Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

Coordination Chemistry Reviews 448 (2021) 214170

Contents lists available at ScienceDirect

Coordination Chemistry Reviews


journal homepage: www.elsevier.com/locate/ccr

Review

Nanozymes: Activity origin, catalytic mechanism, and biological


application
Wenping Yang a, Xin Yang b, Longjiao Zhu a, Huashuo Chu a, Xiangyang Li b, Wentao Xu a,⇑
a
Department of Nutrition and Health, and College of Food Science and Nutrition Engineering, China Agricultural University, Beijing 100191, China
b
Beijing Laboratory of Food Quality and Safety, Faculty of Food Science and Engineering, Beijing University of Agriculture, Beijing 102206, China

a r t i c l e i n f o a b s t r a c t

Article history: Nanozymes are nanomaterials with excellent enzyme-like characteristics. Compared with natural
Received 16 April 2021 enzymes, nanozymes possess significant advantages, such as low cost, high stability, good durability, con-
Accepted 15 August 2021 trollable activity, easy storage, and excellent reusability, thus they have been widely applied in industrial,
medical, and biological fields. In the past decade, nanozymes have made impressive progress in their
activity exploration, mechanism study, and advanced applications. In this review, we analyze the source
Keywords: of nanozyme catalytic activities from two aspects, including the structure comparison of natural enzymes
Nanozyme
and nanozymes and the nano effects of nanomaterials. We also systematically summarize the catalytic
Activity origin
Catalytic mechanism
mechanism of nanozymes, and discuss the reason for their multiple enzyme-like activities. In addition,
RONS regulation RONS regulation-based biological applications are comprehensively reviewed. Finally, we present the
Biological application current challenges of nanozymes and their future research directions.
Ó 2021 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2. Activity origin of nanozymes. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
3. Enzyme-like properties of nanozymes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
3.1. Peroxidase activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
3.2. Oxidative activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
3.3. Catalase activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
3.4. Superoxide dismutase activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
3.5. Hydrolase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
3.6. Diversity of nanozyme activity. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
4. Biological applications of nanozymes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
4.1. Anti-inflammatory and cytoprotection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
4.2. Neurological disease therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
4.3. Cancer therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
4.4. Sterilization and disinfection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
5. Conclusions and prospects. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
Declaration of Competing Interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14

⇑ Corresponding author at: China Agricultural University, Tianxiu Road 10, HaiDianDistrict, Beijing China.
E-mail address: xuwentao@cau.edu.cn (W. Xu).

https://doi.org/10.1016/j.ccr.2021.214170
0010-8545/Ó 2021 Elsevier B.V. All rights reserved.
W. Yang, X. Yang, L. Zhu et al. Coordination Chemistry Reviews 448 (2021) 214170

1. Introduction terns. Nanozymes can also modulate the reactive oxygen and
nitrogen species (RONS) level in cells. For example, cerium nano-
Nanomaterials typically refer to materials with size (at least in zymes with RONS-scavenging capability can disproportionate
one dimension) of 1–100 nm. At this scale, materials may exhibit superoxide anions to H2O2 via SOD mimicking activity, and convert
unique chemical, physical, biological, and mechanical properties H2O2 to H2O and O2 via catalase mimicking activity, thereby
that are distinct from bulk ones.[1] The morphology of nanomate- achieving anti-immunity, cell protection, and disease prevention
rials includes solid, hollow, or porous nanospheres, nanorods, or treatment. Furthermore, some nanozymes offer excellent
nanosheets, nanofibers, nano-polyhedrons, nanoflowers and so RONS-generating ability because of their peroxidase activity and/
on. In 1990 s, scientists found some nanomaterials showed or oxidase activity to produce reactive oxygen species (ROS), which
enzyme-like activity, and Scrimin and co-workers coined the term are successfully applied in cancer treatment and sterilization
‘‘nanozyme’’ in 2004. In 2007, Yan and co-workers verified the (Scheme 1). However, earlier reports on the catalytic mechanism
inherent peroxidase-like activity of Fe3O4 nanoparticles.[2] Since of nanozymes mainly focused on ROS, but rarely focused on the
then, nanozymes have received rising attentions in various fields, reactive nitrogen species (RNS).
and a large number of nanomaterials with enzyme-like character- Compared with natural enzymes, nanozymes have considerable
istics have been proposed. Nanozymes are nanomaterials with advantageous characteristics. First, nanozymes are higher in cat-
enzyme-mimicking characteristics [3], including metals (Au, Ag, alytic stability, lower in cost, easy in recovery and storage. Second,
Pt, Pd, Ir, Au@Pt), metal compounds (Fe3O4, CeO2, MnO2, CuS, nanozymes are more tolerant to surrounding environment, and can
MnSe), non-metals (SWNTs, C-dots, fullerene), non-metal com- work in a wider range of pH and temperature. Third, nanozymes
pounds (g-C3N4, GO). Nanozymes show the behaviors similar to with large surface area are conducive to surface modification. Four,
natural enzymes, such as catalyzing oxidation of substrates, fol- nanozymes have some excellent properties, such as good stability,
lowing the same kinetics, pH- and concentration-dependent pat- tunable activity, specific environmental responsiveness, and adjus-

Scheme 1. An illustration of the application of nanozymes in the biological field.

2
W. Yang, X. Yang, L. Zhu et al. Coordination Chemistry Reviews 448 (2021) 214170

table pore size. Additionally, they can be used as a novel platform like properties.[5] Because the size is reduced to nanometer level,
to achieve internalization, circulation, penetration, absorption, and nanomaterials exhibit remarkable physicochemical properties that
targeting in tissues or cells for certain disease treatment. Five, are distinctly different from bulk inert materials. Due to many sus-
some nanozymes possess unique physicochemical properties, such pended bonds and undercoordination, the atoms on nanomaterial
as magnetism, photothermal qualities, and photodynamic effect, surface have extra energy and become unstable.[6] These high-
which can make their behavior controlled remotely via magnetic energy surface atoms give nanozymes specific activity to react
field, laser, ultrasound, and even heat. with other substances (Fig. 1c, d, e). Particularly, single atom nano-
In the past decade, with the rapid development of nanotechnol- zymes with enzyme-mimicking structure exhibit superior catalytic
ogy and in-depth research, nanozymes have made impressive pro- performance. In addition, nanozymes can catalyze bioorthogonal
gress. In addition to the wide range of biological applications, the reactions not addressed by a natural enzyme, which may be due
basic research has also made significant progress. Therefore, it is to their multivalent elements, multiple active sites, or abundant
necessary to review the theory and application of nanozymes. In coordination structures.[7] Surprisingly, grafting chiral amino
this review, we firstly elaborate the origin of nanozyme activity acids onto nanoparticle surfaces can indirectly endow them with
from the perspectives of enzyme structure and nano effects (vol- a stereoselectivity, in which the functionality of amino acids may
ume effect, quantum size effect, and surface effect), revealing the be similar to that of proteins in natural enzymes.[8,9] Yang and
cause that nanozymes possess enzyme-like properties and func- co-workers also obtained chiral carbon quantum dots with enan-
tions. Secondly, we systematically summarize and compare the tioselectivity using chiral amino acids as raw materials.[10]
catalytic mechanisms, and discuss the reasons for their multiple From the aspects of nano effects (volume effect, quantum size
enzyme-like activities. Then, this paper comprehensively reviews effect, and surface effect). First, in terms of size, nanomaterials
the RONS regulation-based biological applications, including anti- belong to the intermediate domain between atoms and ordinary
inflammation and cytoprotection, neurological disease therapy, particles. Their properties and characteristics are significantly dif-
cancer therapy, sterilization and disinfection. Finally, we discuss ferent from those highly active atoms and ordinary particles. When
the current challenges and future development of nanozymes. the size is equivalent to or smaller than some physical characteris-
tics (such as the wavelength of light, the de Broglie wavelength of
conducting electrons, and the coherence length or projection depth
2. Activity origin of nanozymes of superconducting state), the periodic boundary conditions of
crystal are destroyed and the atom density near the surface of
Nanozymes are nanomaterials in essence, but they possess the amorphous nanoparticles decrease.[13,14] Consequently, their
properties of both enzymes and surface catalyst. It is a cross pro- physical and chemical properties (e.g., light, sound, electricity,
duct of many fields, including biological enzymes, nanomaterials magnetism, chemical activity, and catalysis) changes substantially.
and surface catalyst. To a certain extent, nanozymes surpass them Second, bulk materials possess continuous energy bands. When
with new properties and functions, but do not completely beyond particle size reduces to nanometer level, the electronic energy
them in all aspects. In this section, we revealed the activity origin levels near the Fermi level change from quasi-continuous to dis-
of nanozymes from the aspects of structure and nano effects. In crete energy levels. When the energy level spacing exceeds ther-
addition, the reasons that nanozyme activity can be regulated by mal energy, magnetic energy, electrostatic energy, magnetostatic
their size, morphology, and external stimulation are discussed. energy, photon energy, or superconducting condensed energy,
Nanozymes possess enzyme-mimicking characteristics and the quantum effect appears and alters the properties of nanomate-
functions. From the perspectives of enzymes (Table 1), enzymes rials (light, sound, magnetism, heat, electricity, and superconduc-
are majorly proteins produced by living cells with an excellent cat- tivity). The specific heat, magnetic susceptibility, and catalytic
alytic performance.[4] They often consist of amino acids, saccha- properties of nanoparticles are related to the parity of the con-
rides, metal atoms, and other cofactors.[5] Enzymes generally tained electrons. [13,14] In a word, the nanoscale size endows
have a specific active site, for example, heme molecules coordi- nanomaterials with novel chemical activity and catalysis.
nated in the protein scaffold serve as the active site in many In general, nanozyme activity depends on their size, morphol-
peroxide-related enzymes. The center of heme is occupied by ogy, and external stimulation (e.g., light, sound, and heat). [2,15–
metal atoms like Fe, Cu, Mn, or Zn, which are critical for catalysis 18] In terms of size, the smaller the particle size is, the higher their
(Fig. 1a, b, c). In addition, they possess a substrate binding site activity will be.[2] It can be explained as follows: First, increase of
formed by amino acids with specific structure, which provide the specific surface area. With a decrease in the size of nanoparticles,
stereoscopic space for cofactor and metal atom coordination and the specific surface area increases significantly. For example, when
substrate binding. This complex but precise structure endows the particle size is 10, 5, and 2 nm, their specific surface area is 90,
enzymes with excellent catalytic activity and selectivity.[5] Nano- 180, and 450 m2 g1, respectively. Such a high specific surface area
zymes are a class of nanomaterials that possess intrinsic enzyme- changes surface atomic number and surface energy, which greatly

Table 1
Comparison of structure and properties between natural enzymes and nanozymes.

Natural enzymes Nanozymes


Essence Majorly proteins Nanomaterials
Composition Amino acids, saccharides, metal atoms/ion, and cofactors. Metal or nonmetal atoms/ion.
Structure (a) With active site. E.g., metal atoms in heme’s (a) With active site. E.g., coordination unsaturated surface atoms.
characteristics center. (b) Without specific substrate binding structure, except for chiral
(b) Substrate binding site with specific structure. nanozymes.
Number of active sites Limited Multiple
Utilization of active High Low, except for single-atom nanozymes.
sites
Catalytic characteristics High catalytic activities and substrate specificity. High catalytic activity, low substrate specificity.

3
W. Yang, X. Yang, L. Zhu et al. Coordination Chemistry Reviews 448 (2021) 214170

Fig. 1. Structural comparison of natural enzymes and nanozymes. (a) The three-dimensional X-ray crystal structure of horseradish peroxidase isoenzyme C (the blue
spheres are Ca atoms). [11] (b) The heme iron atom (red sphere) in the heme binding region of horseradish peroxidase isoenzyme C.[11] (c) The active sites of natural
enzymes, nanozymes, and Fe SAEs.[12] (d) Materials and nanomaterials (the illustration is a cross section, red and yellow spheres represent highly active and active atoms,
respectively). (e) The catalytic structure in carbon nanozymes.[5]

affect the catalytic activities of nanoparticles. Second, increase of tion of atomic movement, the saturated atoms closer to the surface
surface atomic number. With the decrease of particle size, the frac- break away from the original bondage to become active atoms. All
tion of surface atoms increases markedly, along with changes in of the above factors may affect the active sites of nanozymes,
the ratio of surface atoms to total atoms. For example, when the resulting in changes of catalytic activity. Active sites generally refer
particle diameter is 10, 2, and 1 nm, the surface atoms account to surface active atoms or sites, which possess high energy because
for 20%, 80%, and 99%, respectively. [14] The rapid increase of sur- of coordination unsaturation and can easily combine with other
face atom ratio changes the properties of nanoparticles. Third, raise atoms, groups, or substances. The common active sites include
of surface energy. The crystal field environment and the binding coordination unsaturated surface atoms and surface defects (e.g.,
energy of surface atoms are different from the internal one. With steps, kinks, adatoms, and vacancies).
a decrease in size, the specific surface area and the surface atomic
number increases, leading to many dangling bonds and unsatu-
3. Enzyme-like properties of nanozymes
rated bonds on surface atoms. [6,19] These surface atoms with high
surface energy are extremely unstable and easily react with other
Until now, many nanomaterials have been discovered with
atoms, groups, or substances, showing significant chemical and
remarkable enzyme-like activities. The types of enzymatic activity
catalytic activity in nanozymes.
mainly include oxidoreductase (e.g., oxidase (OXD), peroxidase
Nanozyme activity is morphology-dependent. Because atom
(POD), catalase (CAT), superoxide dismutase (SOD), and nitrate
coordination environment and dangling bonds are closely related
reductase) and hydrolase (e.g., nuclease, esterase, phosphatase,
to crystal planes. The more saturated the coordination is, the lower
protease, and silicatein),[22] but most of the catalytic mechanisms
the catalytic activity will be, and vice versa. For instance, the coor-
has not been thoroughly studied. Therefore, this section summa-
dination saturation order: (0 0 1) facet > (0 1 0) facets > (1 0 0) facets
rizes the common oxidoreductase activities (Table 2) and partial
> (0 1 0) facets. It has been shown that the glutathione peroxidase-
hydrolase from reaction conditions, catalytic kinetics, and catalytic
like activity of V2O5 nanozymes: only (0 0 1) facet bound
mechanisms. Furthermore, the reasons for their multiple enzyme-
nanowires < large (0 0 1) and minor (0 1 0) facet bound
like activities are discussed from mechanism.
nanosheets < major (0 1 0) and minor (0 0 1) facet bound
nanoflowers < two major (1 0 0), (0 1 0) facet bound nanospheres.
[7,16,20] In addition, certain external factors (e.g., light, sound, 3.1. Peroxidase activity
and heat) may stimulate nanozyme activity due to the activation
of electrons after gaining energy.[21] Taking nanoparticles as an Peroxidase-like activity is the earliest confirmed activity and
example, when given a certain amount of energy, on the one hand, has been discovered in various nanomaterials covering metal,
the coordinated unsaturated atoms on nanoparticle surface will be metal oxide, and carbon nanozymes. Similar with natural peroxi-
excited into more active atoms. On the other hand, with accelera- dase, nanozymes catalyze substrate oxidation with pH-,
4
W. Yang, X. Yang, L. Zhu et al. Coordination Chemistry Reviews 448 (2021) 214170

Table 2 stances in organisms are considered as substrates of peroxidases,


Comparison of typical catalytic mechanism in nanozyme activity. such as nucleic acids, proteins, polysaccharides, and lipids. There-
Activities Mechanisms Specific Functions Reference fore, substrate oxidation by peroxidase occurs frequently.
POD ROS Catalyzing the production of ROS by [2,23–30] Just as horseradish peroxidase, the peroxidase-like activity of
generation certain reactions (e.g., Fenton, Haber- nanozymes follows the typical Michaelis-Menten kinetics and
Weiss reactions), which can come ping-pong mechanism.[2] It binds and reacts with the first sub-
from both substrates and strate, subsequently releasing the first product before reacting
nanozymes.
Electron Enriching and transferring electron [31–34]
with the second substrate.[31] In general, nanozyme peroxidase-
transfer without producing OH or even like activity has a higher Km for H2O2 and a lower Km for another
inhibiting the production of OH. substrate (TMB) than natural enzymes, indicating that nanozymes
OXD Reactive Producing free radicals or ROS by [35–40] show a higher affinity towards TMB but a lower affinity towards
species activating substrates or dissolved
H2O2.[2] Most of the catalytic efficiency is higher than that of nat-
generation oxygen.
Electron Enriching and transferring electron, [41,42] ural enzymes.[51] The reason may be that nanozymes have a larger
transfer or transferring electron by redox specific surface area and more active sites. However, by contrast,
reaction. the specificity of nanozymes is unsatisfactory due to the lack of
CAT Adsorption Promoting the decomposition of [43] substrate binding sites. Interestingly, the profuse functional groups
activation H2O2 by the high adsorption energy.
in carbon nanozymes provide unexpected functions. The ketonic
Redox Decomposing H2O2 by redox [34,44,45]
reaction reaction. carbonyl, carboxylic, and hydroxyl groups are suggested as the cat-
SOD Adsorption Promoting the disproportionation by [46,47] alytic active centers, substrate binding sites and inhibitors, respec-
activation adsorption. tively.[52] Some researchers have recently improved the substrate
Electron Realizing the disproportionation [45,48,49]
selectivity via molecular imprinting technique or assembly of
transfer reaction by free radical reaction or
redox reaction. amino acid residues on nanozymes. [9,10,53]
In general, the catalytic pathway of peroxidase mimics mainly
consists of ROS generation and the electron transfer process. For
temperature-, and concentration-dependence. They generally have the generation of ROS, nanozymes react with H2O2 to produce
two substrates in the reaction, one is peroxide (mostly H2O2), and ROS (e.g., OH, O2, HO2), which are responsible for further oxidiz-
the other is oxidized substrate.[5] For certain nanozymes, some ing substrates (Fig. 2a). Fe3O4 nanoparticles are the first proposed
special substances (such as halide, glutathione, thiol, etc.) may be nanozymes with intrinsic peroxidase-like activity, in which the
necessary to complete the entire catalytic reaction.[50] Many sub- surface ferrous ions play a dominant role in activation of H2O2

Fig. 2. The catalytic mechanisms of nanozymes. (a) ROS generation mechanism of the POD-like activity of Cu-CDs-Fe2+.[58] (b) The proposed electron transfer mechanism
for the POD-like activity of Ti3C2 nanosheets.[59] (c) ROS generation mechanism of TiO2/C-QDs as oxidase mimics.[39] (d) The catalytic mechanism of PB with CAT-like
activity.[34] (e) The energy-based model for the activation of 3O2 by metal surfaces.[46] (f) A schematic illustration of chiral CdTe-based specific DNA cleavage under CPL
irradiation.[9]

5
W. Yang, X. Yang, L. Zhu et al. Coordination Chemistry Reviews 448 (2021) 214170

and generation of ROS via Fenton or Haber-Weiss reactions.[2,23– OH group bonding to the cluster. After CH activation, the barrier-
27] In MOFs, the Fenton-like mechanism are proposed when H2O2 less recombination of CH3 radicals with Cu-bound OH group in
is absorbed and O-O bond breaks down to produce OH radicals on the cluster forms methanol molecules that preferentially coordi-
nanomaterial surface.[28,29] In addition, although it is also a free nates to two neighboring Cu centers of the trinuclear species.[35]
radical mechanism, nanocarbon oxides undergo carboxyl-group- Some reports identified that free radicals came from activated dis-
assisted decomposition of H2O2. The carboxyl groups of nanocar- solved oxygen. Nanozymes with large specific surface areas could
bon oxides are oxidized into peroxycarboxyl groups and homolyt- adsorb and activate massive dissolved oxygen to produce ROS
ically cleaved, generating OH and oxidizing TMB into oxTMB.[30] intermediates (e.g., O2, 1O2, OH), which were responsible for
For the electron transfer process, the substrates are first substrate oxidation (Fig. 2c). [36–39] Huang and coworkers indi-
absorbed on nanozyme surface donating electrons. The large speci- cated that O2 was produced first, then subsequently converted
fic surface area with abundant active sites or variable oxidation into H2O2 and 1O2 immediately via the oxygen radical chain reac-
states endows nanozymes with an increased electron density and tion.[40]
excellent mobility, thereby resulting in the redox reaction between
substrates and H2O2. The rapid electron transfer from substrates to 3.3. Catalase activity
H2O2 further accelerates substrate oxidation and H2O2 reduction.
The characteristic of this process is that it does not produce or even H2O2 is an essential ROS in nature. Nanozymes with catalase-
eliminate OH (Fig. 2b). Recently, Qu and co-workers discovered like activity can catalyze H2O2 to generate water and oxygen
that electron transfer occurred from the top of graphene valence molecular. It depends on the concentration of both substrates
band to the lowest unoccupied molecular orbital (LUMO) of and nanozymes, and its decomposition rates become higher with
H2O2. In this progress, TMB or other substrate molecules absorbed an increased concentration of them.[60] This process can prevent
on graphene surface and donated the lone-pair electrons in their the accumulation of ROS, effectively protecting cells from oxidative
amino groups to graphene, resulting in an increased electron den- stress. The catalase-mimicking activity is generally evaluated by
sity and mobility on graphene.[31] Another study also demon- the level of O2 or dissolved oxygen during the reaction process.
strated that the peroxidase-like activity of Co3O4 NPs was The activity of nanozymes is highly pH- and temperature-
attributed to the electron transfer between reducing substrates dependent. Most prefers neutral and alkaline environments, but a
and H2O2, rather than the production of free radicals.[32,33] Fur- few nanozymes also exhibit catalase-like activity in slightly acidic
thermore, the peroxidase-mimicking activity of Prussian blue environment. [32,61] Similar to catalase, it also follows a typical
nanoparticles could inhibit OH formation, which may be due to Michaelis-Menten catalytic process, and nanozymes have a high
their abundant redox potential as an efficient electron trans- affinity for H2O2.[62]
porter.[34] The catalytic reaction by catalase-like activity is closely associ-
ated with nanoparticle morphology, structure, and surface valence,
3.2. Oxidative activity as well as environmental pH. Currently, the understanding of their
catalytic mechanism of nanozymes is exceedingly limited. Usually,
Oxidases can catalyze the oxidation of substrates into oxidized the catalytic mechanisms include adsorption activation and redox
products and H2O/ H2O2/ O2 with the assistance of molecular reactions. With regard to metal-based catalase-mimicking nano-
oxygen (or other oxidizing reagents).[20] Similarly, nanomaterials zymes, the catalytic mechanism was closely associated with free
(e.g., Ru, Au@Pt, CeO2, N-CNMs) have been discovered with the radical chains. The preabsorbed OH* (* means species adsorbed
ability of oxidizing substrates, and their catalytic activity strongly on metal surfaces) performed a key role on nanozyme surfaces,
depends on temperature, pH and substrates. The nanozyme activ- and could prompt H2O2 to prefer acid-like decomposition, then
ity also shows typical Michaelis-Menten behavior. [54–57] Most the decomposed H reacted with pre-adsorbed OH* to yield HO2*
studies have shown that their Km value are lower than that of nat- and H2O*. Subsequently, the generated HO2* passed H to another
ural enzymes, indicating that the oxidase-like activity displays a H2O2*, leaving an O2* and converting the H2O2* to H2O* and OH*.
higher affinity toward substrates. In other words, only a low con- [43] In addition to free radical reaction, the decomposition process
centration of substrates is needed to achieve high catalytic effi- of H2O2 was accompanied by a redox reaction due to the presence
ciency of nanozymes. of redox forms in Co3O4 nanozyme. At a high pH, H2O2 reacted with
The reaction pathway of nanozyme oxidase-like activity OH– to form OOH, which was more nucleophilic than H2O2 and
includes electron transfer and reactive species generation. In elec- could easily interact with Co (III) to generate O2H and Co (II).
tron transfer process, nanozymes act as the recipient and transfer The produced O2H would further react with OH that generated
of electrons. For the oxidase-mimicking activity of naked gold from H2O2 activation by Co (II). A high pH would facilitate the
nanoparticles (AuNPs), it was considered that the hydrated glucose two reactions, accelerating decomposition of H2O2 by Co3O4.[44]
anion was first adsorbed onto the gold surface to form electron- Similarly, since multiple redox forms and low redox potential of
rich gold species, subsequently activating dissolved oxygen by a H2O2/O2 at a high pH, Prussian blue also exhibited an inherent
nucleophilic attack and generating a dioxo gold intermediate to catalase-like activity (Fig. 2d).[34] Surprisingly, CeO2 as catalase
transform electrons from glucose to dioxygen.[41] Research mimics showed a redox-state-dependent manner. The H2O2 mole-
showed that MoO3 exhibited sulfite oxidase activity. The catalytic cules bound to Ce3+ site and their O–O bond split to release water
mechanism was speculated that SO2- 3 was bonded firstly and oxi- molecules with the electrons transfer from H2O2 to two Ce3+, then
dized to SO2-4 on MoO3 nanoparticles with variable oxidation states, Ce4+ backed to Ce3+ via oxidation reaction.[45] The high valence
along with a reduction of Mo (VI) to Mo (IV). Then, Mo (IV) was oxi- state on nanozyme surfaces is beneficial to their catalase activity.
dized back to Mo (VI) via two one-electron reduction reactions.[42] [61,63]
For reactive species generation, free radicals originate from
either oxygen molecules or substrates. For the mimicking methane 3.4. Superoxide dismutase activity
monooxygenase of trinuclear complex [Cu3(m-O)3]2+, free radicals
are generated from substrates. In this process, methane is activated Superoxide anions, including HOO and O2 (O2 is the main
first via the interaction of the respective single-occupied molecular form), are destructive ROS widely distributed in cells. They can ini-
orbital of [Cu3(m-O)3]2+ with the antibonding CH orbital (s*(CH)) of tiate a radical-chain reaction producing more toxic free radicals. In
methane, then the C–H bond is cleaved, resulting in a CH3 and an general, superoxide anions can self-decay in natural conditions,
6
W. Yang, X. Yang, L. Zhu et al. Coordination Chemistry Reviews 448 (2021) 214170

but the decay rate is exceedingly slow. Hence, superoxide dismu- DMP.[64] Kuang and co-workers [9] also discovered that chiral
tase is an important antioxidant that responsible for the dispropor- cysteine-modified CdTe nanoparticles could mimic restriction
tionation of superoxide anions into H2O2 and O2. Benefiting from endonuclease to specifically recognize and cut at the restriction site
the scavenging capacity of O2, nanozymes (e.g., Pd, MnO2, PB, GAT0 ATC in double-stranded DNA exceeding 90 base pairs (Fig. 2f).
fullerene) are considered as promising alternatives to superoxide During this process, nanoparticles acted as electron donors to
dismutase. When nanozymes possess multiple enzyme activities, induce ROS generation, while DNA was an electron acceptor to
the environmental pH or the structure and surface ions of nanoma- cleave phosphodiester bond. In their another study,[65] chiral cop-
terials make superoxide dismutase activity become the dominant per sulfide quantum dots (d/l-QDs) were used as photocatalysts to
activity. Inappropriate pH leads to a thicker electrostatic barrier cleave proteins. The OH produced by d/l-QDs caused the cleavage
to superoxide radicals, suppressing the catalytic activity.[47] Sim- of peptide bond between amino acids K and L, resulting in the cleav-
ilar with natural enzymes, the kinetics follow a ping-pong age of proteins at specific sites (61 KD + 5KD).
mechanism.
Due to the unpaired electrons and multivalent ions on surface, 3.6. Diversity of nanozyme activity
the primarily contributions of nanozymes are adsorption activa-
tion and electron transfer. However, the actual mechanisms are The most significant feature of nanozymes is their multiple
determined by pH, nanozyme structures, or surface ions. For the enzyme-like activities. Although different nanomaterials have the
superoxide dismutase-like activities of metal and alloy, the mech- same catalytic activities, their catalytic mechanisms are com-
anisms mainly include the protonation of O2 and the adsorption pletely depending on environmental pH, or size, surface ions states,
and rearrangement of HO2 on metal surfaces. O2 is a Brønsted and morphology of nanomaterials.
base that easily captures a proton from water to form HO2 and Nanozymes possess multiple enzyme-mimicking characteris-
OH, and this process is accelerated due to the strong adsorption tics and catalytic mechanisms at different pH. (1) For noble metal
of HO2 on nanozyme surface. Consequently, the rearrangement nanozymes (e.g., Au, Ag, Pt, and Pd), the peroxidase activity is pre-
of HO2* occurs with the formation of H2O2* and O2* (Fig. 2e). sented at pH 4.5 to catalyze H2O2 into OH radicals, while the
[46] Reports showed that C60[C(COOH)2]3 nanozymes (C3) could catalase-mimicking activity is presented at pH 7.4 to decompose
remove superoxide radicals via the attraction of electron- H2O2 into O2. Through density functional theory calculation, Gao
deficient regions. C3 electrostatically drove superoxide anions and co-workers [43] contended that pH-dependent behavior was
toward electron-deficient areas on its surface, then O2 was stabi- mainly attributed to two different decomposition pathways of
lized via the hydrogen bonding and protons of carboxyl groups (or H2O2 caused by preabsorbed H+ or OH on nanozyme surface
intercalated solvating H2O) until the second O2 arriving to com- (Fig. 3a). Under acidic conditions, H* was pre-adsorbed on nano-
bine with the original O2. It allowed O2 dismutation with the zyme surface, which had little influence for the adsorption and
assistance of protons from carboxyl groups or local water mole- decomposition of H2O2. H2O2 preferred base-like decomposition
cules.[47] Due to the presence of many unpaired electrons and pathway, making OO bond broken into two OH* followed by
the planar structure accepting electrons from O2, poly- the formation of H2O* and O*. After O* further attacked organic
(ethylene glycol) hydrophilic carbon clusters (PEG-HCCs) offered substrates (e.g., TMB) to obtain H atoms, the catalytic process of
superior superoxide dismutase-like activity, but was inert to nitric noble metal nanozymes was completed. Under base conditions,
oxide (NO) and peroxynitrite (ONOO).[48] Unlike above nano- OH* was pre-adsorbed on nanozyme surface, resulting in the
zymes, melanin nanoparticles (MeNPs) with superoxide acid-like decomposition pathway of H2O2. The O–H bond of
dismutase-like activity could scavenge O2, NO and ONOO H2O2* broke and transferred a H to OH generating HO2* and
simultaneously.[49] Nanoceria was one of the first reported nano- H2O*. The latter would quickly transferred one H to another
materials that exhibited superoxide dismutase-mimicking activi- H2O2, forming O2*、H2O* and OH*. (2) For iron oxide nanozymes
ties, which were ascribed to convertible valence state (Ce3+ and (IONzymes), in acidic conditions, IONPs with peroxidase-like activ-
Ce4+) and oxygen vacancies. Oxygen vacancy could combine with ity catalyze H2O2 into OH by Fenton or Haber-Weiss reaction.
superoxide, transferring electron via the generation of H2O2, while However, in neutral and alkaline conditions, IONzymes directly
the Ce4+ could switch back to the original Ce3+ through the oxida- decompose H2O2 into H2O and O2 due to the catalase-like activity.
tion of H2O2.[45] Research showed that the multiple enzyme-like behaviors of ION-
zymes were mainly owned to redox potential of H2O2 in different
3.5. Hydrolase pH environments, while H2O2 activation and free radical genera-
tion were closely associated with the reversible redox of iron ion.
Hydrolase, one of common enzyme activities, plays a vital role Since electrons were transferred from a low potential to the high
in biological systems by catalyzing hydrolysis of chemical bond. one, Fe3+/Fe2+ (0.771 V) could not transfer electrons from TMB
Emerging studies have discovered that various nanomaterials (TMBOx /TMBRe 1.13 V) to H2O2 (1.776 V) in low pH environment.
(such as Au, CeO2, Ce-AuNPs, MOFs, Fe3O4, ZrO2) exhibit excellent When in the high pH environment, H2O2 was directly decomposed
hydrolase-like activity. due to the reduced redox potential (0.695 V).[24] (3) Prussian blue
Although minimal studies involve the catalytic mechanism of (PB) nanozymes, one of iron-based nanozymes, are alternately con-
hydrolase-like activity in nanozymes, bond breakage is closely nected by Fe2+–CN–Fe3+ to form cyanide-bridged cubic networks.
related to free radicals. As previously reports, MOF/g-C3N4 displays [7] Unlike IONPs, PB was found with the ability of scavenging ROS
hydrolases-like activity towards dimethyl chlorophosphate (DMCP) [34]. Due to various redox potentials and adjustable energy levels,
via two steps, namely adsorption and decomposition of substrates. Prussian blue served as efficient electron transporters and pos-
First, the vaporized DMCP molecules are adsorbed through the sessed pH-dependent enzyme-like activity. At the acidic environ-
complexation of DMCP molecule on copper sites by its P=O group, ment, high activity H2O2 oxidized Prussian blue (PY/BG: 1.4 V) by
then there are specific interactions between DMCP and copper- transferring electrons from TMB (TMBOx /TMBRe 1.13 V) to H2O2
based amorphous phase, including the formation of hydrogen (H2O2 /H2O 1.776 V), contributing to the peroxidase activity of
bonds or the polar interactions between P=O group and hydroxyl Prussian blue. When in the high pH environment, the redox poten-
groups of uncoordinated BTC units or positively charged Cu2+, tial of H2O2/ O2 was exceedingly low, making H2O2 more easily oxi-
respectively. Second, the chlorine atom of DMCP is replaced by dized into O2. In this case, fewer PB were oxidized into BG or PY to
hydroxyl group in water molecules, and DMCP is hydrolyzed to transfer electrons from TMB to H2O2. Hence, Prussian blue exhib-
7
W. Yang, X. Yang, L. Zhu et al. Coordination Chemistry Reviews 448 (2021) 214170

Fig. 3. The multi-enzyme activities of nanozymes. (a-c) The pH-depended multi-enzyme activities of noble metal nanozymes (a)[7], Prussian blue (b)[34], and Co3O4 (c)[7].
(d) The multi-enzyme activities of CeO2 depend on the size, surface ion, and morphology.[18]

ited catalase-like activity. Interestingly, because H2O2 could be 4. Biological applications of nanozymes
transformed into H2O or O2 by the POD- or CAT-like activity, Prus-
sian blue were capable of catalyzing dismutation of O2 due to the Reactive oxygen and nitrogen species (RONS) play an essential
presence of O2/O2 (0.73 V) and O2/H2O2 (1.5 V). Therefore, Prus- role in cell proliferation, migration, differentiation, signal transduc-
sian blue could mimic superoxide dismutase at different pH levels tion, tissue homeostasis, and immunity, as well as the defense
(including acidic and alkaline) (Fig. 3b). (4) Co3O4 also exhibits pH- against pathogenic invasion.[1,69] However, excessive active spe-
dependent enzyme-mimicking activities due to high redox poten- cies will cause a series of diseases, such as stroke, sepsis, diabetes,
tial of Co3+/CO2+ (1.3 V). The mechanism is similar to that of Prus- hypertension, neurodegenerative diseases, inflammation, and can-
sian blue. Under acidic condition, Co3O4 can transfer the lone pair cer.[69] Under normal physiological conditions, intracellular RONS
electrons of amino group from TMB to H2O2. The increased electron level is modulated by antioxidant enzymes and some small-
density and flow rate on nanozymes accelerate electron transfer molecule antioxidants.[1,70,71] Due to the intrinsic enzyme-
process, which is attributed to the peroxidase-like activity. How- mimicking characteristics, nanozymes can participate in RONS
ever, under neutral /alkaline conditions, the oxidation ability of removal of body. In addition, because of the excellent RONS pro-
Co3O4 is very high, thus Co3+ directly oxidizes H2O2. This is the cat- duction capacity, nanozymes can be applied in cancer therapy,
alytic behaviors of CAT and SOD in Co3O4, and its CAT activity is sterilization, and disinfection. Based on above, this section will
much higher than that of IONzymes (Fig. 3c). [32,33] comprehensively summarize the biological applications of nano-
The multiple enzymatic behaviors in nanozymes are also closely zymes, including anti-inflammatory and cytoprotection, neurolog-
related to their size, surface ions, and morphology. For example, ical disease therapy, cancer therapy, sterilization and disinfection.
the catalytic activities of CeO2 depend on surface ions and size.
In the fluorite structure of CeO2, each cerium atom is surrounded
by eight oxygen anions, and each oxygen atom occupies a tetrahe- 4.1. Anti-inflammatory and cytoprotection
dral position. Many inherent oxygen holes usually exist in this
structure, producing Ce3+.[66] Therefore, there are two valence ROS are the products of normal cell metabolism in organisms,
ions and a special redox cycle (Ce3+/Ce4+) on CeO2 surface. Smaller but ROS in excess will give rise to cellular oxidative stress, affecting
CeO2 possessed more Ce3+ and oxygen vacancy at their surface, its normal growth, metabolism, and basic functions. [72–74] A
which facilitated the SOD activity. On the contrary, large sized variety of nanozymes possess antioxidant properties, which can
CeO2 was conducive to CAT activity. [15,45,63,67,68] In addition, regulate ROS levels to protect cells and promote growth. Cerium
the catalytic activity of CeO2 also depends on morphology. Wu nanoparticles are one of the nanozymes with antioxidant activity.
and co-workers [17] stated that the peroxidase activity of nano- According to Singh’s reports [75], low dose Cerium nanoparticles
cube (1 0 0) plane was much higher than that of nanorod (1 0 0) effectively scavenged free radicals in human liver cells without
plane, but the superoxide dismutase activity of nanocube was eliciting natural antioxidant defense system, and prevented cells
much lower than that of the nanorod. The exposed surface of from early apoptosis and DNA damage. Another study also
CeO2 was related to Ce3+/Ce4+ concentration. CeO2 nanopolyhe- reported a similar result that the mice treated with ceria had a sub-
drons with (1 1 1) and (1 0 0) ((1 1 1)> (1 0 0)) contained a higher stantial decreased ROS level and significantly reduced damage in
Ce4+ concentration, which was beneficial to superoxide dismutase tissue and DNA.[76] Interestingly, by functionally mimicking the
activity (Fig. 3d).[18] antioxidant enzyme glutathione peroxidase, V2O5 nanowires also
8
W. Yang, X. Yang, L. Zhu et al. Coordination Chemistry Reviews 448 (2021) 214170

Fig. 4. The applications based on RONS removal of nanozymes. (a) Schematic diagram depicting the GPx-like antioxidant activity of vanadia nanowires and GSH recycling
by GR for cytoprotection; and (b) The scavenging ability of V2O5 nanowires for extrinsic H2O2 and intrinsic cellular peroxide induced by CuSO4 was measured by using
genetically encoded H2O2-specific probe HyPer in HEK293T cells.[50] (c) Preparation process of CeRMS; (d) Neural differentiation of neural stem cells after treating with
different nanoparticles for 10 days (Scale bar: 50 lm); and (e) Representative swimming path in cognitive function of 3  Tg-AD mice.[85]

exhibited robust antioxidant activity to scavenge the reactive oxy- dismutase- and catalase- mimicking activities. The anti-
gen species in physiologically conditions (Fig. 4 a, b). After being inflammatory effect of CeO2 was studied by enclosing different
internalized into cells, V2O5 nanowires fully restored the redox bal- shaped ceria on titanium surfaces for peri-implantitis prevention.
ance, alleviating the oxidative stress in cells without disturbing the [81] Consequently, nanotube and nano-octahedron CeO2-
cellular antioxidant defense. Furthermore, V2O5 nanowires pre- modified titanium exhibited a satisfactory anti-inflammatory
vented oxidative damage of cellular components (such as lipids, effects and ROS-scavenging ability in vitro, while nano-
proteins, and DNA), providing crucial cytoprotection for biomole- octahedron CeO2 showed stronger anti-inflammatory effect
cules.[50] NACu-Cys nanozymes with superoxide dismutase in vivo. In addition, CeO2 grown on montmorillonite surface could
mimetic activity could also eliminate O2, acting as a protective effectively reduce the inflammatory reaction by scavenging ROS
agent against oxidative stress. Adding NACu-Cys into human (O2, H2O2, OH), decreasing pro-inflammatory cytokines, and
sperm freezing medium, would raise cell motility and viability as increasing anti-inflammatory cytokines.[82] Due to the presence
well as reduce its apoptosis.[77] Compared with natural superox- of double oxidation states (Mn2+ and Mn3+), Mn3O4 nanoparticles
ide dismutase, the NACu-Cys nanozymes showed better cell pro- also exhibited various enzyme-mimicking activities (i.e., superox-
tection ability. ide dismutase and catalase), endowing them with O2 and OH
ROS in excess generally cause uncontrollable inflammation that scavenging ability. [83] By contrast, they were more stable than
is the underlying pathogenesis of most diseases.[78] Nanozymes corresponding natural enzymes and superior to CeO2 nanozymes
with ROS scavenging ability can alleviate inflammation and avoid in ROS elimination. Moreover, the Mn3O4 nanoparticles exhibited
associated diseases emerging.[79] Inorganic nanomaterials are excellent ROS removal efficacy in vitro, and were able to effectively
always utilized in photoacoustic or computed tomography con- protect the mice from ROS-induced ear-inflammation. Most of
trasts to evaluate the diagnosis and treatment of disease, but their anti-inflammatory nanozymes mainly have the ability of ROS scav-
long-term retention in the body usually increases ROS level, lead- enging, while the RNS scavenging capability is relatively weak. Cao
ing to inflammation. To avoid it, bovine serum albumin-iridium and co-workers [84] presented a polyethylene glycol-coated ultra-
oxide nanoparticles were used to catalyze the production of oxy- small rhodium nanodots (Rh-PEG NDs) that showed unexpected
gen due to the inherent catalase-mimicking activity, which not reactive oxygen and nitrogen species (RONS) scavenging properties
only enhanced imaging effect but also protected normal tissues and photothermal activities, which could be utilized in an anti-
against H2O2-induced inflammation.[80] It was also reported that inflammation in colon diseases. They found that Rh-PEG NDs
CeO2 possessed ROS scavenging capability due to the superoxide decreased the level of pro-inflammatory cytokines (TNF-a, and

9
W. Yang, X. Yang, L. Zhu et al. Coordination Chemistry Reviews 448 (2021) 214170

IL-6) because of their multiple enzyme-like activities against RONS, prepared for fast electron capture. In neutral environment, it
resulting in an excellent anti-inflammatory effect on dextran sul- attracted and removed RONS radicals, promoting brain injury
fate sodium induced colitis. treatment.[92] Nanozymes have offered many distinct advantages
in neurological diseases treatment, but the biological safety needs
4.2. Neurological disease therapy to be evaluated in the future.

ROS can cause the dysfunction of proteins and mitochondria, 4.3. Cancer therapy
inducing neuronal damage and neurological diseases. Many natu-
ral enzymes have been applied in the treatment of neurological Cancer is one of the most serious diseases threatening human
diseases. However, the efficacy cannot last long due to the health, and is often featured with vascular abnormalities, glucose
immunogenicity and limited electron-donating ability in vivo. deprivation, mild acidity, hypoxia, hydrogen peroxide overexpres-
[86,87] Nanozymes with intrinsic antioxidant enzyme properties sion, and inflammation. [93–95] The traditional treatments include
and excellent biocompatibility have a high potential to be used surgery, chemotherapy, and radiotherapy, but they usually suffer
in neurological diseases, such as Alzheimer’s disease, ischemic from unsatisfactory therapeutic outcome or even are harmful to
stroke, brain injury, and so on. Qu and co-workers [88] first pre- the function of normal cells because of postoperative infection,
sented a polyoxometalate-based multifunctional nanozyme for hypoxia, and the non-targeting effect. Amazingly, nanozymes can
Alzheimer’s disease treatment. The protease-like activity was used offer many excellent characteristics, including responsive
to deplete amyloid-beta (Ab) aggregates, and superoxide enzyme-like activities, as well as light, thermal and magnetic
dismutase-like activity for scavenging ROS-mediated Ab. Further- effects, which facilitated ROS generation or removal, oxygen gener-
more, the nanozymes with low toxicity was able to cross blood– ation, and remote-control capability. Nanozymes, overcoming the
brain barrier. Since neuron loss often occurs in Alzheimer’s disease, shortcomings of traditional methods, have been successfully
neuron-directional differentiation and neural stem cell niche com- applied in many cancer treatment strategies (Table 3), including
ponent majorization are often utilized to promote nerve growth, photodynamic therapy, sonodynamic therapy, chemodynamic
but their effects are vulnerable to high oxidative stress levels. A therapy, hyperthermia, photothermal therapy, radiotherapy, cat-
MOF-encapsulated nanozyme was designed for the modulation of alytic therapy, oxidative therapy, immunotherapy, starvation ther-
neuron-directional differentiation and the amelioration of oxida- apy, and combination therapy.
tive stress (Fig. 4 c, d, e).[85] In the lesion area, the hydrogen Numerous nanomaterials can be recognized as photosensitizers,
peroxide-responsive MOF released cargos to promote neuron- sonosensitizers, or heat conversion mediums to promote ROS pro-
directional differentiation, while the integrated ceria eliminated duction. Owning to the malignant invasiveness, low toxicity, high
ROS and averted oxidative damage, resulting in a higher survival selectivity, and enhanced therapeutic effect, nanozymes-based
rate and enhanced outgrowth of newborn neurons. Microglia are photodynamic, photothermal and sonodynamic therapy have
resident myeloid cells in the central nervous system, and it partic- become potential anticancer strategy. Recently, the well-defined
ipates in normal central nervous system functions. Elevated ROS single-atom nanozymes (OxgeMCC-r SAE) as a versatile photody-
levels convert M2 microglia into M1 microglia, inducing neurode- namic were fabricated, which could decompose endogenous
generative diseases and Alzheimer’s disease. Hu and co-workers hydrogen peroxide to generate oxygen continuously due to the
[89] designed TPP-MoS2 QDs to scavenge ROS and target mito- catalase-like activities, thereby overcoming tumor hypoxia for
chondria of microglia. Their results proved that TPP-MoS2 QDs photodynamic therapy.[96] Under NIR irradiation, high cytotoxic
could effectively crosse blood–brain barrier, target mitochondria, singlet oxygen (1O2) species were produced, which further
convert microglia, and mitigate Ab-mediated neurotoxicity, enhanced the therapeutic effect of photodynamic therapy in solid
inhibiting neuroinflammation and treating Alzheimer’s disease of tumor. In addition, the enzyme-like activities of carbon-based
mice. nanomaterials and AuNPs can be significantly enhanced, resulting
Nitrogen species (RNS) are common intermediate products of in the generation of high ROS levels. Based on this, Han and co-
many neurological diseases, affecting normal metabolism and workers [97] designed a nanozyme by encapsulating AuNPs within
functions of nerve cells. Melanin is known as a potential radical a hollow porous carbon nanosphere (Au@HCNs). Under 808 nm
scavenger in most organisms. Lu and co-workers developed mela- light irradiation, the nanozymes showed considerable photother-
nin nanoparticles with RONS scavenging capacity for the first time. mal effect. However, photo-based therapeutic models are only
[49] They found that melanin nanoparticles possessed unique suitable for treating superficial and localized cancer due to the lim-
multi-antioxidative, anti-inflammatory, and biocompatible fea- ited penetrating depth. By contrast, sonodynamic therapy, another
tures. Melanin nanoparticles could attenuate RONS-triggered noninvasive treatment mode, provides great opportunities for can-
inflammatory responses by suppressing the expression of inflam- cer treatment, in terms of tissue penetrating capability, conve-
matory mediators and cytokines, effectively protecting ischemic nience, and low cost. However, the low quantum yield and
brains with negligible side effects in vitro and in vivo. Due to the tumor hypoxia hinder the comprehensive applications of sonody-
variable valence state and extraordinary redox capability, as well namic therapy. Cheng and co-workers [98] reported a novel TiO2-
as the unique hollow structure and a large specific surface area, based nanosonosensitizer, which bilaterally enhanced the quan-
Prussian blue nanozymes also showed excellent RONS scavenging tum yield and alleviated tumor hypoxia, faciliting the chemo-
capacity for neuroprotection against ischemic stroke.[90] Experi- sonodynamic synergistic efficacy. In this Pt-TiO2 heterostructure,
mental results indicated that Prussian blue nanozymes effectively TiO2 servesed as a load reservoir of doxorubicin (an chemotherapy
protected neurons against the hypoxic and ischemic injury with drug and a sonosensitizer SDT), while the decorated Pt NPs cat-
negligible side effects. Based on the ultrahigh RNS selectivity and alyze endogenous hydrogen peroxide decomposition to provide
catalytic activity of nanozymes, ultrasmall fluorescent carbogenic sufficient oxygen for chemo-sonodynamic synergistic efficacy. Sur-
was reported.[91] This nanozymes with remarkable RNS / ROS prisingly, the synergistic effect of chemotherapy and dynamic
scavenging efficiency was able to remove harmful peroxide and acoustic therapy displayed an excellent inhibitive effect on tumor
superoxide in vivo, reducing immune responses in traumatic brain growth.
injury mice. Unlike conventional inorganic nanozymes, the nano- Chemodynamic therapy, a typical nanocatalytic therapy strat-
zymes exhibited efficient renal clearance without any side effects egy, employs Fenton or Fenton-like reactions to convert hydrogen
at high doses. In another study, a new trimetallic nanozyme was peroxide into oxidative OH. Tumor site-specific endogenous
10
W. Yang, X. Yang, L. Zhu et al. Coordination Chemistry Reviews 448 (2021) 214170

Table 3
Summary of nanozymes for cancer therapy.

Therapeutic Nanomedicine Function Mechanisms Reference


modality
Radiotherapy MnTCPP-Hf-FA MOF As CAT mimics and radiosensitizer Produce O2 to overcome tumor hypoxia [105]
Ir@liposome As CAT mimics and photothermal reagent Generate O2 to relieve tumor hypoxia [106]
Porous PtNPs As CAT mimics and radiosensitizer Generate O2 to alleviate tumor hypoxia [107]
Chemodynamic FePt-NP2 As Fenton reagent Catalyze the production of OH [104]
therapy Co-Fc@GOx As Fenton reagent Generate OH [100]
MnO2@PtCo As CAT and OXD mimics Catalytic the generation of O2 and ROS [101]
Photochemotherapy UCSRF As Fenton reagent and upconversion nanoparticles NIR light-upconverted UV/vis light, and produce [108]
abundant ROS
Photodynamic Au@ZIF-8 As CAT mimics Catalyze the production of O2 and 1O2 [109]
therapy OxgeMCC-r As CAT mimics For continuous oxygen generation [96]
PCN-224-Pt As CAT mimics and photosensitizer Catalyze the formation of O2 and 1O2 [110]
Lipo-Ogzyme-AIE As CAT mimics Supply oxygen for ROS generation [111]
Hyperthermia magnetic hydrogel As POD mimics Generate OH via Fenton reaction [112]
Au@HCNs As POD, OXD, and photothermal reagent For ROS generation [97]
Catalytic therapy DMSN-Au-Fe3O4-PEG As glucose oxidase and POD mimics Generate H2O2 and OH [93]
GSF@AuNPs As POD mimics Catalyzed H2O2 to form OH [113]
N-PCNSs As OXD, POD, CAT and SOD mimics Boost ROS generation [114]
GOx-Fe3O4@DMSNs As Fenton reagent Generate a considerable amount of OH [115]
FePOs As POD mimics Catalyze OH generation to anti-tumor [116]
PtAu@SiO2 As OXD and POD mimics Catalyze the production of O2 and ROS [117]
Combination PEG/Ce-Bi@DMSN As POD, CAT mimics, and photothermal reagent, Produce O2 and OH, whiling depleting GSH to [118]
therapy with GSH-depletion capability weaken tumor’s reducibility
AuNBPs@MoS2 As POD mimics and photothermal reagent Generate ROS (1O2 and OH) [119]
H-Pt-TiO2-DOX As CAT mimics and sonosensitizer Generate O2 and ROS [98]
GOx/hPB@gellan As CAT mimics and photothermal agent Convert H2O2 into O2 [120]
PtFe@Fe3O4 As POD, CAT, and photothermal agent Generate O2 and ROS (1O2 and OH) [121]
PtCu3-PEG nanocages As POD, GSH peroxidase, and sonosensitizer Generate ROS and reduce ROS clearance via [122]
accelerate GSH depletion
HMPB@GOx As SOD, CAT, POD, OXD, and photothermal agent Catalyze the production of H2O2, O2, and ROS [123]
(OH and 1O2)
HMON-Au-Col@Cu-TA- As glucose oxidase, Fenton reagent, and Self-supply H2O2 and convert into OH [99]
PVP photosensitizer
ABTS@MIL-100/PVP As POD mimics and photothermal agent Produce OH and reduce GSH level [124]
rMGB As CAT mimics Catalyze the production of O2 and ROS [125]
Pt-CuS-P-TAPP As CAT mimics and photothermal agent Generate O2 to overcome hypoxia [126]
Pt@UiO-66-NH2@Aushell- As CAT mimics and photothermal agent Decompose H2O2 to supply O2 [127]
Ce6
Au2Pt-PEG-Ce6 As CAT, POD, photothermal agent, and Generate O2 and OH [62]
sonosensitizer
SnFe2O4 As CAT, GSH peroxidase, Fenton reagent, and Produce O2 and ROS, consume GSH to relieve [128]
photothermal agent antioxidant capability
IrOx-Gox As POD, OXD, and CAT mimics Supply O2 and ROS, and consume GSH [129]
IrRu-GOx@PEG NPs As POD and CAT mimics Convert H2O2 into 1O2 and O2 [130]
Au/Pt star@S-S@ As POD mimics and photothermal reagent Catalyze H2O2 to produce toxic OH [102]
rHSAFA@IR780 @GOx
IMSN-PEG-TI As POD and CAT mimics Decompose H2O2 into OH and O2 [61]
CaO2/DOX@SiO2/DOX– As CAT mimics Generate H2O2 and O2 [131]
MnO2 NR
MON-p53 As Fenton reagent Produce ROS to cause lipid peroxidation [103]

chemical energy is exploited to produce OH, protecting normal tis- Wei [61] proposed an immunomodulation-enhanced nanozyme-
sues from oxidative damage without external energy input. [93,99] based tumor catalytic therapy strategy, in which iron manganese
For instance, Co-Fc NMOF not only exhibited an excellent Fenton silicate nanoparticles (IMSN) with POD- and CAT-like activities
effect, but also acted as a versatile delivery medium of glucose oxi- decomposed hydrogen peroxide into OH and O2 under acidic
dase (GOx) molecules. After glucose oxidase was delivered into TME, while TGF-b inhibitor (TI) regulated tumor immune microen-
tumor and endogenous glucose was catalyzed to form hydrogen vironment to induce the regeneration of hydrogen peroxide, pro-
peroxide, Co-Fc@GOx dramatically amplified the induction of cyto- moting the catalytic activities of IMSN nanozymes (Fig. 5 a, b, c).
toxic OH in situ, enhancing the inhibition effect against cancer The related results suggested that the synergism effect of nano-
cells.[100] Furthermore, the catalase-like activity of MnO2 could zymes and TME regulation could achieve a more optimistic thera-
rapidly decompose hydrogen peroxide into oxygen to address peutic effect. In most cases, due to the complex tumor
tumor hypoxic, while the oxidase-mimicking behavior of PtCo cat- microenvironment, it is generally difficult to achieve desired effect
alyzed oxidation reaction to generate ROS. As a result, MnO2@PtCo by single treatment strategy. Therefore, synergistic therapy strate-
nanoflowers induced significant cell apoptosis via the ROS- gies often attract more attentions. Recently, Au/Pt star@S-S@rHSA-
mediated mechanism, resulting in remarkable and specific tumor FA@IR780@GOx was reported, in which iodide diagnostic probe
growth inhibition.[101] However, the therapeutic outcomes of (IR780) for real-time imaging, photothermal reagent (Au) for pho-
nanozyme-based tumor catalytic therapy are diminished due to tothermal therapy, photosensitizer (IR780) for photodynamic ther-
many factors in tumor microenvironment (TME), including insuffi- apy, glucose consuming enzyme (GOx) for starvation therapy, and
cient endogenous hydrogen peroxide concentration, hypoxia, and peroxidase-mimicking ability (Pt) for oxidative therapy. Once the
immunosuppressive microenvironment. In view of this, Liu and disulfide linker was cleaved by GSH in tumor sites, IR780 were

11
W. Yang, X. Yang, L. Zhu et al. Coordination Chemistry Reviews 448 (2021) 214170

Fig. 5. The applications based on RONS generation of nanozymes. (a) Schematic illustration of synthesis and tumor therapy for IMSN-PEG-TI; (b) Tumor volume of CT26-
tumor-bearing mice after different treatments at 15 days; and (c) Survival of CT26-tumor-bearing mice after different treatments.[61] (d) Schematic illustrating PCN-224-Ag-
HA nanoagents with surface adaptive characteristic for on-demand synergistic bacteria killing; (e) Photographs of MRSA-infected wounds on mice in each group with
different treatments during on the third day; and (f) The bacteria separated from wound tissue after different treatments were cultured on agar plates.[132]

released and GOx catalyzed intracellular glucose to generate healing.[133] Numerous nanozymes as powerful sterilization tools
hydrogen peroxide, enhancing the local acidity. The Pt layer with have been developed due to their excellent ROS production capac-
peroxidase-like properties catalyzed hydrogen peroxide to produce ity. For instance, Au/g-C3N4 hybrid nanozymes displayed strong
toxic OH for tumor oxidative damage. Consequently, it could peroxidase-like activity, which could efficiently catalyze biologi-
simultaneously realize real-time imaging and synergetic cancer cally relevant concentrations of H2O2 decompose into OH radicals,
treatment via starving-like therapy/enzyme oxidative therapy/ showing great potential in preventing bacterial infection. By break-
photothermal therapy/photodynamic therapy.[102] Compared ing down drug-resistant-biofilms and preventing new biofilm for-
with the single treatment strategy, combination therapy is more mation, the hybrid nanozymes possessed remarkable bactericidal
effective and more likely to achieve successful cancer treatment ability against both drug-resistant Gram-negative and Gram-
in the future. positive bacteria. Particularly, the hybrid nanozymes could signifi-
Cancer cells proliferate rapidly, whereas the ROS generated by cantly prevent bacterial infections and accelerate wound healing at
nanozymes causes oxidative damage to lipids, proteins, and DNA lower H2O2 concentration. [133] According to the report, MOF/Ce-
in the cancer cells, affecting their basic biological functions or even based nanozymes are also conducive to sterilization due to the
leading to death.[103] As reported, Metal-organic network (MON) deoxyribonuclease- and peroxidase- mimicking activities.[134]
encapsulated with p53 plasmid (MON-p53) eradicated cancer cells The nanozymes not only killed bacteria via ROS, but also hydro-
via the ferroptosis/apoptosis hybrid pathway by harnessing the lyzed extracellular DNA, disrupting the established biofilms and
oxidative stress regulation ability of p53 and the Fenton avoiding bacteria recolonization and biofilm recurrence. In addi-
reaction-inducing capability of metal–organic network.[104] The tion, metal ion release from metallic-based nanozymes is also a
ferric ions in p53 plasmid catalyzed the overexpressed hydrogen favorable condition for sterilization. It was shown that the released
peroxide to form ROS in the tumor environment, causing serious Cu2+ from CuO-modified copper/carbon nanozymes caused mem-
lipid peroxidation of biofilm. Furthermore, p53 genes encapsulated brane damage, lipid peroxidation, and DNA degradation in Gram-
in p53 plasmid was released, inducing the expression of p53 pro- negative bacteria.[135] Ag ions was also a potential antibacterial
tein, further inhibiting lipid peroxide elimination and sensitizing material, in order to eliminate their toxic effect, a novel antimicro-
cells to ferroptosis. A 75-day anti-cancer experiment treatment bial nanoplatform was fabricated by combining hyaluronic acid
confirmed this treatment strategy. protected Ag ions with metal–organic frameworks (Fig. 5 d, e, f).
[132] Owing to the released Ag ions and generated reactive oxygen
4.4. Sterilization and disinfection species, the nanoplatform provided a synergistic antibacterial
effect and showed good biocompatibility with nontargeted cells.
Bacterial infection often causes significant morbidity and mor- For ROS-based sterilization, effective capture and reaction of bacte-
tality, which has become one of the substantial health challenges. ria by nanozymes are prerequisites for efficient sterilization,
H2O2 is widely used for bacterial inactivation and wound disinfec- because the intrinsical lifetime (shorter than 200 ns) and diffusion
tion. However, its dosage exceeds physiological levels, often caus- distance (approximately 20 nm) of ROS are limited. Wang and co-
ing potential toxicity to healthy tissue or even delaying wound workers [136] provided an antibacterial nanozyme (Cu2WS4

12
W. Yang, X. Yang, L. Zhu et al. Coordination Chemistry Reviews 448 (2021) 214170

nanocrystals, CWS NCs), it first effectively adsorbed bacteria via origin of nanozymes can be revealed from the perspectives
the interaction between copper atoms of CWS NCs and amino of nano effects. Perhaps it is the reason why nanozymes pos-
groups from peptidoglycan of bacterial cell wall. Then bacteria sess properties differed from natural enzymes. However,
were killed by ROS produced by nanozyme oxidase-like and related research is still very limited, thus more exploration
peroxidase-like activities. Consequently, the CWS NCs with a low is needed to reveal the source of nanozyme activity in the
dose achieved > 99.999 % inactivation against both Gram- future.
negative and Gram-positive bacteria, which was far superior to 2) Further studying the relationship between nanozyme activ-
typical antibacterial nanomaterials (Ag and TiO2 nanoparticles) ity and nanomaterial characteristics. Scientists have found
and antibiotics (vancomycin and daptomycin) in the same experi- that nanozyme activity is closely related to the composition,
mental conditions. Recently, an effective method for capture and size, morphology and crystal surface of nanomaterial. For
removal bacteria was proposed by combining hydrogel with example, the activity of metal nanozymes is associated with
molybdenum disulfide (MoS2) nanomaterials.[137] It used hydro- the metal material: Au(1 1 1) < Ag(1 1 1) < Pt(1 1 1) < Pd(1 1 1).
gel with favorable biocompatibility to effectively capture and con- [43] The peroxidase-like activity of iron oxide nanozymes is
fine the bacteria within the scope of ROS damage through as follows: 30 nm > 150 nm > 300 nm.[2] The glutathione
electrostatic interaction, while the MoS2 produced OH relying on peroxidase activity of V2O5 depends on its morphology and
their intrinsic peroxidase-like activity. Treating with MoS2- crystal plane.[16] However, the relevant reports are very
hydrogel had a decidedly antibacterial effect, decreasing inflamma- limited. In addition, the reason why the catalytic activity
tion and promoting wound healing. of nanozymes is closely related to nanomaterial characteris-
Due to the excellent ability of ROS production, nanozymes have tics remains to be studied and analyzed in the future.
drawn a lot of attention in virus inactivation. Besides peroxidase- 3) Systematically researching the catalytic mechanism of dif-
and catalase-like activities, iron oxide nanoparticles performed ferent nanozymes. Although more and more nanozymes
lipoxidase activity that could induce membrane lipid peroxidation and enzymatic activities have been proposed, their catalytic
in synthesized liposome. [138,139] If lipid peroxidation occurred in mechanisms are different as nanomaterials and reaction
virus envelopes, the integrity of neighboring proteins was conditions. So far, the understanding of catalytic mechanism
destroyed, causing virus inactivation. Therefore, an antiviral strat- is far from enough. In order to accurately control the cat-
egy was developed by using iron oxide nanoparticles to target the alytic process of nanozymes, it is important to systemati-
lipid envelope of the influenza virus.[140] These iron oxide cally research the catalytic mechanism of various
nanoparticles showed broad-spectrum antiviral activity in 12 viral nanozymes.
subtypes (H1 ~ H12), and their application in facemask improved 4) Improve their catalytic activity and substrate selectivity. At
the antiviral ability against H1N1, H5N1, and H7N9 subtype. In present, by simulating the structure of enzymes, single atom
addition to various enzyme-like activities of nanozymes, the exten- nanozymes have shown comparable catalytic activity and
sive specific surface area of nanomaterials is conducive to viral substrate selectivity [142], but most nanozymes still require
adsorption. For example, although TiO2 could generate a large to be improved. Therefore, more efforts need to endow
amount of HO, the adsorption of microbial phage MS2 (a common nanozymes with amino acid-like structures, precisely con-
patrol virus) on the unmodified TiO2 was little, showing a limited trol the coordination structure of active center atoms, and
antibacterial effect. However, when photocatalyst SiO2 was modi- improve atom utilization.
fied on TiO2 surface, the adsorption against MS2 was enhanced sig- 5) Further exploring new enzyme-mimicking activities of
nificantly, which was 37 times than that of unmodified TiO2.[141] nanozymes. The activity types discovered in nanozymes
are very limited, mainly including oxidoreductase and
5. Conclusions and prospects hydrolase. Therefore, it is crucial to further explore potential
enzymatic activity of nanozymes.
Since the discovery of Fe3O4 nanozyme in 2007, nanozymes 6) Evaluating the biological safety of nanozymes. A remarkable
have attracted much attention due to the same activity and similar feature of nanozymes is that they can act as nanodrugs
function as natural enzymes. Compared with natural enzymes, in vivo and their catalytic behaviors can be controlled remo-
nanozymes offer distinct advantages, including low cost, high sta- tely. However, the biological safety should be considered
bility, good durability, controllable activity, easy storage, and first when acting in vivo. The safety and potential toxicity
excellent reusability. Therefore, they have been widely applied in are main challenges in further application of nanozymes.
many fields, including online monitoring, imaging, anti- Therefore, the control and decrease of potential toxicity or
immunity, cell protection, disease prevention and therapy, antibac- developing safe and non-toxic nanozymes is worthy of our
terial and promote wound healing, and simulating complex biolog- attention.
ical structure /system. With the technology development and in- 7) Further refining the definition of related enzymes. With the
depth research, nanozymes have made great progress in theory development of science and technology, scientists have
and application. In this review, we revealed the activity origin of found more and more types of ‘‘enzymes”, covering natural
nanozymes, summarized the catalytic mechanism of common enzymes, artificial enzymes and nanozymes, but some are
activities, and reviewed RONS regulation-based biological applica- still controversial [143]. Thus, in order to promote people’s
tions of nanozymes. Although fruitful progress has been made, understanding of enzymes, it is necessary for scientists to
nanozymes are still facing many challenges. update or further clarify the definition and characteristics
of related enzymes.
1) The activity origin of nanozymes. By structure comparison of
natural enzymes and nanozymes, it is found that they have
many similarities. However, the structure of natural Declaration of Competing Interest
enzymes is much more complex than that of nanozymes.
Therefore, in order to obtain the same function as natural The authors declare that they have no known competing finan-
enzymes, it is necessary to further study the microstructure cial interests or personal relationships that could have appeared
of natural enzymes and nanozymes. In addition, the activity to influence the work reported in this paper.

13
W. Yang, X. Yang, L. Zhu et al. Coordination Chemistry Reviews 448 (2021) 214170

Acknowledgments [44] J. Mu, L. Zhang, M. Zhao, Y. Wang, Journal of Molecular Catalysis a-Chemical
378 (2013) 30–37.
[45] I. Celardo, J.Z. Pedersen, E. Traversa, L. Ghibelli, Nanoscale 3 (2011) 1411–
This work was supported by the National Natural Science Foun- 1420.
dation of China (No. 31671922 and No. 31871875); the National [46] X. Shen, W. Liu, X. Gao, Z. Lu, X. Wu, X. Gao, J. Am. Chem. Soc. 137 (2015)
15882–15891.
Key Research and Development Program (No. 2018YFC1603704);
[47] S.S. Ali, J.I. Hardt, K.L. Quick, J.S. Kim-Han, B.F. Erlanger, T.T. Huang, C.J.
and 2115 Talent Development Program of China Agricultural Epstein, L.L. Dugan, Free Radical Biol. Med. 37 (2004) 1191–1202.
University (No. 00109016). [48] E.L.G. Samuel, D.C. Marcano, V. Berka, B.R. Bitner, G. Wu, A. Potter, R.H. Fabian,
R.G. Pautler, T.A. Kent, A.-L. Tsai, J.M. Tour, PNAS 112 (2015) 2343–2348.
[49] Y. Liu, K. Ai, X. Ji, D. Askhatova, R. Du, L. Lu, J. Shi, J. Am. Chem. Soc. 139 (2017)
856–862.
References [50] A.A. Vernekar, D. Sinha, S. Srivastava, P.U. Paramasivam, P. D’Silva, G. Mugesh,
Nat. Commun. 5 (2014).
[51] H. Ye, K. Yang, J. Tao, Y. Liu, Q. Zhang, S. Habibi, Z. Nie, X. Xia, ACS Nano 11
[1] H. Wang, K. Wan, X. Shi, Adv. Mater. 31 (2019).
(2017) 2052–2059.
[2] L. Gao, J. Zhuang, L. Nie, J. Zhang, Y. Zhang, N. Gu, T. Wang, J. Feng, D. Yang, S.
[52] H. Sun, A. Zhao, N. Gao, K. Li, J. Ren, X. Qu, Angewandte Chemie-International
Perrett, X. Yan, Nat. Nanotechnol. 2 (2007) 577–583.
Edition 54 (2015) 7176–7180.
[3] H. Wei, E. Wang, Chem. Soc. Rev. 42 (2013) 6060–6093.
[53] Z. Zhang, X. Zhang, B. Liu, J. Liu, J. Am. Chem. Soc. 139 (2017) 5412–5419.
[4] S. Wu, J. Zhang, P. Wu, Anal. Methods 11 (2019) 5081–5088.
[54] H. Tan, Y. Zhao, X. Xu, Y. Sun, Y. Li, J. Du, Microchim. Acta 187 (2020).
[5] L. Gao, X. Gao, X. Yan, Kinetics and Mechanisms for Nanozymes, Springer,
[55] F. Li, N. Li, C. Xue, H. Wang, Q. Chang, H. Liu, J. Yang, S. Hu, Chem. Eng. J. 382
Singapore, 2020.
(2020).
[6] H. She, B. Wang, Finite Elem. Anal. Des. 45 (2009) 463–467.
[56] T. Cai, G. Fang, X. Tian, J.-J. Yin, C. Chen, C. Ge, ACS Nano 13 (2019) 12694–
[7] H. Dong, Y. Fan, W. Zhang, N. Gu, Y. Zhang, Bioconjug. Chem. 30 (2019) 1273–
12702.
1296.
[57] Y. Ding, G. Wang, F. Sun, Y. Lin, ACS Appl. Mater. Interfaces 10 (2018) 32567–
[8] Y. Sun, C. Zhao, N. Gao, J. Ren, X. Qu, Chemistry-a European Journal 23 (2017)
32578.
18146–18150.
[58] D. Yang, Q. Li, S.K. Tammina, Z. Gao, Y. Yang, Sensors and Actuators B-
[9] M. Sun, L. Xu, A. Qu, P. Zhao, T. Hao, W. Ma, C. Hao, X. Wen, F.M. Colombari, A.
Chemical 319 (2020).
F. de Moura, N.A. Kotov, C. Xu, H. Kuang, Nat. Chem. 10 (2018) 821–830.
[59] X. Wu, T. Chen, Y. Chen, G. Yang, J. Mater. Chem. B 8 (2020) 2650–2659.
[10] F. Li, S. Li, X. Guo, Y. Dong, C. Yao, Y. Liu, Y. Song, X. Tan, L. Gao, D. Yang,
[60] S. Lin, Y. Cheng, H. Zhang, X. Wang, Y. Zhang, Y. Zhang, L. Miao, X. Zhao, H.
Angewandte Chemie-International Edition 59 (2020) 11087–11092.
Wei, Small 16 (2020).
[11] N.C. Veitch, Phytochemistry 65 (2004) 249–259.
[61] B. Xu, Y. Cui, W. Wang, S. Li, C. Lyu, S. Wang, W. Bao, H. Wang, M. Qin, Z. Liu,
[12] C. Zhao, C. Xiong, X. Liu, M. Qiao, Z. Li, T. Yuan, J. Wang, Y. Qu, X. Wang, F.
W. Wei, H. Liu, Adv. Mater. 32 (2020).
Zhou, Q. Xu, S. Wang, M. Chen, W. Wang, Y. Li, T. Yao, Y. Wu, Y. Li, Chem.
[62] M. Wang, M. Chang, Q. Chen, D. Wang, C. Li, Z. Hou, J. Lin, D. Jin, B. Xing,
Commun. 55 (2019) 2285–2288.
Biomaterials 252 (2020).
[13] W. Du, Research on theory and engineering application of nano material
[63] T. Pirmohamed, J.M. Dowding, S. Singh, B. Wasserman, E. Heckert, A.S.
chemistry, University of Electronic Science and Technology Press, Chengdu,
Karakoti, J.E.S. King, S. Seal, W.T. Self, Chem. Commun. 46 (2010) 2736–2738.
2018.
[64] D.A. Giannakoudakis, Y. Hu, M. Florent, T.J. Bandosz, Nanoscale Horiz. 2
[14] L. Cao, J. Huang, X. Zhao, J. Li, Y. Zhuo, F. Yu, S. Zhou, X. Zhang, W. Zhou, Y. Li,
(2017) 356–364.
X. He, Wet chemical synthesis and photocatalytic properties of tungstate
[65] C. Hao, R. Gao, Y. Li, L. Xu, M. Sun, C. Xu, H. Kuang, Angewandte Chemie-
nanomaterials, 2017.
International Edition 58 (2019) 7371–7374.
[15] S. Deshpande, S. Patil, S. Kuchibhatla, S. Seal, Appl. Phys. Lett. 87 (2005).
[66] J.C. Conesa, Surf. Sci. 339 (1995) 337–352.
[16] S. Ghosh, P. Roy, N. Karmodak, E.D. Jemmis, G. Mugesh, Angewandte Chemie-
[67] V. Baldim, F. Bedioui, N. Mignet, I. Margaill, J.F. Berret, Nanoscale 10 (2018)
International Edition 57 (2018) 4510–4515.
6971–6980.
[17] Y. Yang, Z. Mao, W. Huang, L. Liu, J. Li, J. Li, Q. Wu, Sci. Rep. 6 (2016).
[68] E.G. Heckert, A.S. Karakoti, S. Seal, W.T. Self, Biomaterials 29 (2008) 2705–
[18] T. Naganuma, Nano Res. 10 (2017) 199–217.
2709.
[19] M. Peng, C. Dong, R. Gao, D. Xiao, H. Liu, D. Ma, ACS Central Science (2020).
[69] C.A. Ferreira, D. Ni, Z.T. Rosenkrans, W. Cai, Nano Res. 11 (2018) 4955–4984.
[20] J. Wu, X. Wang, Q. Wang, Z. Lou, S. Li, Y. Zhu, L. Qin, H. Wei, Chem. Soc. Rev. 48
[70] M. Lu, C. Wang, Y. Ding, M. Peng, W. Zhang, K. Li, W. Wei, Y. Lin, Chem.
(2019) 1004–1076.
Commun. 55 (2019) 14534–14537.
[21] B. Rogers, S. Pennathur, J. Adams, Nanotechnology : understanding small
[71] N. Singh, M.A. Savanur, S. Srivastava, P. D’Silva, G. Mugesh, Angewandte
systems /, 2nd ed., CRC Press, 2011.
Chemie-International Edition 56 (2017) 14267–14271.
[22] Y. Huang, J. Ren, X. Qu, Chem. Rev. 119 (2019) 4357–4412.
[72] C. Hao, A. Qu, L. Xu, M. Sun, H. Zhang, C. Xu, H. Kuang, J. Am. Chem. Soc. 141
[23] X. Ai, Y. Wang, X. Hou, L. Yang, C. Zheng, L. Wu, Analyst 138 (2013) 3494–
(2019) 1091–1099.
3501.
[73] H. Xiang, W. Feng, Y. Chen, Adv. Mater. 32 (2020).
[24] Z. Chen, J.-J. Yin, Y.-T. Zhou, Y. Zhang, L. Song, M. Song, S. Hu, N. Gu, ACS Nano
[74] B. Yang, Y. Chen, J. Shi, Chem. Rev. 119 (2019) 4881–4985.
6 (2012) 4001–4012.
[75] R. Singh, S. Singh, Colloids and Surfaces B-Biointerfaces 175 (2019) 625–635.
[25] R.C.C. Costa, M.F.F. Lelis, L.C.A. Oliveira, J.D. Fabris, J.D. Ardisson, R. Rios, C.N.
[76] S. Das, C.J. Neal, J. Ortiz, S. Seal, Nanoscale 10 (2018) 21069–21075.
Silva, R.M. Lago, J. Hazard. Mater. 129 (2006) 171–178.
[77] F. Dashtestani, H. Ghourchian, A. Najafi, Bioorg. Chem. 80 (2018) 621–630.
[26] H. Niu, D. Zhang, S. Zhang, X. Zhang, Z. Meng, Y. Cai, J. Hazard. Mater. 190
[78] J. Gao, S. Wang, Z. Wang, Biomaterials 135 (2017) 62–73.
(2011) 559–565.
[79] S. Munir, A.A. Shah, H. Rahman, M. Bilal, M.S.R. Rajoka, A.A. Khan, M.
[27] J.F. Perez-Benito, J. Phys. Chem. A 108 (2004) 4853–4858.
Khurshid, Biotechnol. Lett. 42 (2020) 357–373.
[28] H. Yang, R. Yang, P. Zhang, Y. Qin, T. Chen, F. Ye, Microchim. Acta 184 (2017)
[80] W. Zhen, Y. Liu, L. Lin, J. Bai, X. Jia, H. Tian, X. Jiang, Angewandte Chemie-
4629–4635.
International Edition 57 (2018) 10309–10313.
[29] D. Jiang, D. Ni, Z.T. Rosenkrans, P. Huang, X. Yan, W. Cai, Chem. Soc. Rev. 48
[81] X. Li, M. Qi, X. Sun, M.D. Weir, F.R. Tay, T.W. Oates, B. Dong, Y. Zhou, L. Wang,
(2019) 3683–3704.
H.H.K. Xu, Acta Biomater. 94 (2019) 627–643.
[30] R. Zhao, X. Zhao, X. Gao, Chemistry-a European Journal 21 (2015) 960-+.
[82] S. Zhao, Y. Li, Q. Liu, S. Li, Y. Cheng, C. Cheng, Z. Sun, Y. Du, C.J. Butch, H. Wei,
[31] Y. Song, K. Qu, C. Zhao, J. Ren, X. Qu, Adv. Mater. 22 (2010) 2206–2210.
Adv. Funct. Mater. 30 (2020).
[32] J. Dong, L. Song, J.-J. Yin, W. He, Y. Wu, N. Gu, Y. Zhang, ACS Appl. Mater.
[83] J. Yao, Y. Cheng, M. Zhou, S. Zhao, S. Lin, X. Wang, J. Wu, S. Li, H. Wei, Chem.
Interfaces 6 (2014) 1959–1970.
Sci. 9 (2018) 2927–2933.
[33] J. Mu, Y. Wang, M. Zhao, L. Zhang, Chem. Commun. 48 (2012) 2540–2542.
[84] Z. Miao, S. Jiang, M. Ding, S. Sun, Y. Ma, M.R. Younis, G. He, J. Wang, J. Lin, Z.
[34] W. Zhang, S. Hu, J.-J. Yin, W. He, W. Lu, M. Ma, N. Gu, Y. Zhang, J. Am. Chem.
Cao, P. Huang, Z. Zha, Nano Lett. 20 (2020) 3079–3089.
Soc. 138 (2016) 5860–5865.
[85] D. Yu, M. Ma, Z. Liu, Z. Pi, X. Du, J. Ren, X. Qu, Biomaterials 255 (2020).
[35] S. Grundner, M.A.C. Markovits, G. Li, M. Tromp, E.A. Pidko, E.J.M. Hensen, A.
[86] A. Sood, M.S. Granick, N.L. Tomaselli, Advances in wound care 3 (2014) 511–529.
Jentys, M. Sanchez-Sanchez, J.A. Lercher, Nat. Commun. 6 (2015).
[87] Y. Guan, M. Li, K. Dong, N. Gao, J. Ren, Y. Zheng, X. Qu, Biomaterials 98 (2016)
[36] Q. Wang, H. Wei, Z. Zhang, E. Wang, S. Dong, Trac-Trends in Analytical
92–102.
Chemistry 105 (2018) 218–224.
[88] N. Gao, K. Dong, A. Zhao, H. Sun, Y. Wang, J. Ren, X. Qu, Nano Res. 9 (2016)
[37] L. Wang, S. Li, X. Zhang, Y. Huang, Talanta 216 (2020).
1079–1090.
[38] X. Zhang, A. Yuan, X. Mao, Q. Chen, Y. Huang, Sensors and Actuators B-
[89] C. Ren, D. Li, Q. Zhou, X. Hu, Biomaterials 232 (2020).
Chemical 299 (2019).
[90] K. Zhang, M. Tu, W. Gao, X. Cai, F. Song, Z. Chen, Q. Zhang, J. Wang, C. Jin, J. Shi,
[39] Z. Jin, G. Xu, Y. Niu, X. Ding, Y. Han, W. Kong, Y. Fang, H. Niu, Y. Xu, J. Mater.
X. Yang, Y. Zhu, W. Gu, B. Hu, Y. Zheng, H. Zhang, M. Tian, Nano Lett. 19 (2019)
Chem. B 8 (2020) 3513–3518.
2812–2823.
[40] X.J. Yang, R.S. Li, C.M. Li, Y.F. Li, C.Z. Huang, Talanta 215 (2020).
[91] X. Mu, H. He, J. Wang, W. Long, Q. Li, H. Liu, Y. Gao, L. Ouyang, Q. Ren, S. Sun, J.
[41] M. Comotti, C. Della Pina, E. Falletta, M. Rossi, Adv. Synth. Catal. 348 (2006)
Wang, J. Yang, Q. Liu, Y. Sun, C. Liu, X.-D. Zhang, W. Hu, Nano Lett. 19 (2019)
313–316.
4527–4534.
[42] R. Ragg, F. Natalio, M.N. Tahir, H. Janssen, A. Kashyap, D. Strand, S. Strand, W.
[92] X. Mu, J. Wang, Y. Li, F. Xu, W. Long, L. Ouyang, H. Liu, Y. Jing, J. Wang, H. Dai,
Tremel, ACS Nano 8 (2014) 5182–5189.
Q. Liu, Y. Sun, C. Liu, X.-D. Zhang, ACS Nano 13 (2019) 1870–1884.
[43] J. Li, W. Liu, X. Wu, X. Gao, Biomaterials 48 (2015) 37–44.

14
W. Yang, X. Yang, L. Zhu et al. Coordination Chemistry Reviews 448 (2021) 214170

[93] S. Gao, H. Lin, H. Zhang, H. Yao, Y. Chen, J. Shi, Adv. Sci. 6 (2019). [119] S.K. Maji, S. Yu, K. Chung, M.S. Ramasamy, J.W. Lim, J. Wang, H. Lee, D.H. Kim,
[94] H. Lin, Y. Chen, J. Shi, Chem. Soc. Rev. 47 (2018) 1938–1958. ACS Appl. Mater. Interfaces 10 (2018) 42068–42076.
[95] G. Yang, L. Xu, Y. Chao, J. Xu, X. Sun, Y. Wu, R. Peng, Z. Liu, Nat. Commun. 8 [120] Y. Hao, Y. Liu, Y. Wu, N. Tao, D. Lou, J. Li, X. Sun, Y.-N. Liu, Biomater. Sci. 8
(2017). (2020) 1830–1839.
[96] D. Wang, H. Wu, S.Z.F. Phua, G. Yang, W.Q. Lim, L. Gu, C. Qian, H. Wang, Z. Guo, [121] S. Li, L. Shang, B. Xu, S. Wang, K. Gu, Q. Wu, Y. Sun, Q. Zhang, H. Yang, F. Zhang,
H. Chen, Y. Zhao, Nat. Commun. 11 (2020). L. Gu, T. Zhang, H. Liu, Angewandte Chemie-International Edition 58 (2019)
[97] L. Fan, X. Xu, C. Zhu, J. Han, L. Gao, J. Xi, R. Guo, ACS Appl. Mater. Interfaces 10 12624–12631.
(2018) 4502–4511. [122] X. Zhong, X. Wang, L. Cheng, Y.A. Tang, G. Zhan, F. Gong, R. Zhang, J. Hu, Z. Liu,
[98] S. Liang, X. Deng, G. Xu, X. Xiao, M. Wang, X. Guo, P.A. Ma, Z. Cheng, D. Zhang, X. Yang, Adv. Funct. Mater. 30 (2020).
J. Lin, Adv. Funct. Mater. 30 (2020). [123] Y. Chen, Z.-H. Li, J.-J. Hu, S.-Y. Peng, L. Rong, Y. Sun, X.-Z. Zhang, Nanoscale
[99] L. Li, Z. Yang, W. Fan, L. He, C. Cui, J. Zou, W. Tang, O. Jacobson, Z. Wang, G. Niu, Horiz. 5 (2020) 283–293.
S. Hu, X. Chen, Adv. Funct. Mater. 30 (2020). [124] F. Liu, L. Lin, Y. Zhang, Y. Wang, S. Sheng, C. Xu, H. Tian, X. Chen, Adv. Mater.
[100] C. Fang, Z. Deng, G. Cao, Q. Chu, Y. Wu, X. Li, X. Peng, G. Han, Adv. Funct. 31 (2019).
Mater. 30 (2020). [125] X. Yang, Y. Yang, F. Gao, J.-J. Wei, C.-G. Qian, M.-J. Sun, Nano Lett. 19 (2019)
[101] Z. Wang, Y. Zhang, E. Ju, Z. Liu, F. Cao, Z. Chen, J. Ren, X. Qu, Nat. Commun. 9 4334–4342.
(2018). [126] S. Liang, X. Deng, Y. Chang, C. Sun, S. Shao, Z. Xie, X. Xiao, P.A. Ma, H. Zhang, Z.
[102] A. Zhang, Q. Zhang, G. Alfranca, S. Pan, Z. Huang, J. Cheng, Q. Ma, J. Song, Y. Cheng, J. Lin, Nano Lett. 19 (2019) 4134–4145.
Pan, J. Ni, L. Ma, D. Cui, Nano Res. 13 (2020) 160–172. [127] C. Liu, L. Luo, L. Zeng, J. Xing, Y. Xia, S. Sun, L. Zhang, Z. Yu, J. Yao, Z. Yu, O.U.
[103] D.-W. Zheng, Q. Lei, J.-Y. Zhu, J.-X. Fan, C.-X. Li, C. Li, Z. Xu, S.-X. Cheng, X.-Z. Akakuru, M. Saeed, A. Wu, Small 14 (2018).
Zhang, Nano Lett. 17 (2017) 284–291. [128] L. Feng, B. Liu, R. Xie, D. Wang, C. Qian, W. Zhou, J. Liu, D. Jana, P. Yang, Y.
[104] P.A. Ma, H. Xiao, C. Yu, J. Liu, Z. Cheng, H. Song, X. Zhang, C. Li, J. Wang, Z. Gu, J. Zhao, Adv. Funct. Mater. (2020).
Lin, Nano Lett. 17 (2017) 928–937. [129] W. Zhen, Y. Liu, W. Wang, M. Zhang, W. Hu, X. Jia, C. Wang, X. Jiang,
[105] Y. Chen, H. Zhong, J. Wang, X. Wan, Y. Li, W. Pan, N. Li, B. Tang, Chem. Sci. 10 Angewandte Chemie-International Edition 59 (2020) 9491–9497.
(2019) 5773–5778. [130] C. Wei, Y. Liu, X. Zhu, X. Chen, Y. Zhou, G. Yuan, Y. Gong, J. Liu, Biomaterials
[106] L. Feng, Z. Dong, C. Liang, M. Chen, D. Tao, L. Cheng, K. Yang, Z. Liu, 238 (2020).
Biomaterials 181 (2018) 81–91. [131] J. Wang, L. Fang, P. Li, L. Ma, W. Na, C. Cheng, Y. Gu, D. Deng, Nano-Micro
[107] Y. Li, K.-H. Yun, H. Lee, S.-H. Goh, Y.-G. Suh, Y. Choi, Biomaterials 197 (2019) Letters 11 (2019).
12–19. [132] Y. Zhang, P. Sun, L. Zhang, Z. Wang, F. Wang, K. Dong, Z. Liu, J. Ren, X. Qu, Adv.
[108] P. Hu, T. Wu, W. Fan, L. Chen, Y. Liu, D. Ni, W. Bu, J. Shi, Biomaterials 141 Funct. Mater. 29 (2019).
(2017) 86–95. [133] Z. Wang, K. Dong, Z. Liu, Y. Zhang, Z. Chen, H. Sun, J. Ren, X. Qu, Biomaterials
[109] Y.-C. Ma, Y.-H. Zhu, X.-F. Tang, L.-F. Hang, W. Jiang, M. Li, M.I. Khan, Y.-Z. You, 113 (2017) 145–157.
Y.-C. Wang, Biomater. Sci. 7 (2019) 2740–2748. [134] Z. Liu, F. Wang, J. Ren, X. Qu, Biomaterials 208 (2019) 21–31.
[110] Y. Zhang, F. Wang, C. Liu, Z. Wang, L. Kang, Y. Huang, K. Dong, J. Ren, X. Qu, [135] J. Xi, G. Wei, L. An, Z. Xu, Z. Xu, L. Fan, L. Gao, Nano Lett. 19 (2019) 7645–7654.
ACS Nano 12 (2018) 651–661. [136] J. Shan, X. Li, K. Yang, W. Xiu, Q. Wen, Y. Zhang, L. Yuwen, L. Weng, Z. Teng, L.
[111] F. Gao, J. Wu, H. Gao, X. Hu, L. Liu, A.C. Midgley, Q. Liu, Z. Sun, Y. Liu, D. Ding, Y. Wang, ACS Nano 13 (2019) 13797–13808.
Wang, D. Kong, X. Huang, Biomaterials 230 (2020). [137] Y. Sang, W. Li, H. Liu, L. Zhang, H. Wang, Z. Liu, J. Ren, X. Qu, Adv. Funct. Mater.
[112] H. Wu, L. Liu, L. Song, M. Ma, N. Gu, Y. Zhang, ACS Nano 13 (2019) 14013– 29 (2019).
14023. [138] L. Wang, Y. Min, D. Xu, F. Yu, W. Zhou, A. Cuschieri, Chem. Commun. 50
[113] S.K. Maji, A.K. Mandal, N. Kim Truc, P. Borah, Y. Zhao, ACS Appl. Mater. (2014) 11147–11150.
Interfaces 7 (2015) 9807–9816. [139] F. Xiong, H. Wang, Y. Feng, Y. Li, X. Hua, X. Pang, S. Zhang, L. Song, Y. Zhang, N.
[114] K. Fan, J. Xi, L. Fan, P. Wang, C. Zhu, Y. Tang, X. Xu, M. Liang, B. Jiang, X. Yan, L. Gu, Sci. Rep. 5 (2015).
Gao, Nat. Commun. 9 (2018). [140] T. Qin, R. Ma, Y. Yin, X. Miao, S. Chen, K. Fan, J. Xi, Q. Liu, Y. Gu, Y. Yin, J. Hu, X.
[115] M. Huo, L. Wang, Y. Chen, J. Shi, Nat. Commun. 8 (2017). Liu, D. Peng, L. Gao, Theranostics 9 (2019) 6920–6935.
[116] Z. Wang, G. Li, Y. Gao, Y. Yu, P. Yang, B. Li, X. Wang, J. Liu, K. Chen, J. Liu, W. [141] M.V. Liga, S.J. Maguire-Boyle, H.R. Jafry, A.R. Barron, Q. Li, Environ. Sci.
Wang, Chem. Eng. J. 404 (2020) 125574. Technol. 47 (2013) 6463–6470.
[117] Z. Ma, L. Wu, K. Han, H. Han, Nanoscale Horiz. 4 (2019) 1124–1131. [142] J. Xi, R. Zhang, L. Wang, W. Xu, Q. Liang, J. Li, J. Jiang, Y. Yang, X. Yan, K. Fan, L.
[118] S. Dong, Y. Dong, T. Jia, S. Liu, J. Liu, D. Yang, F. He, S. Gai, P. Yang, J. Lin, Adv. Gao, Adv. Funct. Mater. 31 (2021).
Mater. 32 (2020). [143] S. Scott, H. Zhao, A. Dey, T.B. Gunnoe, ACS Catal. 10 (2020) 14315–14317.

15

You might also like