nanoparticles that
can protect against ischemic stroke. Angew Chem Int Ed
Engl, 2012,51(44):11 039-11 043
9 Heckman KL, DeCoteau W, Estevez A, et al. Custom
cerium oxide nanoparticles protect against a free radical
mediated autoimmune degenerative disease in the brain.
ACS Nano, 2013,7(12):10 582-10 596
10 Niu J, Azfer A, Rogers LM, et al. Cardioprotective effects
of cerium oxide nanoparticles in a transgenic murine
model of cardiomyopathy. Cardiovasc Res,
2007,73(3):549-559
11 Demaerschalk BM, Kleindorfer DO, Adeoye OM, et al.
Scientific Rationale for the Inclusion and Exclusion
Criteria for Intravenous Alteplase in Acute Ischemic
Stroke: A Statement for Healthcare Professionals From the
American Heart Association/American Stroke Association.
Stroke, 2016,47(2):581-641
12 Davis SM, DonnanGA. 4.5 hours: the new time window
for tissue plasminogen activator in stroke. Stroke,
2009,40(6):2266-2267
13 Messé SR, Fonarow GC, Smith EE, et al. Use of tissue-
type plasminogen activator before and after publication of
the European Cooperative Acute Stroke Study III in Get
With The Guidelines-Stroke. Circ Cardiovasc Qual
Outcomes, 2012,5(3):321-326
14 Doyle KP, Simon RP, Stenzel-Poore MP. Mechanisms of
ischemic brain damage. Neuropharmacology, 2008,55(3):
310-318
15 Wang D, Yuan X, Liu T, et al. Neuroprotective activity of
lavender oil on transient focal cerebral ischemia in mice.
Molecules, 2012,17(8):9803-9817
16 Chan PH. Reactive oxygen radicals in signaling and
damage in the ischemic brain. J Cereb Blood Flow Metab,
2001,21(1):2-14
17 Mehta SH, Webb RC, Ergul A, et al. Neuroprotection by
tempol in a model of iron-induced oxidative stress in acute
ischemic stroke. Am J Physiol Regul Integr Comp Physiol,
2004,286(2):R283-288
18 Nakamura T, Kume T, Katsuki H, et al. Protective effect
of serofendic acid on ischemic injury induced by
occlusion of the middle cerebral artery in rats. Eur J
Pharmacol, 2008,586(1-3):151-155
19 Ray PD, Huang BW, Tsuji Y. Reactive oxygen species
(ROS) homeostasis and redox regulation in cellular
signaling. Cell Signal, 2012,24(5):981-990
20 Gutteridge JM, Halliwell B. Comments on review of Free
Radicals in Biology and Medicine, second edition, by
Barry Halliwell and John M. C. Gutteridge. Free Radic
Biol Med, 1992,12(1):93-95
21 Matsuda S, Umeda M, Uchida H, et al. Alterations of
oxidative stress markers and apoptosis markers in the
striatum after transient focal cerebral ischemia in rats. J
Neural Transm, 2009,116(4):395-404
22 Lee B, Clarke D, Al Ahmad A, et al. Perlecan domain V is
neuroprotective and proangiogenic following ischemic
stroke in rodents. J Clin Invest, 2011,121(8):3005-3023
23 Rodrigo R, Fernández-Gajardo R, Gutiérrez R, et al.
Oxidative stress and pathophysiology of ischemic stroke:
novel therapeutic opportunities. CNS Neurol Disord Drug
Targets, 2013,12(5):698-714
24 Celardo I, De Nicola M, Mandoli C, et al. Ce(3)+ ions
determine redox-dependent anti-apoptotic effect of cerium
oxide nanoparticles. ACS Nano, 2011,5(6):4537-4549
25 Neumann G, Hicks J. Effects of Cerium and Aluminum in
Cerium-Containing Hierarchical HZSM-5 Catalysts for
Biomass Upgrading. Topics in Catalysis, 2012,55(3-
4):196-208
26 Korsvik C, Patil S, Seal S, et al. Superoxide dismutase
mimetic properties exhibited by vacancy engineered ceria
486J Huazhong Univ Sci TechnolMed Sci 36(4):2016
nanoparticles. Chem Commun (Camb), 2007,10:1056-
1058
27 Ganesana M, Erlichman JS, Andreescu S. Real-time
monitoring of superoxide accumulation and antioxidant
activity in a brain slice model using an electrochemical
cytochrome c biosensor. Free Radic Biol Med,
2012,53(12):2240-2249
28 Pirmohamed T, Dowding JM, Singh S, et al. Nanoceria
exhibit redox state-dependent catalase mimetic activity.
Chem Commun (Camb), 2010,46(16):2736-2738
29 Dowding JM, Dosani T, Kumar A, et al. Cerium oxide
nanoparticles scavenge nitric oxide radical (NO). Chem
Commun (Camb), 2012,48(40):4896-4898
30 Xue Y, Luan QF, Yang D, et al. Direct evidence for
hydroxyl radical scavenging activity of cerium oxide
nanoparticles. J Phys Chem C, 2011,115(11):4433-4438
31 Asati A, Santra S, Kaittanis C, et al. Oxidase-like activity
of polymer-coated cerium oxide nanoparticles. Angew
Chem Int Ed Engl, 2009,48(13):2308-2312
32 Wang Y, Yang F, Zhang HX, et al. Cuprous oxide
nanoparticles inhibit the growth and metastasis of
melanoma by targeting mitochondria. Cell Death Dis,
2013,4:e783
33 Das M, Patil S, Bhargava N, et al. Auto-catalytic ceria
nanoparticles offer neuroprotection to adult rat spinal cord
neurons. Biomaterials, 2007,28(10):1918-1925
34 Schubert D, Dargusch R, Raitano J, et al. Cerium and
yttrium oxide nanoparticles are neuroprotective. Biochem
Biophys Res Commun, 2006,342(1):86-91
35 Chen XM, Chen HS, Xu MJ, et al. Targeting reactive
nitrogen species: a promising therapeutic strategy for
cerebral ischemia-reperfusion injury. Acta Pharmacol Sin,
2013,34(1):67-77
36 Pinzon-Daza ML, Campia I, Kopecka J, et al.
Nanoparticle- and liposome-carried drugs: new strategies
for active targeting and drug delivery across blood-brain
barrier. Curr Drug Metab, 2013,14(6):625-640
37 Matoba T, Egashira K. Nanoparticle-mediated drug
delivery system for cardiovascular disease. Int Heart J,
2014,55(4):281-286
38 Thompson BJ, Ronaldson PT. Drug delivery to the
ischemic brain. Adv Pharmacol, 2014,71:165-202
39 Reddy MK, Labhasetwar V. Nanoparticle-mediated
delivery of superoxide dismutase to the brain: an effective
strategy to reduce ischemia-reperfusion injury. Faseb J,
2009,23(5):1384-1395
40 Yun X, Maximov VD, Yu J, et al. Nanoparticles for
targeted delivery of antioxidant enzymes to the brain after
cerebral ischemia and reperfusion injury. J Cereb Blood
Flow Metab, 2013,33(4):583-592
41 Karakoti AS, Monteiro-Riviere NA, Aggarwal R, et al.
Nanoceria as Antioxidant: Synthesis and Biomedical
Applications. JOM (1989), 2008,60(3):33-37
42 Wong LL, Hirst SM, Pye QN, et al. Catalytic nanoceria
are preferentially retained in the rat retina and are not
cytotoxic after intravitreal injection. PLoS One,
2013,8(3):e58431
43 Zhou X, Wong LL, Karakoti AS, et al. Nanoceria inhibit
the development and promote the regression of pathologic
retinal neovascularization in the Vldlr knockout mouse.
PLoS One, 2011,6(2):e16733
44 Cimini A, D'Angelo B, Das S, et al. Antibody-conjugated
PEGylated cerium oxide nanoparticles for specific
targeting of Abeta aggregates modulate neuronal survival
pathways. Acta Biomater, 2012,8(6):2056-2067
45 Pagliari F, Mandoli C, Forte G, et al. Cerium oxide
nanoparticles protect cardiac progenitor cells from
oxidative stress. ACS Nano, 2012,6(5):3767-3775
46 Lin W, Huang YW, Zhou XD, et al. Toxicity of cerium
oxide nanoparticles in human lung cancer cells. Int J
Toxicol, 2006,25(6):451-457
47 Alili L, Sack M, Karakoti AS, et al. Combined cytotoxic
and anti-invasive properties of redox-active nanoparticles
in tumor-stroma interactions. Biomaterials, 2011,32(11):
2918-2929
48 Karakoti AS, Satyanarayana VNTK, Suresh Babu K, et al.
Direct Synthesis of Nanoceria in Aqueous Polyhydroxyl
Solutions. J Phys Chem C, 2007,111(46):17232-17240
49 Portioli C, Benati D, Pii Y, et al. Short-term biodistr-
ibution of cerium oxide nanoparticles in mice: focus on
brain parenchyma. Nanosci Nanotechnol Lett, 2013,5(11):
1174-1181
50 Ma JY, Mercer RR, Barger M, et al. Cerium oxide
nanoparticle-induced pulmonary inflammation and
alveolar macrophage functional change in rats.
Nanotoxicology, 2011,5(3):312-325
51 Ma JY, Zhao H, Mercer RR, et al. Induction of pulmonary
fibrosis by cerium oxide nanoparticles. Toxicol Appl
Pharmacol, 2012,262(3):255-264
52 Perez JM, Asati A, Nath S, et al. Synthesis of
biocompatible dextran-coated nanoceria with pH-
dependent antioxidant properties. Small, 2008,4(5):552-
556
53 Asati A, Santra S, Kaittanis C, et al. Surface-charge-
dependent cell localization and cytotoxicity of cerium
oxide nanoparticles. ACS Nano, 2010,4(9):5321-5331
54 Karakoti AS, Munusamy P, Hostetler K, et al. Preparation
and Characterization Challenges to Understanding
Environmental and Biological Impacts of Nanoparticles.
Surf Interface Anal, 2012,44(5):882-889
55 Dowding JM, Das S, Kumar A, et al. Cellular interaction
and toxicity depend on physicochemical properties and
surface modification of redox-active nanomaterials. ACS
Nano, 2013,7(6):4855-4868
56 Patil S, Reshetnikov S, Haldar MK, et al. Surface-
Derivatized Nanoceria with Human Carbonic Anhydrase
II Inhibitors and Fluorophores: A Potential Drug
Delivery Device. J Phys Chem C, 2007,111(24):8437-
8442
57 Vincent A, Babu S, Heckert E, et al. Protonated
nanoparticle surface governing ligand tethering and
cellular targeting. ACS Nano, 2009,3(5):1203-1211
58 Li M, Shi P, Xu C, et al. Cerium oxide caged metal
chelator: anti-aggregation and anti-oxidation integrated
H2O2-responsive controlled drug release for potential
Alzheimer's disease treatment. Chem Sci, 2013,4(6):2536-
2542
59 Bi JJ, Yi L. Effects of integrins and integrin alphavbeta3
inhibitor on angiogenesis in cerebral ischemic stroke. J
Huazhong Univ Sci Technolog Med Sci, 2014,34(3):299-
305
60 Shimamura N, Matchett G, Yatsushige H, et al. Inhibition
of integrin alphavbeta3 ameliorates focal cerebral
ischemic damage in the rat middle cerebral artery
occlusion model. Stroke, 2006,37(7):1902-1909
(Received June 2, 2015; revised June 15, 2016)
... ROS, a series of oxygen intermediates, leads to lipid peroxidation damage in the brain tissues (Xiang et al., 2021). Some endogenous enzymes and non-enzymes such as superoxide dismutase (SOD) and glutathione (GSH) can be involved in tissue repair after ischemia-reperfusion injury (Margaill et al., 2005;Zhou et al., 2016). These enzymes are encoded by specific genes that carry antioxidant response elements in promoters (Zhang et al., 2015). ...
... Oxidative stress is responsible for the cerebral ischemiareperfusion injury that leads to apoptosis, autophagy, and necrosis of the brain cells (Deng et al., 2016). Excess ROS produced due to oxidative stress reacts rapidly with several sites on the cytoplasmic membrane, resulting in membrane protein oxidation and lipid peroxidation (Kahles and Brandes, 2012;Zhou et al., 2016). Toxic products of lipid peroxidation, such as malondialdehyde (MDA), have longer half-lives than free radicals (Mao et al., 1999). ...
Article
Full-text available
Background: Oxidative stress is associated with the pathogenesis of ischemic stroke (±)-5-bromo-2-(5-fluoro-1-hydroxyamyl) benzoate (BFB) is a novel compound modified by dl-3-n-butylphthalide (NBP). Here, we hypothesized that BFB may protect the PC12 cells against H 2 O 2 -induced oxidative stress injury through activation of the Nrf2 pathway. Methods: We measured the cell viability and levels of lactate dehydrogenase (LDH), malondialdehyde (MDA), glutathione (GSH), and reactive oxygen species (ROS) to determine the construction of the H 2 O 2 -induced models of oxidative stress in PC12 cells. Additionally, apoptotic cell death, mitochondrial membrane potential, and cellular morphology were examined to determine the effect of BFB on oxidative stress injury in H 2 O 2 -treated PC12 cells. The expression levels of Nrf2-related and autophagy-related genes and proteins were detected using real time quantative PCR (RT-qPCR), Western Blot, and immunofluorescence analyses. Results: Our study showed that BFB treatment reduced the elevated levels of MDA, LDH, and ROS, and decreased cell viability and GSH in H 2 O 2 -treated PC12 cells. We also observed the elevated expression of Nrf2 pathway-related factors and intranuclear transitions and found that Nrf2 inhibitors (ML385) could block the protective effect of BFB. The inhibitory effect of BFB on oxidative stress may be partially regulated by Nrf2 activation, and the initiation and induction of autophagy. Conclusion: BFB inhibited H 2 O 2 -induced oxidative stress injury in PC12 cells by activating the Nrf2 pathway, initiating and inducing autophagy, suggesting that BFB may be a promising therapeutic agent in treating neurological disorders like cerebral ischemia.
... 153 For example, a work reported that cerium oxide nanoparticles were able to scavenge ROS and reduce inflammation in a rat model of acute lung injury. 154 While nanoparticles show promise as anti-inflammatory agents, it is important to note that further research is needed in order fully to understand their potential benefits and risks. It is important to ensure that any nanoparticles used in medical applications are safe and do not cause unintended harm to patients. ...
Article
Full-text available
Background Conventional nanoparticle synthesis methods involve harsh conditions, high costs, and environmental pollution. In this context, researchers are actively searching for sustainable, eco-friendly alternatives to conventional chemical synthesis methods. This has led to the development of green synthesis procedures among which the exploration of the plant-mediated synthesis of nanoparticles experienced a great development. Especially, because plant extracts can work as reducing and stabilizing agents. This opens up new possibilities for cost-effective, environmentally-friendly nanoparticle synthesis with enhanced size uniformity and stability. Moreover, bio-inspired nanoparticles derived from plants exhibit intriguing pharmacological properties, making them highly promising for use in medical applications due to their biocompatibility and nano-dimension. Objective This study investigates the role of specific phytochemicals, such as phenolic compounds, terpenoids, and proteins, in plant-mediated nanoparticle synthesis together with their influence on particle size, stability, and properties. Additionally, we highlight the potential applications of these bio-derived nanoparticles, particularly with regard to drug delivery, disease management, agriculture, bioremediation, and application in other industries. Methodology Extensive research on scientific databases identified green synthesis methods, specifically plant-mediated synthesis, with a focus on understanding the contributions of phytochemicals like phenolic compounds, terpenoids, and proteins. The database search covered the field’s development over the past 15 years. Results Insights gained from this exploration highlight plant-mediated green synthesis for cost-effective nanoparticle production with significant pharmacological properties. Utilizing renewable biological resources and controlling nanoparticle characteristics through biomolecule interactions offer promising avenues for future research and applications. Conclusion This review delves into the scientific intricacies of plant-mediated synthesis of nanoparticles, highlighting the advantages of this approach over the traditional chemical synthesis methods. The study showcases the immense potential of green synthesis for medical and other applications, aiming to inspire further research in this exciting area and promote a more sustainable future.
... To validate sites-specific targeting of lesions and to significantly reduce ischemic stroke harm, many nanomedical medicines have been created [41] . For the therapy of ischemic stroke injury, a range of nanoparticles (NPs) have been reported, primarily polymeric micelles [42] , inorganic NPs (cerium oxide [43] , selenium [44] , gold NPs [45] , and carbon nanotubes [46] , and biodegradable nanoparticles like (PLGA [47] , albumin [48] , chitosan nanoparticles [49] as well as other nanoparticles such as liposomes [50] and nanogels [51] . Additionally, these nanoparticles can enhance the half-life and bioavailability of a variety of payloads, including small molecules, peptides, antibodies and nucleic acid [52,53] . ...
Article
Full-text available
The development of biomimetic drug delivery systems for biomedical applications has attracted significant research attention. As the use of cell membrane as a surface coating has shown to be a promising platform for several disease treatments. Cell-membrane-coated nanoparticles exhibit enhanced immuno-compatibility and prolonged circulation time. Use of various modified shapes have improved the bioavailability aspects of various drugs. Widely used in cancer treatment to enhance the retention time of drug at the affected site. The nano-bio interface represents the key facilitator of this communication exchange, where the surface properties of NPs govern their in vivo fate. Cell membrane-based biomimetic nanoparticles have emerged as one approach to achieve targeted drug delivery by actively engaging and communicating with the biological milieu.
... Biosensors Bioelectronics, 249-268. Zhang, S. X., Xue, S. F., Deng, J., Zhang, M., Shi, G., Zhou, T. (2016). Polyacrylic acidcoated cerium oxide nanoparticles: An oxidase mimic applied for colorimetric assay to organophosphorus pesticides. ...
Book
The research activities on nanozymes have witnessed boom over the past decade due to their high catalytic activity, low cost and high stability. The burgeoning field of nanozymes is still in its infancy as research is underway on various aspects of nanozymes such as their underlying mechanism of action, engineering efficient nanozymes, and exploring their wide applications. Nanozymes are being used in various spheres of science such as diagnostics, therapeutics, and electronics, however their applicability in different ways to monitor and remediate the environmental pollutants is naïve and bears immense future potential. In recent years, applications of nanozymes are being explored for environmental monitoring. They are being used to develop new methods for sensitive detection of environmental pollutants, removal/extraction of these pollutants from soil or water, and their degradation to relatively safer by products. However, their effective implementation in environmental field to support sustainable growth is yet challenging as it needs deeper understanding of the fundamental principles of their catalytic mechanism, and subsequently design new nanozymes with high selectivity and reusability
... Biosensors Bioelectronics, 249-268. Zhang, S. X., Xue, S. F., Deng, J., Zhang, M., Shi, G., Zhou, T. (2016). Polyacrylic acidcoated cerium oxide nanoparticles: An oxidase mimic applied for colorimetric assay to organophosphorus pesticides. ...
Book
The book is about the applications of Nanozymes in monitoring Environmental pollutants and the removal/extraction of these pollutants. Chapters address the various role of nanozymes in real-time monitoring and developing sensors for the same.
... Hence, it was suggested to use a particle size of less than 30 nm to increase the percentage of Ce3+ valence status [23,52]. In addition, Zhou et al. [53] elucidated that CeNPs has catalase-and superoxide dismutase-like activities. Co-treatment with nanoceria upregulated the molecular level of PGC1-α, a strong regulator of mitochondrial biogenesis, Nrf2 and its antioxidant response element Ho-1, thus increasing the intrinsic antioxidant defense mechanism by increasing the production of the antioxidant enzymes. ...
Article
Full-text available
Chemo fog is one of the most serious health concerns encountered by cancer survivors receiving doxorubicin (DOX)-based chemotherapy. Oxidative stress, neuroinflammation, apoptosis and impairment of synaptic plasticity are regarded as the key factors implicated in DOX-induced cognitive impairment. This research aimed to assess the possible neuroprotective effect of cerium oxide nanoparticles (CeNPs) against DOX-induced neurotoxicity. Forty-eight rats were divided into four groups (12 rats/group): control group, CeNPs group (received oral CeNPs solution (35 mg/kg) daily for 4 weeks), and DOX group (were administered DOX intraperitoneally (2 mg/kg, once/week for 4 weeks)) and DOX+ CeNPs group. The findings revealed that CeNPs mitigated behavioral alterations in DOX-induced cognitive deficit. Additionally, CeNPs alleviated the histopathological abnormalities in hippocampus and ameliorated DOX-induced neuroinflammation by downregulating the expression of NF-κB, TNF-α, IL-1β and IL6. In addition, CeNPs antagonized the apoptosis through reducing the protein expression of cytochrome c and caspase 3. In addition, it stimulated the antioxidant defense, as indicated by upregulating the expression of the Nrf2, HO-1 and PGC-1α genes. CeNPs improved synaptic plasticity via acting on the BDNF. These actions were related through the modification of SIRT-1 expression. Based on the aforementioned results, CeNPs antagonized the doxorubicin-induced neurodegeneration by its antioxidant, anti-inflammatory and antiapoptotic effects, alongside its SIRT-1 mediated mechanisms.
... Cependant peu d'études ont été réalisées dans des modèles d'ischémie cérébrale. Une revue publiée en 2016 discute du potentiel protecteur des NPC pour traiter l'ischémie cérébrale en citant des applications dont les pathologies de la rétine, les maladies neurodégénératives, les cardiomyopathies, le cancer, l'inflammation chronique et l'ischémie cérébrale(Zhou et al., 2016). ...
Thesis
Les accidents vasculaires cérébraux (AVC) constituent la 2ème cause de mortalité dans le monde et la 1ère chez les femmes en France. Pour les AVC ischémiques (AVCi), seules des stratégies de recanalisation pharmacologique ou mécanique ont été approuvées mais aucune stratégie protectrice n'est aujourd'hui disponible. Bien que le rôle délétère du stress oxydant ait été clairement établi dans les lésions neuronales et vasculaires à la suite d'une ischémie cérébrale (IC) dans les études précliniques, aucune stratégie anti-oxydante n'a démontré d'efficacité clinique à ce jour. Or, les nanoparticules d'oxyde de cérium (NPC) possèdent de multiples capacités antioxydantes (enzymatique et non enzymatique). Afin d'améliorer la biocompatibilité des NPC, la société Specific Polymers® a développé des copolymères de polyéthylène glycol (PEG)/ polyméthacrylate de méthyle/ phosphonate pour recouvrir leur surface. De plus, ces polymères peuvent être fonctionnalisés avec un peptide ciblant l'endothélium ce qui permettrait d'y concentrer l'effet antioxydant des NPC, afin de réduire la survenue d'hémorragies cérébrales, complications graves chez les patients victimes d'AVCi. L'objectif de cette thèse est d'évaluer l'impact du recouvrement des NPC sur leur potentiel thérapeutique dans l'IC. Les études ont été menées in vitro pour établir la toxicité, l'effet antioxydant et l'internalisation cellulaire des NPC et in vivo, pour examiner leur biodistribution et leur toxicité, ainsi que leur potentiel thérapeutique dans un modèle d'IC. Les études in vitro ont été effectuées sur des cellules endothéliales cérébrales murines de la lignée b.End3. Nous avons démontré que les NPC n'induisaient ni mortalité, ni perturbation de l'activité métabolique jusqu'à 100µg/ml. A 1000µg/mL, les NPC nues augmentent la mortalité, contrairement aux NPC PEGylées. Nous avons modélisé l'excitotoxicité survenant lors d'une IC et qui contribue au stress oxydant, grâce à un traitement des cellules par le glutamate. L'augmentation de la production d'espèces réactives de l'oxygène par les cellules b.End3 et l'oxydation des acides nucléiques dans ces conditions ont été réduites par les NPC, démontrant que leur recouvrement n'interfère pas avec leurs propriétés anti-oxydantes. La fonctionnalisation des NPC a permis le greffage d'un fluorophore pour suivre leur internalisation par cytométrie en flux et microscopie confocale. Nous avons ainsi mis en évidence que les NPC étaient rapidement internalisées dans les cellules b.End3. Des études de microscopie électronique ont ensuite montré que les NPC sont principalement localisées dans des endosomes périnucléaires. Enfin, nous avons réalisé le greffage sur les NPC d'un peptide ciblant une protéine d'adhésion vasculaire surexprimée lors de l'IC. La suite de ces études consistera à vérifier l'interaction spécifique de ces NPC avec la molécule d'adhésion. Les études in vivo ont permis d'établir la biodistribution des NPC chez des souris Swiss : des NPC sont retrouvées durant les premières heures suivant leur injection, avant leur élimination par voie rénale. L'histopathologie n'a révélé aucune toxicité des NPC sur le foie, les reins, la rate, les poumons et le cerveau de ces souris et aucune modification de leur numération sanguine n'a été observée. Les NPC ont ensuite été administrées dans un modèle murin d'IC, mais n'ont pas réduit le volume de la lésion dans nos conditions. En conclusion, le recouvrement des NPC par des polymères innovants a réduit leur toxicité sans altérer leurs capacités antioxydantes et leur internalisation dans des cellules endothéliales cérébrales. L'absence d'accumulation à long terme et de toxicité in vivo sont encourageantes quant à leur biocompatibilité. Bien que les NPC n'aient pas montré d'effet protecteur in vivo, celles ciblant l'endothélium pourraient réduire les lésions vasculaires et le risque hémorragique consécutif à une IC.
Article
The purpose of this work was to examine the physiochemical and biological aspects of plant-mediated cerium oxide nanomaterials (CeO2 NMs) using Zingiber officinale extract using stirring-based and autoclaved-based syntheses. X-ray diffraction analysis confirmed that the synthesized CeO2 NMs exhibited the cubic fluorite crystal structure with the particle diameters of 3.73 nm (stirred) and 3.81 nm (autoclaved). Fourier transform infrared analysis confirmed the attachment of various functional groups that are covalently bonded to the CeO2 NMs. Scanning electron microscopy images confirmed the spherical-agglomerated morphology of CeO2 NMs. The assessment of the CeO2 NM antioxidant properties using the radical scavenging assays showed a higher scavenging capacity of 34% and 43% for stirred-CeO2 NMs, respectively. Similarly, stirred-CeO2 NMs exhibited significant total antioxidant capacity (61 µgAAE/mg) and total reduction potential (52 µgAAE/mg) than autoclaved-CeO2 NMs. Likewise, stirred-CeO2 NM potential was determined to be higher for enzyme inhibitory activities (α-amylase, urease, and lipase) as compared to autoclaved-CeO2 NMs. The significant peroxidase-like activity was depicted by stirred-CeO2 NMs. Antibacterial activity of CeO2 NMs and the ginger extract was found to be mild (7–7.5 mm zone of inhibition) against tested bacterial strains except for S. entertica, against which ginger extract showed a moderate zone of inhibition (9 mm). The study concludes that ginger extract can synthesize CeO2 NMs following different synthetic modes. The presence of enough oxygen vacancies in stirred-CeO2 NMs facilitates the enhanced shifting between Ce3+ and Ce4+ by interaction with hydrogen peroxide (H2O2), implying that nanomaterials hold promise for further exploration in biomedical applications.
Article
Tendon disorders are common medical conditions, which can be greatly debilitating as they are often accompanied by great pain and inflammation. The techniques used nowadays for the treatment of chronic tendon injuries often involve surgery. However, one critical aspect of this procedure involves the scar tissue, characterized by mechanical properties that vary from healthy tissue, rendering the tendons inclined to reinjury or rupture. Synthetic polymers, such as thermoplastic polyurethane, are of special interest in the tissue engineering field as they allow the production of scaffolds with controlled elastic and mechanical properties, which could guarantee an effective support during the new tissue formation. The aim of this work was the design and the development of tubular nanofibrous scaffolds based on thermoplastic polyurethane and enriched with cerium oxide nanoparticles and chondroitin sulfate. The scaffolds were characterized by remarkable mechanical properties, especially when tubular aligned, reaching values comparable to the ones of the native tendons. A weight loss test was performed, suggesting a degradation in prolonged times. In particular, the scaffolds maintained their morphology and also remarkable mechanical properties after 12 weeks of degradation. The scaffolds promoted the cell adhesion and proliferation, in particular when in aligned conformation. Finally, the systems in vivo did not cause any inflammatory effect, representing interesting platforms for the regeneration of injured tendons.
Article
Full-text available
Cerebral ischemia occurs when blood flow to the brain is insufficient to meet metabolic demand. This can result from cerebral artery occlusion that interrupts blood flow, limits CNS supply of oxygen and glucose, and causes an infarction/ischemic stroke. Ischemia initiates a cascade of molecular events in neurons and cerebrovascular endothelial cells including energy depletion, dissipation of ion gradients, calcium overload, excitotoxicity, oxidative stress, and accumulation of ions and fluid. Blood-brain barrier (BBB) disruption is associated with cerebral ischemia and leads to vasogenic edema, a primary cause of stroke-associated mortality. To date, only a single drug has received US Food and Drug Administration (FDA) approval for acute ischemic stroke treatment, recombinant tissue plasminogen activator (rt-PA). While rt-PA therapy restores perfusion to ischemic brain, considerable tissue damage occurs when cerebral blood flow is reestablished. Therefore, there is a critical need for novel therapeutic approaches that can "rescue" salvageable brain tissue and/or protect BBB integrity during ischemic stroke. One class of drugs that may enable neural cell rescue following cerebral ischemia/reperfusion injury is the HMG-CoA reductase inhibitors (i.e., statins). Understanding potential CNS drug delivery pathways for statins is critical to their utility in ischemic stroke. Here, we review molecular pathways associated with cerebral ischemia and novel approaches for delivering drugs to treat ischemic disease. Specifically, we discuss utility of endogenous BBB drug uptake transporters such as organic anion transporting polypeptides and nanotechnology-based carriers for optimization of CNS drug delivery. Overall, this chapter highlights state-of-the-art technologies that may improve pharmacotherapy of cerebral ischemia.
Article
Full-text available
Administration of drugs and other therapeutic agents has been the central strategy of contemporary medicine for cardiovascular disease. The use of a drug delivery system (DDS) is always demanded to enhance the efficacy and safety of therapeutic agents, and improve the signal-to-noise ratio of imaging agents. Nano-scale materials modify in vivo drug kinetics, depending on (patho)physiological mechanisms such as vascular permeability and incorporation by the mononuclear phagocyte system, which constitute 'passive-targeting' properties of nano-DDS. By contrast, an 'active-targeting' strategy employs a specific targeting structure on nano-DDS, which binds to the target molecule that is specific for a certain disease process, such as tumor specific antigens and the induction of adhesion molecules. In this review, we summarize recent studies that applied nano-DDS for the diagnosis and treatment of cardiovascular disease, especially focusing on atherosclerosis and myocardial ischemia-reperfusion (IR) injury. Pathophysiological changes in atherosclerosis and myocardial IR injury are successfully targeted by nano-DDS and preclinical studies in animals showed positive effects of nano-DDS enhancing efficacy and reducing adverse effects. The development of nano-DDS in clinical medicine is keenly being awaited.
Article
Full-text available
With the development of many nanomedicines designed for tumor therapy, the diverse abilities of cerium oxide nanoparticles (CONPs) have encouraged researchers to pursue CONPs as a therapeutic agent to treat cancer. Research data have shown CONPs to be toxic to cancer cells, to inhibit invasion, and to sensitize cancer cells to radiation therapy and chemotherapy. CONPs also display minimal toxicity to normal tissues and provide protection from various forms of reactive oxygen species generation. Differential cytotoxicity is important for anticancer drugs to distinguish effectively between tumor cells and normal cells. The antioxidant capabilities of CONPs, which enable cancer therapy protection, have also resulted in the exploration of these particles as a potential anticancer treatment. Taken together, CONPs might be a potential nanomedicine for cancer therapy and this review highlights the current research into CONPs as a novel therapeutic for the treatment of cancer.
Article
Purpose: To critically review and evaluate the science behind individual eligibility criteria (indication/inclusion and contraindications/exclusion criteria) for intravenous recombinant tissue-type plasminogen activator (alteplase) treatment in acute ischemic stroke. This will allow us to better inform stroke providers of quantitative and qualitative risks associated with alteplase administration under selected commonly and uncommonly encountered clinical circumstances and to identify future research priorities concerning these eligibility criteria, which could potentially expand the safe and judicious use of alteplase and improve outcomes after stroke. Methods: Writing group members were nominated by the committee chair on the basis of their previous work in relevant topic areas and were approved by the American Heart Association Stroke Council's Scientific Statement Oversight Committee and the American Heart Association's Manuscript Oversight Committee. The writers used systematic literature reviews, references to published clinical and epidemiology studies, morbidity and mortality reports, clinical and public health guidelines, authoritative statements, personal files, and expert opinion to summarize existing evidence and to indicate gaps in current knowledge and, when appropriate, formulated recommendations using standard American Heart Association criteria. All members of the writing group had the opportunity to comment on and approved the final version of this document. The document underwent extensive American Heart Association internal peer review, Stroke Council Leadership review, and Scientific Statements Oversight Committee review before consideration and approval by the American Heart Association Science Advisory and Coordinating Committee. Results: After a review of the current literature, it was clearly evident that the levels of evidence supporting individual exclusion criteria for intravenous alteplase vary widely. Several exclusionary criteria have already undergone extensive scientific study such as the clear benefit of alteplase treatment in elderly stroke patients, those with severe stroke, those with diabetes mellitus and hyperglycemia, and those with minor early ischemic changes evident on computed tomography. Some exclusions such as recent intracranial surgery are likely based on common sense and sound judgment and are unlikely to ever be subjected to a randomized, clinical trial to evaluate safety. Most other contraindications or warnings range somewhere in between. However, the differential impact of each exclusion criterion varies not only with the evidence base behind it but also with the frequency of the exclusion within the stroke population, the probability of coexistence of multiple exclusion factors in a single patient, and the variation in practice among treating clinicians.
Article
Integrins such as αvβ3, α5β1 play a key role in angiogenesis regulation, invasion and metastasis, inflammation, wound healing, etc. The up-regulation of integrin αvβ3 after cerebral ischemic stroke can promote angiogenesis, which in turn improves functional recovery. In addition, the integrin αvβ3 inhibitor can block the blood-brain barrier (BBB) leakage induced by vascular endothelial growth factor (VEGF) and also can reduce inflammatory reaction, decrease the deposition of fibrinogen. Other studies showed that integrin αvβ3 is not essential in revascularization. Therefore, the effect of integrin αvβ3 in the whole process of brain function recovery merits further study.
Article
Cerium oxide nanoparticles, nanoceria, have been proposed as drug delivery devices and therapeutic tools also for neurological diseases due to their antioxidant properties and neuroprotective effects. However, very little evidence has been hitherto obtained on the passage of nanoceria across the blood-brain barrier in vivo. Distribution of in-house synthesized 10 nm nanoceria in spleen, liver, lung, kidney and brain was here examined at 24 h after a single iv injection of a nanoceria suspension at two concentrations (1 or 20 mg/kg). The use of fluorescein isothiocyanate-conjugated nanoceria allowed their visualization in confocal microscopy. The investigation was also pursued with inductively coupled plasma-mass spectrometry and with transmission and scanning electron microscopy. Consistently with previous investigations, these approaches pointed to accumulation of the largest amount of nanoceria in the liver and spleen, with deposition also in the kidney and lung, and no signs of overt pathology of these tissues. In the brain, confocal microscopy revealed penetration of fluorescent nanoceria into the parenchyma, albeit in limited amount, and astrocytic activation. Occurrence of electron-dense nanoceria-like particles in the cerebral cortex, sparse or in agglomerates, was confirmed by the ultrastructural study. The present findings contribute novel knowledge by showing nanoceria entry into the brain parenchyma.
Article
Cerium oxide nanoparticles are potent antioxidants, based on their ability to either donate or receive electrons as they alternate between the +3 and +4 valence states. The dual oxidation state of ceria has made it an ideal catalyst in industrial applications, and more recently, nanoceria's efficacy in neutralizing biologically generated free radicals has been explored in biological applications. Here we report the in vivo characteristics of custom-synthesized cerium oxide nanoparticles (CeNPs) in an animal model of immunological and free-radical mediated oxidative injury leading to neurodegenerative disease. The CeNPs are 2.9 nm in diameter, monodispersed and have a -23.5 mV zeta potential when stabilized with citrate/EDTA. This stabilizer coating resists being 'washed' off in physiological salt solutions, and the CeNPs remain monodispersed for long durations in high ionic strength saline. The plasma half-life of the CeNPs is ~4.0 hours, far longer than previously described, stabilized ceria nanoparticles. When administered intravenously to mice, the CeNPs were well tolerated and taken up by the liver and spleen much less than previous nanoceria formulations. The CeNPs were also able to penetrate the brain, reduce reactive oxygen species levels and alleviate clinical symptoms and motor deficits in mice with a murine model of multiple sclerosis. Thus, CeNPs may be useful in mitigating tissue damage arising from free radical accumulation in biological systems.
Article
We report the synthesis of multifunctional, hierarchical, cerium-containing HZSM-5 catalysts with various aluminum and cerium contents using a dry-gel, dual templating method. The catalysts were characterized by X-ray diffraction, N2 physisorption, scanning electron microscopy, diffuse reflectance UV–Visible spectroscopy, diffuse reflectance Fourier transform infrared spectroscopy, Fourier transform Raman spectroscopy, NH3 temperature programmed desorption and X-ray photoelectron spectroscopy. At low theoretical Si:Al ratios (15 and 20), the formation of amorphous silica–alumina materials was observed. However, at higher theoretical Si:Al ratios (30, 40, and 50), crystalline materials were formed. Furthermore, with Si:Al ratios of 50, cerium incorporation as high as 2.2 wt% was obtained using this procedure. To understand the role of aluminum and cerium in the catalyst, the crystalline materials were studied in the catalytic fast pyrolysis of glucose at 600 °C. The catalysts with cerium incorporated showed an increase in the production of oxygenated chemicals as well as CO, compared with their non-cerium containing analogs.