Back to Journals » Journal of Inflammation Research » Volume 15

Nanozymes in the Treatment of Diseases Caused by Excessive Reactive Oxygen Specie

Authors Liang S, Tian X, Wang C 

Received 21 July 2022

Accepted for publication 11 October 2022

Published 15 November 2022 Volume 2022:15 Pages 6307—6328

DOI https://doi.org/10.2147/JIR.S383239

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Professor Ning Quan



Shufeng Liang,1,2 Xin Tian,3 Chunyan Wang4

1Department of Molecular Biology, Shanxi Province Cancer Hospital/Shanxi Hospital, Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, People’s Republic of China; 2Institute of Environmental Sciences, Shanxi University, Taiyuan, People’s Republic of China; 3State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, People’s Republic of China; 4Department of Transfusion, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, People’s Republic of China

Correspondence: Xin Tian; Chunyan Wang, Department of Transfusion, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, No 3 Zhigongxin Street, Xinghualing, Taiyuan, Shanxi, People’s Republic of China, Tel +86 15110313926, Email [email protected]; [email protected]

Abstract: Excessive reactive oxygen species (ROS) may generate deleterious effects on biomolecules, such as DNA damage, protein oxidation and lipid peroxidation, causing cell and tissue damage and eventually leading to the pathogenesis of diseases, such as neurodegenerative diseases, ischemia/reperfusion ((I/R)) injury, and inflammatory diseases. Therefore, the modulation of ROS can be an efficient means to relieve the aforementioned diseases. Several studies have verified that antioxidants such as Mitoquinone (a mitochondrial-targeted coenzyme Q10 derivative) can scavenge ROS and attenuate related diseases. Nanozymes, defined as nanomaterials with intrinsic enzyme-like properties that also possess antioxidant properties, are hence expected to be promising alternatives for the treatment of ROS-related diseases. This review introduces the types of nanozymes with inherent antioxidant activities, elaborates on various strategies (eg, controlling the size or shape of nanozymes, regulating the composition of nanozymes and environmental factors) for modulating their catalytic activities, and summarizes their performances in treating ROS-induced diseases.

Keywords: nanozymes, enzyme-mimic, oxidative stress, antioxidant, ROS-related diseases

Introduction

Reactive oxygen species (ROS) contain various reactive molecules and free radicals (species with unpaired electrons) derived from molecular oxygen, such as hydrogen peroxide (H2O2), superoxide (O2·−) and hydroxyl radicals (·OH).1–3 ROS are produced by a cell converting oxygen into oxygen radicals during physiological processes in biological systems and play roles as signaling molecules in cells, including activating cell signaling cascades, inducing apoptosis, and influencing gene expression.4–7

However, oxidative stress occurs when ROS generation increases or antioxidant defense systems become weaker or both, which means unquenched ROS may trigger further reactions.5,8,9 In turn, oxidative stress destroys biomacromolecules and plays a part in a series of pathologies, including neurodegeneration, inflammation, atherosclerosis, diabetes, and aging.4,10–12 As massive ROS production and associated oxidative damage are also identified as important causative factors for diseases such as I/R, researchers speculated that scavenging excessive ROS might be a viable method for oxidative stress-related diseases in the future.13,14

Nanozymes are inorganic nanomaterials that mimic enzyme activities.15 Since ferromagnetic nanoparticles (NPs) were detected to possess enzyme-like activity for the first time,16 various nanomaterials have been developed with peroxidase-like, oxidase-like, catalase-like and superoxide dismutase (SOD)-like activities and are widely used in biomedicine fields such as biosensing, disease diagnosis and treatment.17,18 Furthermore, as Gao and Yan et al concluded, nanozymes should be consistent with the enzyme kinetics and catalytic mechanism of natural enzymes, such as the Michaelis–Menten equation. As a substitute for natural enzymes, even compared to other artificial enzymes, nanozymes are more prominent in their stability to pH and temperature, low production cost, ease of synthesis in large quantities, and reusability.19,20 In addition, the unique physicochemical properties of nanomaterials endow them with a variety of functions and provide various methods for rational design.21 By virtue of enzyme-like activities, especially SOD-like and catalase-like activities, nanozymes are expected to eliminate intracellular ROS and have therapeutic effects in ROS-related damages or diseases.

In this review, we focus on the catalytic activity of antioxidant nanozymes and their significant research progress in the treatment of excessive ROS-caused diseases. First, nanozymes that show antioxidant activity, including carbon-based, metal-based, and metal oxide-based nanozymes, are summarized. Second, strategies for modulating nanozymes to elevate their ROS-quenching capacities are outlined. Third, the applications of nanozymes in relevant treatments are introduced. We hope that this review will provide an overview of antioxidant nanozymes in the ROS-induced disease treatment field, as well as inspiration for future clinical applications.

Antioxidant Nanozymes

Here, we introduce the representative types of antioxidant nanozymes, which have been reported as antioxidants in recent years in the following section (Table 1).

Table 1 Representative Antioxidant Nanozymes and Their Applications

Carbon-Based Nanozymes

Carbon-based nanomaterials have outstanding physicochemical properties, such as electrical, thermal, and optical properties, which make them promising materials for biomedical applications.22 They contain fullerene, graphene nanosheets, graphene oxide (GO), graphene quantum dots (GQDs), and carbon nanotubes (CNTs), which can be divided into zero-dimensional nanomaterials (fullerene, carbon dots, GQDs), one-dimensional nanomaterials (CNTs), two-dimensional nanomaterials (graphene), and three-dimensional nanomaterials (nanodiamonds) based on the number of dimensions exceeding the nanoscale (100 nm).23

Fullerenes are carbon spheres, in which C-atoms are sp2-hybridized carbon atoms, and presently are used for various applications in nanomedicine.24,25 The existence of abundant conjugated double bonds and low lying lowest unoccupied molecular orbital endow them the ability to scavenge radical species.26 With the abovementioned characteristics, fullerenes show antioxidant activity in dealing with O2·−, ·OH, and H2O2, and are called “free radical sponges”.27–29

Through inhibiting cellular ROS levels, fullerenes could suppress proinflammatory cytokine release in synovial inflammation-related cells in vitro, and alleviate synovitis and joint destruction significantly in vivo.30 Fullerenol belongs to fullerene derivatives and exhibits excellent water solubility. With the intrinsic capability of eliminating ROS, fullerenol/alginate hydrogels were synthesized to protect brown adipose-derived stem cells (BADSCs) against the oxidative stress damage after myocardial infarction (MI) and improve the cardiomyogenic differentiation.31 In MI rat model, the fullerenol/alginate hydrogel seeded with BADSCs reduced infarct size, increased wall thickness, and improved cardiac functions successfully.

CNTs, with hollow and cylindrical structure, are formed by rolled graphene sheets. Single-walled CNT consist of one graphene sheet, while multiwalled carbon nanotubes (MWCNTs) consist of multiple single graphene layers.32 For the first time, Fubini et al reported MWCNTs exhibited a remarkable ROS scavenging capacity, including ·OH and O2·−.33 The excellent electron affinity of the molecular orbitals or electronic bands of the carbon atoms in the nanotube framework was conductive to the scavenging reaction.

Graphene is a flat-like layer of densely packed sp2 carbon atoms. For graphene-based nanomaterials, Akhavan et al had summarized that the electron density, chemical composition, sp2-hybridized carbon content and chemical properties were responsible for their ROS-scavenging abilities.13 Just as Hurt et al reported that a series of graphene-based materials showed ·OH scavenging activities.34 Among them, few layer graphene exhibited the best performance, followed by reduced graphene oxide (rGO) and GO. The activity order was in inverse to their total surface area. Thus, the main antioxidant activities of graphene-based materials were ascribed to pristine sp2 carbon domains rather than oxygen-containing functional groups.

In virtue of the antioxidant activities, graphene-based nanomaterials are used for medical treatment applications, such as neurotoxicity and cardiac repair.35,36

GOQDS could decompose H2O2 to H2O and O2, thus verifying their catalase-like activity.36 Pretreatment with GOQDS in 1-methyl-4-phenyl-pyridinium ion (MPP+) induced PC12 cells and the brains of larval zebrafish could prevent overproduction of ROS and provide neuroprotection.35

GOs was modified with polyethylenimine and folic acid−polyethylene glycol (PEG) to form a macrophage-targeting/polarizing GO complex (MGC).36 The as-prepared material was verified to eliminate ·OH which invoked cardiac failure after MI. In turn, inflammatory biomarkers, such as tumor necrosis factor-α (TNF-α), were less produced during the inflammatory phase. Additionally, DNA-functionalized MGC promoted the polarization of M1 to M2 macrophages and accelerated secretion of cardiac repair-favorable cytokines. Collectively, this compound played dual roles to attenuate inflammation and improve heart function.

Metal and Metal Oxide-Based Nanozymes

Iron Based

Under different reaction conditions, iron oxide nanozymes (including Fe3O4 and Fe2O3) exhibited either peroxidase-like or catalase-like activities.16,37,38 These activities conferred iron oxide nanozymes the abilities to scavenge ROS under appropriate conditions.

Intrigued by the discovery of the catalase-like activity of iron oxide NPs, researchers have explored how these nanoparticles dealt with oxidative stress in aging or neurodegeneration.39 Fe3O4 NPs alleviated a cell model of Parkinson’s disease (PD) death and reduced the levels of α-synuclein and cleaved caspase-3, which are presynaptic neuronal proteins tightly linked to PD pathogenesis and apoptosis markers, respectively. Finally, the effects of dietary Fe3O4 NPs on aging and the Alzheimer’s disease (AD) model of Drosophila were tested, and Fe3O4 NPs were effective in increasing life span, antiaging, promoting locomotor activity and reducing apoptosis in these disease models.

Combining nanozymes and natural antioxidants has also been used as free radical scavengers, as shown in Figure 1A.40 As Zhao et al designed, Fe3O4 NPs and the polyphenol tannic acid (TA), which is a natural antioxidant, were combined into Fe3O4@TAn nanoflowers (NFs), as shown in Figure 1B. The Fe3O4 @ TAn NFs could mimic peroxidase, catalase, and SOD. Through the complementary action of Fe3O4 and TA, the as-designed nanomaterial exhibited improved and broad-spectrum ROS eliminating activity. Additionally, the nanoflower morphology of the Fe3O4@TAn NFs made them fully exposed to oxidative stress conditions, thus contributing to the excellent activity. Owing to a remarkable multiple ROS scavenging activity, the Fe3O4@Tan NFs were employed to treat mouse endotoxemia with excellent results.

Figure 1 Iron oxide nanozyme and nonenzymatic antioxidant constructed a nanoplatform to mimic the antioxidant defense system. (A) The antioxidant ability of Fe3O4 @ TAn NFs. (B) Schematic illustration of the petal structure of Fe3O4@TA1.0 NFs. Reproduced with permission from Wei Z, Wang L, Tang C et al. Metal-phenolic networks nanoplatform to mimic antioxidant defense system for broad-spectrum radical eliminating and endotoxemia treatment. Adv Funct Mater. 2020;30(49). Copyright (2020) John Wiley and Sons.40

In addition, Prussian blue nanoparticles (PBNPs), as Fe-containing nanomaterials with good biosafety, were also found to possess multienzyme activities. The different redox potentials of PB, Prussian white (PW), Berlin green (BG), and Prussian yellow (PY) conferred these materials tremendous abilities to transfer electrons.41–44 Dong et al proposed that PBNPs represented the first example of a bioinspired nanozyme.45 They looked for the active site of PBNPs and concluded that N-coordinated Fe units (FeNx (x=4–6)) with intrinsic heme-like structures endowed PBNPs with peroxidase-like activity.

Thus, PBNPs could efficiently control ROS-induced cell damage and treat diseases such as mouse liver inflammation and injury, colitis, acute pancreatitis, cutaneous wound healing and ischemic stroke.42,46–50

Gold-Based

Au NPs were demonstrated to decompose H2O2 to O2 at alkaline pH and scavenge superoxide at acidic pH.51 The activities made Au NPs potential catalase and SOD mimetics. In addition, Guo and Lu et al showed that Au NPs played important roles in ameliorating neurologic deficits and infarction volumes and protecting the cerebra from I/R injury in rats via the regulation of inflammatory and immune responses.52

To eliminate the toxicity induced by ·OH produced by the catalytic action of Au nanoclusters (AuNCs), polymeric 3°-amine was added to Au NCs to simultaneously restrain the generation of ·OH and retain their catalase-like activity. The derived AuNCs-NH2 acted as H2O2 scavengers, thus protecting primary mouse neurons against oxidative stress.53

Cerium Oxide (Ceria)-Based

As early as 2006, Schubert et al proposed that cerium oxide NPs could protect nerve cells from oxidative stress through the ability to limit the amount of ROS.54

As relevant studies developed, cerium oxide NPs were proven to have excellent multiple enzyme-mimicking properties due to their Ce3+/Ce4+ oxidation state and the presence of oxygen vacancies.55–57

Numerous studies are performed on nanoceria and its biomedical application. CeO2 NPs and related materials have been widely used to protect cells that are destroyed by excessive ROS, such as nerve cells, macrophages, cardiac progenitor cells, human breast cells, fibrosarcoma cells and human hepatic cells.54,57–61 Furthermore, they may serve as a novel therapy for neurodegenerative diseases, depression, ischemic stroke, autoimmune diseases, hepatic injury, sepsis, psoriasis, inflammatory bowel disease (IBD), periodontitis, and acute gout.62–77 Based on the excellent ROS eliminating capabilities of CeO2, researchers have explored various kinds of targeted methods to address the aforementioned diseases.

During the treatment of ROS-related diseases, CeO2 also plays roles in downregulating proinflammatory cytokine levels, upregulating anti-inflammatory cytokine levels, inhibiting the ROS–NFκB pathway, etc.69,73,77

Copper Based

Cu-based nanozymes have been widely used to eliminate ROS and improve ROS-related diseases, including PD, acute liver injury, acute kidney injury, and wound healing.20,78,79

Deng’s group considered that large surface-to-volume ratios would confer high catalytic activity, so they made ultrasmall Cu5.4O NPs (Cu5.4O USNPs), the average hydrodynamic diameter of which was as small as 4.5 nm.20 The as-designed nanoparticles were verified to have multiple enzyme-mimicking activities, including catalase, glutathione peroxidase (GPx), SOD and excellent broad-spectrum ROS scavenging ability. The reason for the name “Cu5.4O USNPs” was because the materials were a mixture of Cu and Cu2O NPs.

Manganese-Based

Lv et al first discovered that manganese dioxide (MnO2) nanoparticles possessed enzyme-mimicking activities, which was further demonstrated in another study in 2017.80,81

Recently, Mn3O4 nanozymes were also confirmed to possess antioxidant enzyme-like properties, and when compared with the most widely used CeO2, researchers found that Mn3O4 nanozymes were more effective in terms of ROS elimination.82,83 The mixed valance states of Mn2+/Mn3+, tolerance to oxidation, large surface area and extremely large pore size of Mn3O4 nanozymes contributed to their outstanding multienzyme activity.82

Mn3O4 nanozymes could provide efficient cytoprotection in SHSY-5Y, which is a human cell line model for the PD phenotype, and protect mice from phorbol 12-myristate 13-acetate (PMA)-induced ear inflammation.82,84 Furthermore, Mn3O4 nanozymes were proven to protect cells suffering from oxidative stress.83 The related expression of various stress markers and antioxidant enzymes in the presence of Mn3O4 nanozymes was also detected. Conclusively, Mn3O4 nanozymes complemented the antioxidant machinery and rescued the cells under oxidative stress independent of the endogenous antioxidant machinery.

Other Metal-Based Nanozymes

Qu and his cooperators synthesized GO-Se nanocomposites and Se@PDA (polydopamine) nanozymes, which both showed GPx-like properties.85,86 Se@PDA nanocomposites showed excellent antioxidant activity, which was ascribed to the enzyme-like effect of the Se component and the nonenzymatic antioxidant ability of pDA.86 By decomposing excessive intracellular H2O2, the nanocomposites successfully alleviated neumonia in the mouse model. To achieve higher GPx-like activity, mesoporous selenium NPs (MSeNPs) with a high specific surface were synthesized.87 Then, hyaluronic acid (HA), which could specifically attach to CD44 on the surface of macrophages, was assembled on the MSe NPs to facilitate the accumulation of the as-designed nanozyme at the site of inflammation. Taken together, treatment with MSe-HANPs successfully relieved local inflammation and sepsis.

Metal Organic Frameworks (MOFs) Based

MOFs are a class of supramolecular coordination complexes with metal ions/clusters bridged with organic linkers by coordination bonds.88,89 They also have natural enzyme-mimicking properties. For example, MOF-derived copper nanoparticle@carbon nanocomposites, iron(III)-based MOFs (MIL-68 and MIL-100) (MIL=Material Institute of Lavoisier), PA-Tb-Cu MOF, Cu2+ ion-modified UiO-type MOF NPs (UiO = University of Oslo), MOF-808 and zirconium-ferriporphyrin (Zr-FeP) MOF were verified to possess peroxidase-like activities.90–95

Tb3+ as a metal ion and a boronic acid ligand as a linker constituted a luminescent Tb-MOF.96 The boronic acid groups of Tb-MOF reacted with ROS through the rapid nucleophilic addition reaction, which endowed the MOF with ROS quenching capability. Further investigation proved that Tb-MOF effectively scavenges exogenous or endogenous ROS in living cells, including HeLa cells and RAW 264.7 cells.

According to the size and active sites of natural SOD, Qu and Ren et al designed ultrasmall and bioinspire Cu-TCPP MOF nanodots (CTMDs), the average size of which was less than 5 nm.97 The Cu active sites of CTMDs showed a similar coordination of N and O atoms to that of SOD, while the ordered channels were similar to substrate channels that could accumulate superoxide, thus contributing to the potent SOD-like activities of CTMDS. Additionally, CTMDs could catalyze H2O2 into H2O with glutathione (GSH), which mimicked the activity of GPx. Combining SOD-like and GPx-like activity, CTMDs could effectively destroy O2·− and H2O2. As demonstrated in vivo, CTMDs could alleviate endotoxemia efficiently by eliminating ROS and reducing systemic inflammation.

Others

Melanin and Its Derivatives

Nanomaterials derived from natural organisms have been used in biological and medical applications due to their biocompatibility and biodegradability. As a natural biopolymer, melanin is distributed in the human body, such as skin, eye, brain, and hair.98,99 It showed scavenging activity toward ROS and protected skin from UV damage.100 In addition, PDA NPs, as the most typical synthetic melanin, are produced by the oxidative polymerization of dopamine. The reductive functional groups of PDA NPs, including catechol and imine, endow them with robust potential in scavenging ROS.98,101,102

Ultrasmall Mn2+-chelated melanin NPs (MMPPs) were also verified to exhibit multiple ROS scavenging activities and used to treat murine acute kidney injury.103 Considering the characteristics of targeted disease, MMPPs were designed with an ultrasmall hydrodynamic size, good physiological stability, and stable radiolabeling performance. Thus, they could preferentially concentrate in the kidney and show a robust antioxidative protection response with few side effects.

Single Atom Nanozymes (SAzymes)

SAzymes contain single metal atoms on various supports, which can maximize the utilization of metal atoms, thus achieving high activity and selectivity and shrinking the difference between nanozymes and natural enzymes.104–106

To obtain an efficient antioxidant therapy for sepsis, Qu and Ren et al synthesized a SAzyme, Co/PMCS, characterized by atomically dispersed coordinatively unsaturated active Co porphyrin centers. The SAzymes could mimic SOD, catalase and GPx, thus obliterating multiple RONS.107 In another experiment, the researchers tried to develop a clinically applicable bandage based on a single-atom Pt/CeO2 nanozyme for brain trauma (Figure 2). 108 The nanozyme had high and persistent catalytic activity, which could fight against oxidative stress, decrease neuroinflammation and improve neurotrauma. The high catalytic activity was ascribed to the 2% lattice expansion induced by Pt single atoms dispersed on the CeO2 (111) matrix, thus decreasing the binding energy of the chemical reactions. Moreover, Pt/CeO2 avoided obvious decay due to continuous exposure to oxygen and maintained catalytic activity for up to 30 days, which was ascribed to its sustained electron-donating ability via the Mars–van Krevelen reaction.

Figure 2 Schematic diagram of the nanozyme-based bandage to treat brain trauma. Reprinted with permission from Yan R, Sun S, Yang J et al. Nanozyme-based bandage with single-atom catalysis for brain trauma. ACS nano. 2019;13(10):11,552–11,560. Copyright (2019) American Chemical Society.108

Regulatory Factors for Nanozyme Activity

Elevating the catalytic properties of nanozymes is a topic of common concern. Studies focused on activity regulation have been performed in the past few years. The following are several important factors that influence the antioxidant activity of nanozymes.

Physicochemical Properties

Size

Generally, smaller nanozymes show better catalytic activity than larger nanozymes. As the size decreases, nanozymes obtain a higher surface-to-volume ratio, expose more active sites, and have a larger surface energy.21,109 However, there are some special cases; for example, the catalytic performance of Pd-Ir NPs increased with increasing size.110 To exclude other physicochemical factors, the researchers synthesized nanomaterials with the same morphologies and surface structures except for size. Catalytic activity parameters, such as Kcat (catalytic rate constant) and Kcat/S (area-specific Kcat), were compared. As the size of the Pd-Ir NPs increased, Kcat continuously increased by nearly 12-fold. However, the Kcat/S values for different sizes of Pd-Ir NPs were similar. Combining the results, the conclusion was that the catalytic activity of the NPs was mainly ascribed to the catalytic surface areas, which could promote the interaction of individual nanoparticles and more substrates.

Shape and Morphology

The catalytic activity of nanomaterials also depends on shape and morphology.111 For instance, Ge et al speculated that antioxidant activity was structure-dependent for the carbon nanomaterial family.29 They evaluated the radical scavenging activities of buckyball-shaped fullerene derivatives and sheet-shaped GQDs by electron spin resonance (ESR) measurements. After comparison, the activities were in the order C70>C60>GQD. By theoretical calculations of the Gibbs free energies of reactions (Gr), the Gr’s of C60 and C70 were much lower than that of GQD, suggesting that larger radical scavenging activity depends on the lower Gr values.

More deeply, scientists revealed the prominent effect of surface facets in determining the enzyme-like activity of Pd nanocrystals.112 They discovered that Pd octahedrons enclosed by 111 facets had greater enzyme-mimic activity than Pd nanocubes enclosed by 100 facets through ESR experiments (Figure 3A). Taking advantage of theoretical simulations, the ROS clearance that reacted on Pd 111 and 100 facets was calculated. The reaction energy (Er) of the rate-limiting step could be used as the descriptor of the enzyme activity of the metal. As shown in Figure 3B, the Er values of H2O2 and O2·− on the 111 facet were both stronger than those on the 100 facet, which indicated greater H2O2 and O2·− scavenging activities.

Figure 3 The regulation of nanozyme activity based on physicochemical properties. (A) Lower surface energy {111}-faceted Pd octahedrons and higher surface energy {100}-faceted Pd nanocubes. (B) Lowest-energy adsorption structures and reaction energies (in eV) for the reactions on structures having either Pd 111 or 100 facets. Reprinted with permission from Ge C, Fang G, Shen X et al. Facet energy versus enzyme-like activities: the unexpected protection of palladium nanocrystals against oxidative damage. ACS Nano. 2016;10(11):10,436–10,445. Copyright (2016) American Chemical Society.112 (C) STEM, energy dispersive X-ray spectroscopy (EDS) mapping and electron tomographic reconstruction of CuxO NCs. Reprinted with permission from Hao C, Qu A, Xu L et al. Chiral molecule-mediated porous CuxO nanoparticle clusters with antioxidation activity for ameliorating Parkinson’s disease. J Am Chem Soc. 2019;141(2):1091–1099. Copyright (2019) American Chemical Society.79 Synthetic process (D) and catalytic performance (E) of the Nanocage. Reprinted with permission from Wang C, Ren G, Yuan B et al. Enhancing enzyme-like activities of Prussian blue analog nanocages by molybdenum doping: toward cytoprotecting and online optical hydrogen sulfide monitoring. Anal Chem. 2020;92(11):7822–7830. Copyright (2020) American Chemical Society.120

Interestingly, Kuang et al prepared a complex of CuO and Cu2O nanoparticle clusters (CuxO NCs) by selecting L-phenylalanine (L-Phe) as the structure-directing agent.79 The Phe ligand was confirmed to take part in determining the CuxO NC structure, which had a uniform diameter and morphology, a larger volume of pores and a larger pore size than those of other materials, such as CuxO-aspartic acid (Asp) (Figure 3C), thus endowing the CuxO NCs with higher catalytic performance.

Composition

Tailoring the composition of nanomaterials, such as doping other elements or forming bimetallic or multimetallic nanocomposites, poses another way to tune the activity of nanozymes.113–118

Hyeon and Lee et al introduced Zr4+ into ceria nanoparticles to increase the Ce3+/Ce4+ ratio and convert Ce4+ to Ce3+ since Ce3+ targets O2·− and ·OH.72 Through investigation, the Ce0.7Zr0.3O2 (7CZ) NPs showed the best ROS-scavenging performance. Additionally, in vivo tests also demonstrated that 7CZ NPs effectively reduced mortality and systemic inflammation in sepsis. By Li doping, the catalytic activities of ZnMn2O4 nanozyme developed from single antioxidant activity to multiple activities, including SOD-like, catalase-like and GPx-like activities, with enhanced performance.119 Through XPS analysis, the molar ratio of Mn4+/Mn3+ increased from 0.56 to 2.29 along with gradual Li doping.

Another doping strategy was adopted to improve the catalytic activity of Prussian blue analog (PBA)-based nanozymes (Figure 3D and E).120 In this study, Ni-Fe bimetal PBA-based nanocages doped by molybdenum-polysulfide (Nanocages) were prepared and were proved to have multiple enzyme-like activities. Doping molybdenum into PBA successfully regulated the size (decreased), shape (from solid to hollow), composition, and electronic structure of the nanocage, thus bringing in more catalytic sites and binding sites.

In addition, the peroxidase-like, catalase-like and SOD-like activity of PtCu nanoalloys was related to the atomic ratio of Pt/Cu.121 At the same time, by changing the atomic ratio of Pt/Cu, the particle size, alloy composition and crystal structure of PtCu nanoalloys were controlled correspondingly.

Surface Coating and Modification

Controlling surface chemistry by surface coating and modification is also an advantageous way to improve catalytic performance because chemical reactions take place mainly on the surface of catalysts.122 Ions (fluoride), small molecules such as amino acids, citrate, Prussian blue, amino acids, and polymers (protein, dextran, DNA) can act as materials for modification.123–128

Yan and Gao et al modified Fe3O4 NPs with histidine (His) residues to mimic the active site of horseradish peroxidase (HRP) and expected hydrogen bond formation between the distal imidazole group and H2O2.129 The bionic design effectively altered the catalytic properties of the Fe3O4 nanozyme.

Specifically, abundant efforts have been made to improve the specificity of nanozymes for chiral substrates. By virtue of the surface coating of amino acids and DNA with chiral structures on nanozymes as chiral ligands, nanozymes can be designed with chiral selectivity.130–133

Natural phenolic antioxidants gave Qu and Ren et al inspiration; they used NaBH4 as the reductant to convert C=O on classical GQDs (c-GQDs) to C–OH and formed GQDs (h-GQDs) with phenol-like groups.134 Through comparison, the antioxidant activity of h-GQDs was at least 5 times that of c-GQDs under the same conditions. By means of experimental research and theoretical calculations, the researchers discovered step by step that phenol-like groups played an important role in the catalytic performance of h-GQDs. In addition, carbonyl groups, which suppress the H-donating ability of the phenol-like group, were removed from h-GQDs, which also promoted the antioxidative activity of h-GQDs.

Form Complex or Hybrid Nanozymes

The construction of different nanozymes may improve their catalytic performance and result in synergistic effects and even multistep cascade reaction activity, thus attracting many intentions in recent years.135–139

A multinanozyme cooperative platform was assembled in which V2O5 nanowires acted as GPX mimics, while MnO2 nanoparticles mimicked SOD and catalase (Figure 4A). 140 V2O5@pDA@MnO2 exhibited excellent ROS depletion performance that could mimic a natural antioxidant enzyme system (Figure 4B). Compared with Se NPs, GO-Se nanocomposites exhibited much higher GPx-like activity.85 The reason might be that GO endows the GO-Se hybrid with a greater surface area and rapid electron transfer capacity.

Figure 4 A multinanozyme cooperative platform to efficiently reduce ROS levels. (A) The synthesis process of V2O5@pDA@MnO2 nanocomposites. (B) The multinanozyme system mimics the intracellular antioxidant enzyme-based defense system. Reprinted with permission from Huang Y, Ren J, Qu X. Nanozymes: classification, catalytic mechanisms, activity regulation, and applications. Chem. Rev. 2019, 119, 6: 4357–4412. Copyright (2019) American Chemical Society.176

Zeolitic imidazolate framework-8 (ZIF-8) was used to encapsulate CeO2 NPs to form CeO2@ZIF-8 and overcame some limitations of CeO2.141 According to the design, ZIF-8 mimicked peroxidase to maintain antioxidant activity and controlled the size, morphology, and surface potential of CeO2, which was beneficial for biological applications.

Environmental Factor

The antioxidant performance of nanozymes is also sensitive to the surrounding environment, such as pH, temperature, and light. The catalytic activity of nanozymes is greatly influenced by pH. As Wei concluded, under acidic conditions, nanozymes showed peroxidase-mimicking activities, while under neutral and alkaline conditions, they exhibited SOD-like and catalase-like properties.21

Application of Nanozymes in Excessive ROS-Induced Diseases Treatment

In this section, we introduce the application of nanozymes in excessive ROS-induced diseases, including neurodegenerative diseases, inflammatory diseases, and stroke et al.

Treatment of Inflammatory Diseases

The accumulation of ROS may be one of the signaling pathways connecting the UPR (unfolded-protein response) and inflammation, thus initiating an inflammatory response.12,142 Excessive inflammation can cause chronic or systemic inflammatory diseases, including cardiovascular disease, atherosclerosis, asthma, cystic fibrosis, and rheumatoid arthritis.143,144

IBD, as a troublesome colorectal inflammatory disease, is also closely related to excessive ROS.145 Current treatment for IBD mainly relies on antibiotics, which will induce problems such as antibiotic resistance. Prussian blue, which has been approved as an antidote by the US Food and Drug Administration (FDA) and with good biosafety, can also act as an artificial nanozyme, effectively scavenging ROS.146 To improve the solubility of PB NPs, PPB NPs were prepared with the help of poly(vinylpyrrolidone) via a one-pot strategy.43 The in vivo experiments showed that through intravenous administration, PPBs targeted the inflamed sites and alleviated colitis in mice; in addition, proinflammatory cytokines were inhibited significantly. Furthermore, to realize oral administration, it was an ingenious design to combine clinically approved montmorillonite (MMT) and CeO2 in targeting the inflamed colon.69 MTT, with negative charges and high tolerance to pH, endowed CeO2@MMT with stability to pH and specific adsorption onto the inflamed colon, which was positively charged based on electrostatic interactions. Thus, the nanozyme could be efficiently delivered through the digestive tract, which made it suitable for oral administration. MMT could simultaneously decrease the systemic absorption of CeO2, thus reducing potential toxicity. Budesonide, a promising drug for IBD, and MnO2 nanozyme, a nanocarrier, when taken together, became more effective and could be taken orally, as Tong et al demonstrated in mouse models.147 The main reasons for selecting hollow MnO2 NPs were as follows: first, they possessed intrinsic enzyme-like properties, which could scavenge ROS; second, they had an adjustable hollow structure, which was conducive to the loading of drugs; and thirdly, they could be modified with dextran sulfate sodium (DSS), which could target activated macrophages. In addition, hollow MnO2 NPs were influenced by the inflammatory microenvironment and were prone to release budesonide, thus fulfilling their role as nanocarriers. Compared with free budesonide, the nanocarrier system exhibited fewer side effects and had a better curative effect on IBD.

The researchers noticed some problems in the therapy of osteoarthritis, such as antioxidants, melatonin and N-acetylcysteine (NAC), were too small to stay long enough in the joint.148 Therefore, they designed appropriately sized dopamine-melanin (DM) NPs, which were 112.5 nm in size, and made the nanomaterials suitable for retention in the joint for a long time. In a rat osteoarthritis model, DM NPs were found to combat damage caused by inflammation and protect chondro by scavenging intracellular RONS and activating antioxidant enzymes by autophagy. Sharma et al chose cartilage that suffered irreversible damage under oxidative stress as the therapeutic target.149 PEG-MnO2 NPs were designed with a size of less than 20 nm and positive surface charge. Due to the small size and zeta potential, the nanoparticles enabled cartilage penetration and chondrocyte uptake in vitro and in vivo. After one week, 75% of the initial signal remained in the joint space, which might be connected with the electrostatic interactions between nanoparticles and anionic cartilage tissue. Combined with the intrinsic ROS scavenging capability of MnO2 NPs, the materials improved chondrocyte viability and extracellular matrix preservation by reducing inflammation-induced oxidative stress in cartilage.

Yang et al pointed out that in periodontal disease suffering from oxidative stress, PDA NPs with strong ROS removal capacity successfully decreased local periodontal inflammation.150 PDA NPs that could satisfy the following conditions, such as robust antioxidative activity, degradability and low toxicity, might act as intelligent treatments for clinical application.

Treatment of Ischemic Stroke

Ischemic stroke, caused by artery thrombi in blood vessels, in turn leads to excessive ROS production and the resulting damaged neurons during the I/R process.151,152

Bioinspired melanin NPs (Me NPs) were employed as a novel antioxidant to treat ischemic stroke disease, as shown in Figure 5. The infarction area of the brain in a murine model was greatly reduced compared to that in the control.153 The melanin NPs not only scavenged ROS, including O2·−, H2O2, and ·OH, but also reduced inflammatory responses by inhibiting the expression of inflammatory mediators and cytokines.

Figure 5 Comparison of the infarction area of 2,3,5-triphenyltetrazolium chloride-stained brain slices from different groups. Reprinted with permission from Liu Y, Ai K, Ji X et al. Comprehensive insights into the multi-antioxidative mechanisms of melanin nanoparticles and their application to protect brain from injury in ischemic stroke. J Am Chem Soc. 2017;139(2):856–862. Copyright (2017) American Chemical Society.153

A stable composite formed by human serum albumin (HSA) bound to the Mn3O4 nanozyme was used to alleviate nervous system injury caused by ischemic stroke reperfusion.154 Owing to HSA, the as-designed nanozyme showed excellent stability, which was important for high bioavailability and prolonged blood circulation time. Moreover, Mn ions were released from HSA-Mn3O4 into bodily circulation and thus promoted SOD2 expression. PEG- and angiopep-2 (ANG)-modified CeO2 nanozyme was loaded with edaravone for the treatment of ischemic stroke.62 PEGylation endowed CeO2 nanozyme with superior characteristics of uniformity, small size, and prolonged blood circulation, while ANG led to effective cross blood–brain barrier (BBB) (through receptor-mediated transcytosis), which solved the problem of maintaining BBB integrity and the high accumulation need of nanozyme in damaged areas (Figure 6). The loaded edaravone and CeO2 core both contributed to the removal of ROS, thus providing an efficient strategy for treating strokes. To solve the problems that exist in nanozyme treatment, such as less nanozyme accumulation in the ischemic brain site, a neutrophil-like cell membrane-coated mesoporous Prussian blue nanozyme (MPBzyme@NCM) was prepared.50 The preparation of the nanomaterials translocated the entire cell membrane to the surface of the nanozyme and realized interaction between inflamed brain microvascular endothelial cells and neutrophils. The operation could help to promote nanozyme to access damaged brain and realize a noninvasive active-targeting therapy for ischemic stroke. PBNPs also polarized microglia from M1 to M2, decreased the accumulation of neutrophils, reduced neuronal apoptosis, and upregulated neurogenesis. This work proposed a new way to treat brain diseases by combining biomembranes and nanozymes.

Figure 6 TPP-CeO2 NPs acted as antioxidants for AD. (A) Formations of oxygen vacancies, cerium(III) and cerium(IV) species in CeO2 NPs. (B) Main roles of components on edaravone-carried and PEG/ANG-conjugated CeO2 NPs (E-A/P-CeO2). (C) Synthetic process for E-A/P-CeO2. (D) ANG-LRP endocytosis of E-A/P-CeO2. Reprinted with permission from Bao Q, Hu P, Xu Y et al. Simultaneous blood-brain barrier crossing and protection for stroke treatment based on edaravone-loaded ceria nanoparticles. ACS Nano. 2018;12(7):6794–6805. Copyright (2018) American Chemical Society.62

Treatment of Neurological Diseases

The common feature of neurodegenerative diseases, such as AD and PD, is a gradual loss of neuron function. More deeply, the deposition of the peptide amyloid-β (Aβ) for plaques and the deposition of the protein tau for neurofibrillary tangles comprise the histopathological hallmarks of AD.155–157 Studies have confirmed that abnormal ROS affect the production and aggregation of Aβ peptides and metal ion homeostasis and cause mitochondrial dysfunction in AD.158 ROS can also contribute to the degeneration of dopamine cells in PD.159

For the treatment of AD, researchers synthesized small and positively charged triphenylphosphonium-conjugated CeO2 NPs (TPP-CeO2 NPs), which could enter mitochondria and destroy ROS to relieve oxidative stress in a 5XFAD transgenic AD mouse model.64 Concretely, the derived nanoparticles mitigated reactive gliosis and maintained mitochondrial morphology. The TPP orientation verified the feasibility of mitochondrial therapeutics against neuroinflammation. Another strategy aimed at the accumulation of Aβ is shown in Figure 7.65 Aβ antibody-conjugated magnetite/CeO2 nanoparticle assemblies (MCNAs) with core/shell structures were synthesized to remove Aβ peptides from blood. When applying an external magnetic field, MCNAs isolated the captured Aβ peptides through magnetic force, while CeO2 NPs in MCNAs scavenged ROS overproduced by the immune response. Collectively, the assemblies reduced the levels of blood Aβ and brain Aβ in 5XFAD transgenic mice and prevented spatial working memory deficits.

Figure 7 Schematic illustration of MCNA synthesis and mechanism. Reprinted with permission from Kim D, Kwon HJ, Hyeon T. Magnetite/ceria nanoparticle assemblies for extracorporeal cleansing of amyloid-beta in Alzheimer’s disease. Adv Mater. 2019;31(19):e1807965. Copyright (2019) John Wiley and Sons.65

Ling and Tian et al exploited a new approach aimed at the tau pathway to deal with AD, involving tau hyperphosphorylation and aggregation of hyperphosphorylated tau.66 As Figure 8 shows, the nanocomposite CeNC/IONC/MSN was formed by modifying CeO2 and iron oxide nanocrystals on mesoporous silica NPs (MSN). Then, T807, which can bind tau protein, was immobilized on its surface; while methylene blue (MB), which can inhibit tau aggregation, was adsorbed into the pores of MSNs. CeO2 scavenged ROS and suppressed tau hyperphosphorylation, while MB inhibited hyperphosphorylated tau aggregation. Through the cooperation of components, the as-nanocomposite successfully protected neuronal survival and ameliorated learning and memory impairments developed in AD.

Figure 8 CeNC/IONC/MSN-T807-MB and its tau-targeted synergistic treatment for AD. (A) Schematic illustration of the progressive fabrication of CeNC/IONC/MSN-T807-MB. (B) Treatment mechanism of the nanocomposite. Reprinted with permission from Chen Q, Du Y, Zhang K et al. Tau-targeted multifunctional nanocomposite for combinational therapy of Alzheimer’s disease. ACS Nano. 2018;12(2):1321–1338. Copyright (2018) American Chemical Society.66

PD is characterized by the accumulation of misfolded α-synuclein (a prion-like protein) into Lewy bodies, which plays a central role in the pathogenesis of PD.160 A concept of proof that used nanozymes to fight against α-synuclein spreading was provided by Mao’s group.121 PtCu nanoalloys (NAs) were prepared and possessed three enzyme-like activities, including peroxidase-like, catalase-like and SOD-like activities. The application of PtCu NAs in vitro or in vivo significantly inhibited α-synuclein preformed fibril (PFF)-induced α-synuclein pathology, cell death, and neuron-to-neuron transmission by scavenging ROS. Moreover, the PtCu NAs strongly inhibited α-synuclein spreading in the PD brain. Zheng and Cai et al considered that inflammasome-mediated pyroptosis may be a therapeutic target for PD and discovered PB NPs as a pyroptosis inhibitor.161 Due to their potent ROS-scavenging activity, PB NPs inhibited the activation of the nucleotide-binding domain and leucine-rich repeat family pyrin domain containing 3 (NLRP3) inflammasome and caspase-1, reduced the cleavage of gasdermin D and the production of inflammatory factors, and effectively suppressed the pyroptosis of microglia in PD cell and mouse models. Although the study reveals a good effect of PB NPs on the treatment of PD, the deeper mechanism in vivo is currently unclear. The pharmacokinetics and long-term toxicity of PB NPs remain an issue of concern.

Treatment of Acute Liver/Kidney Injury

The researchers used Cu5.4O USNPs to treat acute kidney injury and acute liver injury, and verified that Cu5.4O USNPs functioned as antioxidants to eliminate ROS and induced remission of the diseases.20 In the meantime, the ultrafine size of the nanoparticles accelerated renal clearance and conferred high biocompatibility. The therapeutic mechanism involved in acute kidney injury was uncovered in a further study. GSH metabolism, the MAPK signaling pathway, and the TNF signaling pathway, which exacerbated renal injury, were inhibited by Cu5.4O USNPs. The nanoparticles also maintained the expression of antioxidant genes, inhibited excessive proinflammatory factors such as TNF-α and interleukin-1β (IL-1β), and promoted the expression of kidney-associated genes. The Cu5.4O USNPs were reused three times to decompose H2O2. The results showed that the nanomaterials could maintain almost the same catalytic activity as the original solution, indicating that they had good stability and recyclability.

Mo-based polyoxometalate (POM) nanoclusters were synthesized with potent ROS-scavenging activity owing to the dual oxidation states of Mo5+/Mo6+.162 Furthermore, POM nanoclusters acted as new antioxidants for ameliorating acute kidney injury in a murine model. By positron emission tomography (PET) imaging, radionuclide 89Zr-labeled POM nanoclusters were observed to be more likely to accumulate in the injured kidneys due to their ultrasmall hydrodynamic diameter. The treatment effect demonstrated that POM improved the renal function of acute kidney injury mice with excellent safety.

The therapeutic mechanisms of PBNPs were probed in anthracycline-induced liver injury.46 PBNPs activated the Nrf2/ARE pathway to upregulate antioxidative genes, thus reducing oxidative stress in response to liver injury. They also attenuated inflammatory reactions by decreasing the expression levels of myeloperoxidase and F4/80 (a specific indicator for macrophages) in the liver.

Others

The main pathological features of depression, a serious mental disease, are oxidative stress and excessive ROS. Thus, Zheng and Zhang et al considered using CeO2 nanozyme as a novel drug to treat depression.67 To overcome the disadvantage of CeO2, such as its large size, bovine serum albumin (BSA) served as a space constraint to avoid nanoparticle aggregation. The as-designed CeO2@BSA nanoclusters were endowed with an ultrasmall size as small as 2 nm, potent ROS scavenging activity and BBB penetration ability. When used in a depressive model induced by chronic restraint stress, CeO2@BSA decreased depression-like behaviors, relieved neuroinflammation and provided neuroprotection with few side effects.

Conclusion and Future Perspective

An imbalance of ROS leads to oxidative stress and is linked to various diseases, including diabetes, neurodegeneration, and aging.4 Nanozymes with intrinsic antioxidant ability are widely employed to treat ROS-involved diseases.

In this review, we mainly introduce considerable experimental studies that showed satisfactory performances of nanozymes in disease models such as inflammation, neurodegenerative diseases and stroke. In addition, we summarize the enzyme-mimicking traits of nanozymes and show the rational design of nanozymes to improve antioxidant enzyme-like activities. For example, shapes, surface areas, surface facets, particle morphologies, porous structures, pore sizes and volumes, metal doping, and combinations of different functional enzymes may lead to the different catalytic abilities of nanozymes. Although abundant efforts have been made on nanozymes in ROS-involved diseases, there are still shortcomings and deficiencies that need further improvement.

  1. The low catalytic activity and poor substrate selectivity of nanozymes are still big problems, which will be a huge stumbling block for their practical application in the future. According to the above summary, doping, control of shape and morphology, forming complex nanozymes, single atom technology, etc., are effective measures to improve the activity of nanozymes. A biomimetic strategy that simulates the active sites of natural enzymes may help to enhance the efficiency of nanozymes.
  2. It is well known that living organisms are complex systems. When nanozymes are used in vivo, their pharmacokinetics, ie, the absorption, distribution, biochemical conversion and excretion of nanozymes in the body, need a detailed examination, and based on this information, the application prospects of nanozymes can be overall assessed. In addition, the lack of detailed evaluation of the biosafety and biocompatibility of nanozymes is another problem. Fears have been raised about the potential toxicity of nanozymes on biological systems. The problem in the usage of nanozymes in the central nervous system also includes BBB permeability. It is worth noting that the compatibility of nanozymes can be improved by the loading of natural proteins, which may be a feasible strategy.163,164
  3. At present, the use of nanozymes for ROS-related diseases involves AD, PD, colitis, acute kidney injury and acute liver injury, which are serious and incurable; however, the treatment should expand to diseases that are not life threatening but also bring pain to patients, such as oral ulcers caused by radiation therapy and difficult-to-heal wounds.

With respect to the problems above, further refinement for high-performance, excellent biosafety, and detailed research in diverse animal models, nanozymes, which are easy to synthesize and low-cost, may be further developed and hold promise as a new therapeutic strategy for ROS-related diseases in clinics.

Funding

This work is supported by the Medical Key Science Project of Shanxi Province, No. 2020XM52, the Scientific and Technological Activities for Overseas Students in Shanxi Province, No. 20200042, and Priority Academic Program Development of Jiangsu Higher Education institutions (PAPD).

Disclosure

The authors report no conflicts of interest in this work.

References

1. Turrens JF. Mitochondrial formation of reactive oxygen species. J Physiol. 2003;552(2):335–344. doi:10.1113/jphysiol.2003.049478

2. Held P. An introduction to reactive oxygen species. Tech Resources-App Guides. 2012;802:5–9.

3. Bayir H. Reactive oxygen species. Crit Care Med. 2005;33(12 Suppl):S498–501. doi:10.1097/01.ccm.0000186787.64500.12

4. Ray PD, Huang BW, Tsuji Y. Reactive oxygen species (ROS) homeostasis and redox regulation in cellular signaling. Cell Signal. 2012;24(5):981–990. doi:10.1016/j.cellsig.2012.01.008

5. Dhiman A, Handa M, Ruwali M, Singh DP, Kesharwani P, Shukla R. Recent trends of natural based therapeutics for mitochondria targeting in Alzheimer’s disease. Mitochondrion. 2022;64:112–124. doi:10.1016/j.mito.2022.03.006

6. Mittler R. ROS are good. Trends in Plant Sci. 2017;22(1):11–19. doi:10.1016/j.tplants.2016.08.002

7. Checa J, Aran JM. Reactive oxygen species: drivers of physiological and pathological processes. J Inflamm Res. 2020;13:1057. doi:10.2147/JIR.S275595

8. Kang DH. Oxidative stress, DNA damage, and breast cancer. AACN Adv Crit Care. 2002;13(4):540–549. doi:10.1097/00044067-200211000-00007

9. Monaghan P, Metcalfe NB, Torres R. Oxidative stress as a mediator of life history trade-offs: mechanisms, measurements and interpretation. Ecol Lett. 2009;12(1):75–92. doi:10.1111/j.1461-0248.2008.01258.x

10. Andersen JK. Oxidative stress in neurodegeneration: cause or consequence? Nat Med. 2004;10(S7):S18–25. doi:10.1038/nrn1434

11. Melo A, Monteiro L, Lima RM, de Oliveira DM, de Cerqueira MD, El-Bachá RS. Oxidative stress in neurodegenerative diseases: mechanisms and therapeutic perspectives. Oxid Med Cell Longev. 2011;2011:1–14. doi:10.1155/2011/467180

12. Mittal M, Siddiqui MR, Tran K, Reddy SP, Malik AB. Reactive oxygen species in inflammation and tissue injury. Antioxid Redox Signal. 2014;20(7):1126–1167. doi:10.1089/ars.2012.5149

13. Amani H, Habibey R, Hajmiresmail SJ, Latifi S, Pazoki-Toroudi H, Akhavan O. Antioxidant nanomaterials in advanced diagnoses and treatments of ischemia reperfusion injuries. J Mater Chem B. 2017;5(48):9452–9476. doi:10.1039/c7tb01689a

14. Droge W. Free radicals in the physiological control of cell function. Physiol Rev. 2002;82(1):47–95. doi:10.1152/physrev.00018.2001

15. Wei H, Wang E. Nanomaterials with enzyme-like characteristics (nanozymes): next-generation artificial enzymes. Chem Soc Rev. 2013;42(14):6060–6093. doi:10.1039/c3cs35486e

16. Gao L, Zhuang J, Nie L, et al. Intrinsic peroxidase-like activity of ferromagnetic nanoparticles. Nat Nanotechnol. 2007;2(9):577–583. doi:10.1038/nnano.2007.260

17. Yu X, Xu Z, Wang X, Xu Q, Chen J. Bactrian camel serum albumins-based nanocomposite as versatile biocargo for drug delivery, biocatalysis and detection of hydrogen peroxide. Mater Sci Eng C. 2020;109:110627. doi:10.1016/j.msec.2020.110627

18. Wang D, Jana D, Zhao Y. Metal–organic framework derived nanozymes in biomedicine. Acc Chem Res. 2020;53(7):1389–1400. doi:10.1021/acs.accounts.0c00268

19. Yan X, Gao L. Nanozymology: an Overview. In: Yan X, editor. Nanozymology. Nanostructure Science and Technology. Singapore: Springer; 2020:3–16. doi:10.1007/978-981-15-1490-6_1

20. Liu T, Xiao B, Xiang F, et al. Ultrasmall copper-based nanoparticles for reactive oxygen species scavenging and alleviation of inflammation related diseases. Nat Commun. 2020;11(1):2788. doi:10.1038/s41467-020-16544-7

21. Wu J, Wang X, Wang Q, et al. Nanomaterials with enzyme-like characteristics (nanozymes): next-generation artificial enzymes (II). Chem Soc Rev. 2019;48(4):1004–1076. doi:10.1039/c8cs00457a

22. Gaur M, Misra C, Yadav AB, et al. Biomedical applications of carbon nanomaterials: fullerenes, quantum dots, nanotubes, nanofibers, and graphene. Materials. 2021;14(20):5978. doi:10.3390/ma14205978

23. Karimi-Maleh H, Beitollahi H, Kumar PS, et al. Recent advances in carbon nanomaterials-based electrochemical sensors for food azo dyes detection. Food Chem Toxicol. 2022;164:112961. doi:10.1016/j.fct.2022.112961

24. Dellinger A, Zhou Z, Connor J, et al. Application of fullerenes in nanomedicine: an update. Nanomedicine. 2013;8(7):1191–1208. doi:10.2217/nnm.13.99

25. Speranza G. Carbon nanomaterials: synthesis, functionalization and sensing applications. Nanomaterials. 2021;11(4):967. doi:10.3390/nano11040967

26. Bakry R, Vallant RM, Najam-ul-Haq M, et al. Medicinal applications of fullerenes. Int J Nanomedicine. 2007;2(4):639–649.

27. Xiao L, Takada H, Gan X, Miwa N. The water-soluble fullerene derivative “Radical Sponge” exerts cytoprotective action against UVA irradiation but not visible-light-catalyzed cytotoxicity in human skin keratinocytes. Bioorg Med Chem Lett. 2006;16(6):1590–1595. doi:10.1016/j.bmcl.2005.12.011

28. Castro E, Garcia AH, Zavala G, Echegoyen L. Fullerenes in Biology and Medicine. J Mater Chem B. 2017;5(32):6523–6535. doi:10.1039/C7TB00855D

29. Cheng X, Ni X, Wu R, et al. Evaluation of the structure–activity relationship of carbon nanomaterials as antioxidants. Nanomedicine. 2018;13(7):733–747. doi:10.2217/nnm-2017-0314

30. Yudoh K, Karasawa R, Masuko K, Kato T. Water-soluble fullerene (C60) inhibits the development of arthritis in the rat model of arthritis. Int J Nanomedicine. 2009;4:217–225. doi:10.2147/ijn.s7653

31. Hao T, Li J, Yao F, et al. Injectable fullerenol/alginate hydrogel for suppression of oxidative stress damage in brown adipose-derived stem cells and cardiac repair. ACS Nano. 2017;11(6):5474–5488. doi:10.1021/acsnano.7b00221

32. Aqel A, Abou El-Nour KM, Ammar RA, Al-Warthan A. Carbon nanotubes, science and technology part (I) structure, synthesis and characterisation. Arab J Chem. 2012;5(1):1–23. doi:10.1016/j.arabjc.2010.08.022

33. Fenoglio I, Tomatis M, Lison D, et al. Reactivity of carbon nanotubes: free radical generation or scavenging activity? Free Radic Biol Med. 2006;40(7):1227–1233. doi:10.1016/j.freeradbiomed.2005.11.010

34. Qiu Y, Wang Z, Owens AC, et al. Antioxidant chemistry of graphene-based materials and its role in oxidation protection technology. Nanoscale. 2014;6(20):11744–11755. doi:10.1039/c4nr03275f

35. Ren C, Hu X, Zhou Q. Graphene oxide quantum dots reduce oxidative stress and inhibit neurotoxicity in vitro and in vivo through catalase-like activity and metabolic regulation. Adv Sci. 2018;5(5):1700595. doi:10.1002/advs.201700595

36. Han J, Kim YS, Lim MY, et al. Dual roles of graphene oxide to attenuate inflammation and elicit timely polarization of macrophage phenotypes for cardiac repair. ACS Nano. 2018;12(2):1959–1977. doi:10.1021/acsnano.7b09107

37. Chen Z, Yin JJ, Zhou YT, et al. Dual enzyme-like activities of iron oxide nanoparticles and their implication for diminishing cytotoxicity. ACS Nano. 2012;6(5):4001–4012. doi:10.1021/nn300291r

38. Gao L, Fan K, Yan X. Iron oxide nanozyme: a multifunctional enzyme mimetic for biomedical applications. Theranostics. 2017;7(13):3207–3227. doi:10.7150/thno.19738

39. Zhang Y, Wang Z, Li X, et al. Dietary iron oxide nanoparticles delay aging and ameliorate neurodegeneration in Drosophila. Adv Mater. 2016;28(7):1387–1393. doi:10.1002/adma.201503893

40. Wei Z, Wang L, Tang C, et al. Metal-phenolic networks nanoplatform to mimic antioxidant defense system for broad-spectrum radical eliminating and endotoxemia treatment. Adv Funct Mater. 2020;30(49):2002234. doi:10.1002/adfm.202002234

41. Zhang W, Ma D, Du J. Prussian blue nanoparticles as peroxidase mimetics for sensitive colorimetric detection of hydrogen peroxide and glucose. Talanta. 2014;120:362–367. doi:10.1016/j.talanta.2013.12.028

42. Zhang W, Hu S, Yin JJ, et al. Prussian blue nanoparticles as multienzyme mimetics and reactive oxygen species scavengers. J Am Chem Soc. 2016;138(18):5860–5865. doi:10.1021/jacs.5b12070

43. Zhao J, Cai X, Gao W, et al. Prussian blue nanozyme with multienzyme activity reduces colitis in mice. ACS Appl Mater Interfaces. 2018;10(31):26108–26117. doi:10.1021/acsami.8b10345

44. Estelrich J, Busquets MA. Prussian blue: a nanozyme with versatile catalytic properties. Int J Mol Sci. 2021;22(11):5993. doi:10.3390/ijms22115993

45. Chen J, Wang Q, Huang L, et al. Prussian blue with intrinsic heme-like structure as peroxidase mimic. Nano Res. 2018;11(9):4905–4913. doi:10.1007/s12274-018-2079-8

46. Bai H, Kong F, Feng K, et al. Prussian blue nanozymes prevent anthracycline-induced liver injury by attenuating oxidative stress and regulating inflammation. ACS Appl Mater Interfaces. 2021;13(36):42382–42395. doi:10.1021/acsami.1c09838

47. Xie X, Zhao J, Gao W, et al. Prussian blue nanozyme-mediated nanoscavenger ameliorates acute pancreatitis via inhibiting TLRs/NF-κB signaling pathway. Theranostics. 2021;11(7):3213–3228. doi:10.7150/thno.52010

48. Sahu A, Jeon J, Lee MS, Yang HS, Tae G. Antioxidant and anti-inflammatory activities of Prussian blue nanozyme promotes full-thickness skin wound healing. Mater Sci Eng C Mater Biol Appl. 2021;119:111596. doi:10.1016/j.msec.2020.111596

49. Zhang K, Tu M, Gao W, et al. Hollow Prussian blue nanozymes drive neuroprotection against ischemic stroke via attenuating oxidative stress, counteracting inflammation, and suppressing cell apoptosis. Nano Lett. 2019;19(5):2812–2823. doi:10.1021/acs.nanolett.8b04729

50. Feng L, Dou C, Xia Y, et al. Neutrophil-like cell-membrane-coated nanozyme therapy for ischemic brain damage and long-term neurological functional recovery. ACS Nano. 2021;15(2):2263–2280. doi:10.1021/acsnano.0c07973

51. He W, Zhou YT, Wamer WG, et al. Intrinsic catalytic activity of Au nanoparticles with respect to hydrogen peroxide decomposition and superoxide scavenging. Biomaterials. 2013;34(3):765–773. doi:10.1016/j.biomaterials.2012.10.010

52. Liu Z, Shen Y, Wu Y, et al. An intrinsic therapy of gold nanoparticles in focal cerebral ischemia-reperfusion injury in rats. J Biomed Nanotechnol. 2013;9(6):1017–1028. doi:10.1166/jbn.2013.1597

53. Liu CP, Wu TH, Lin YL, Liu CY, Wang S, Lin SY. Tailoring enzyme-like activities of gold nanoclusters by polymeric tertiary amines for protecting neurons against oxidative stress. Small. 2016;12(30):4127–4135. doi:10.1002/smll.201503919

54. Schubert D, Dargusch R, Raitano J, Chan SW. Cerium and yttrium oxide nanoparticles are neuroprotective. Biochem Biophys Res Commun. 2006;342(1):86–91. doi:10.1016/j.bbrc.2006.01.129

55. Pirmohamed T, Dowding JM, Singh S, et al. Nanoceria exhibit redox state-dependent catalase mimetic activity. Chem Commun (Camb). 2010;46(16):2736–2738. doi:10.1039/b922024k

56. Li Y, He X, Yin JJ, et al. Acquired superoxide-scavenging ability of ceria nanoparticles. Angew Chem Int Ed Engl. 2015;54(6):1832–1835. doi:10.1002/anie.201410398

57. Akhtar MJ, Ahamed M, Alhadlaq HA, Khan MAM, Alrokayan SA. Glutathione replenishing potential of CeO2 nanoparticles in human breast and fibrosarcoma cells. J Colloid Interface Sci. 2015;453:21–27. doi:10.1016/j.jcis.2015.04.049

58. Hirst SM, Karakoti AS, Tyler RD, Sriranganathan N, Seal S, Reilly CM. Anti-inflammatory properties of cerium oxide nanoparticles. Small. 2009;5(24):2848–2856. doi:10.1002/smll.200901048

59. 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. doi:10.1021/nn2048069

60. Dowding JM, Song W, Bossy K, et al. Cerium oxide nanoparticles protect against Abeta-induced mitochondrial fragmentation and neuronal cell death. Cell Death Differ. 2014;21(10):1622–1632. doi:10.1038/cdd.2014.72

61. Singh R, Singh S. Redox-dependent catalase mimetic cerium oxide-based nanozyme protect human hepatic cells from 3-AT induced acatalasemia. Colloids Surf B Biointerfaces. 2019;175:625–635. doi:10.1016/j.colsurfb.2018.12.042

62. Bao Q, Hu P, Xu Y, et al. Simultaneous blood-brain barrier crossing and protection for stroke treatment based on edaravone-loaded ceria nanoparticles. ACS Nano. 2018;12(7):6794–6805. doi:10.1021/acsnano.8b01994

63. Kim CK, Kim T, Choi IY, et al. Ceria nanoparticles that can protect against ischemic stroke. Angew Chem Int Ed Engl. 2012;51(44):11039–11043. doi:10.1002/anie.201203780

64. Kwon HJ, Cha MY, Kim D, et al. Mitochondria-targeting ceria nanoparticles as antioxidants for Alzheimer’s disease. ACS Nano. 2016;10(2):2860–2870. doi:10.1021/acsnano.5b08045

65. Kim D, Kwon HJ, Hyeon T. Magnetite/ceria nanoparticle assemblies for extracorporeal cleansing of amyloid-beta in Alzheimer’s disease. Adv Mater. 2019;31(19):e1807965. doi:10.1002/adma.201807965

66. Chen Q, Du Y, Zhang K, et al. Tau-targeted multifunctional nanocomposite for combinational therapy of Alzheimer’s disease. ACS Nano. 2018;12(2):1321–1338. doi:10.1021/acsnano.7b07625

67. Fu S, Chen H, Yang W, et al. ROS-targeted depression therapy via BSA-incubated ceria nanoclusters. Nano Lett. 2022;22(11):4519–4527. doi:10.1021/acs.nanolett.2c01334

68. Kim J, Hong G, Mazaleuskaya L, et al. Ultrasmall antioxidant cerium oxide nanoparticles for regulation of acute inflammation. ACS Appl Mater Interfaces. 2021;13(51):60852–60864. doi:10.1021/acsami.1c16126

69. Zhao S, Li Y, Liu Q, et al. An orally administered CeO2 montmorillonite nanozyme targets inflammation for inflammatory bowel disease therapy. Adv Funct Mater. 2020;30(45). doi:10.1002/adfm.202004692

70. Lin A, Sun Z, Xu X, et al. Self-cascade uricase/catalase mimics alleviate acute gout. Nano Lett. 2022;22(1):508–516. doi:10.1021/acs.nanolett.1c04454

71. Wu L, Liu G, Wang W, et al. Cyclodextrin-modified CeO2 nanoparticles as a multifunctional nanozyme for combinational therapy of psoriasis. Int J Nanomedicine. 2020;15:2515–2527. doi:10.2147/IJN.S246783

72. Soh M, Kang DW, Jeong HG, et al. Ceria-zirconia nanoparticles as an enhanced multi-antioxidant for sepsis treatment. Angew Chem Int Ed Engl. 2017;56(38):11399–11403. doi:10.1002/anie.201704904

73. Li F, Qiu Y, Xia F, et al. Dual detoxification and inflammatory regulation by ceria nanozymes for drug-induced liver injury therapy. Nano Today. 2020;35:100925. doi:10.1016/j.nantod.2020.100925

74. Adebayo OA, Akinloye O, Adaramoye OA. Cerium oxide nanoparticles attenuate oxidative stress and inflammation in the liver of diethylnitrosamine-treated mice. Biol Trace Elem Res. 2020;193(1):214–225. doi:10.1007/s12011-019-01696-5

75. Kwon HJ, Kim D, Seo K, et al. Ceria nanoparticle systems for selective scavenging of mitochondrial, intracellular, and extracellular reactive oxygen species in Parkinson’s disease. Angew Chem Int Ed Engl. 2018;57(30):9408–9412. doi:10.1002/anie.201805052

76. 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):10582–10596. doi:10.1021/nn403743b

77. Yu Y, Zhao S, Gu D, et al. Cerium oxide nanozyme attenuates periodontal bone destruction by inhibiting the ROS-NFκB pathway. Nanoscale. 2022;14(7):2628–2637. doi:10.1039/d1nr06043k

78. Korschelt K, Ragg R, Metzger CS, et al. Glycine-functionalized copper(ii) hydroxide nanoparticles with high intrinsic superoxide dismutase activity. Nanoscale. 2017;9(11):3952–3960. doi:10.1039/c6nr09810j

79. Hao C, Qu A, Xu L, et al. Chiral molecule-mediated porous CuxO nanoparticle clusters with antioxidation activity for ameliorating Parkinson’s disease. J Am Chem Soc. 2019;141(2):1091–1099. doi:10.1021/jacs.8b11856

80. Liu X, Wang Q, Zhao H, Zhang L, Su Y, Lv Y. BSA-templated MnO2 nanoparticles as both peroxidase and oxidase mimics. Analyst. 2012;137(19):4552–4558. doi:10.1039/c2an35700c

81. Li W, Liu Z, Liu C, Guan Y, Ren J, Qu X. Manganese dioxide nanozymes as responsive cytoprotective shells for individual living cell encapsulation. Angew Chem Int Ed Engl. 2017;56(44):13661–13665. doi:10.1002/anie.201706910

82. Yao J, Cheng Y, Zhou M, et al. ROS scavenging Mn3O4 nanozymes for in vivo anti-inflammation. Chem Sci. 2018;9(11):2927–2933. doi:10.1039/c7sc05476a

83. Singh N, Savanur MA, Srivastava S, D’Silva P, Mugesh G. A manganese oxide nanozyme prevents the oxidative damage of biomolecules without affecting the endogenous antioxidant system. Nanoscale. 2019;11(9):3855–3863. doi:10.1039/c8nr09397k

84. Singh N, Savanur MA, Srivastava S, D’Silva P, Mugesh G. A redox modulatory Mn3O4 nanozyme with multi-enzyme activity provides efficient cytoprotection to human cells in a Parkinson’s disease model. Angew Chem Int Ed Engl. 2017;56(45):14267–14271. doi:10.1002/anie.201708573

85. Huang Y, Liu C, Pu F, Liu Z, Ren J, Qu X. A GO-Se nanocomposite as an antioxidant nanozyme for cytoprotection. Chem Commun (Camb). 2017;53(21):3082–3085. doi:10.1039/c7cc00045f

86. Huang Y, Liu Z, Liu C, Zhang Y, Ren J, Qu X. Selenium-based nanozyme as biomimetic antioxidant machinery. Chemistry. 2018;24(40):10224–10230. doi:10.1002/chem.201801725

87. Chen X, Zhu X, Gong Y, et al. Porous selenium nanozymes targeted scavenging ROS synchronize therapy local inflammation and sepsis injury. Appl Mater Today. 2021;22. doi:10.1016/j.apmt.2020.100929

88. Chen B, Xiang S, Qian G. Metal-organic frameworks with functional pores for recognition of small molecules. Acc Chem Res. 2010;43(8):1115–1124. doi:10.1021/ar100023y

89. Kreno LE, Leong K, Farha OK, Allendorf M, Van Duyne RP, Hupp JT. Metal-organic framework materials as chemical sensors. Chem Rev. 2012;112(2):1105–1125. doi:10.1021/cr200324t

90. Tan H, Ma C, Gao L, et al. Metal–organic framework‐derived copper nanoparticle@ carbon nanocomposites as peroxidase mimics for colorimetric sensing of ascorbic acid. Chem Eur J. 2014;20(49):16377–16383. doi:10.1002/chem.201404960

91. Zhang JW, Zhang HT, Du ZY, Wang X, Yu SH, Jiang HL. Water-stable metal-organic frameworks with intrinsic peroxidase-like catalytic activity as a colorimetric biosensing platform. Chem Commun (Camb). 2014;50(9):1092–1094. doi:10.1039/c3cc48398c

92. Qi Z, Wang L, You Q, Chen Y. PA-Tb-Cu MOF as luminescent nanoenzyme for catalytic assay of hydrogen peroxide. Biosens Bioelectron. 2017;96:227–232. doi:10.1016/j.bios.2017.05.013

93. Chen WH, Vazquez-Gonzalez M, Kozell A, Cecconello A, Willner I. Cu2+-modified metal-organic framework nanoparticles: a peroxidase-mimicking nanoenzyme. Small. 2018;14(5). doi:10.1002/smll.201703149

94. Zheng HQ, Liu CY, Zeng XY, et al. MOF-808: a metal-organic framework with intrinsic peroxidase-like catalytic activity at neutral pH for colorimetric biosensing. Inorg Chem. 2018;57(15):9096–9104. doi:10.1021/acs.inorgchem.8b01097

95. Zhang K, Meng X, Cao Y, et al. Metal-organic framework nanoshuttle for synergistic photodynamic and low-temperature photothermal therapy. Adv Funct Mater. 2018;28(42):1804634. doi:10.1002/adfm.201804634

96. Li H, Cao X, Fei X, Zhang S, Xian Y. Nanoscaled luminescent terbium metal–organic frameworks for measuring and scavenging reactive oxygen species in living cells. J Mater Chem B. 2019;7(18):3027–3033. doi:10.1039/c9tb00361d

97. Zhang L, Zhang Y, Wang Z, et al. Constructing metal–organic framework nanodots as bio-inspired artificial superoxide dismutase for alleviating endotoxemia. Mater Horiz. 2019;6(8):1682–1687. doi:10.1039/c9mh00339h

98. Liu H, Yang Y, Liu Y, et al. Melanin-like nanomaterials for advanced biomedical applications: a versatile platform with extraordinary promise. Adv Sci. 2020;7(7):1903129. doi:10.1002/advs.201903129

99. Yue Y, Zhao X. Melanin-like nanomedicine in photothermal therapy applications. Int J Mol Sci. 2021;22(1):399. doi:10.3390/ijms22010399

100. Tada M, Kohno M, Niwano Y. Scavenging or quenching effect of melanin on superoxide anion and singlet oxygen. JClinbiochem nutr. 2010;46:224–228. doi:10.3164/jcbn.09-84

101. Liu Y, Ai K, Lu L. Polydopamine and its derivative materials: synthesis and promising applications in energy, environmental, and biomedical fields. Chem Rev. 2014;114(9):5057–5115. doi:10.1021/cr400407a

102. Hu J, Yang L, Yang P, Jiang S, Liu X, Li Y. Polydopamine free radical scavengers. Biomater Sci. 2020;8(18):4940–4950. doi:10.1039/d0bm01070g

103. Sun T, Jiang D, Rosenkrans ZT, et al. A melanin-based natural antioxidant defense nanosystem for theranostic application in acute kidney injury. Adv Funct Mater. 2019;29(48):1904833. doi:10.1002/adfm.201904833

104. Yang XF, Wang A, Qiao B, Li J, Liu J, Zhang T. Single-atom catalysts: a new frontier in heterogeneous catalysis. Acc Chem Res. 2013;46(8):1740–1748. doi:10.1021/ar300361m

105. Jiao L, Yan H, Wu Y, et al. When nanozymes meet single-atom catalysis. Angew Chem Int Ed Engl. 2020;59(7):2565–2576. doi:10.1002/anie.201905645

106. Pei J, Zhao R, Mu X, Wang J, Liu C, Zhang XD. Single-atom nanozymes for biological applications. Biomater Sci. 2020;8(23):6428–6441. doi:10.1039/d0bm01447h

107. Cao F, Zhang L, You Y, Zheng L, Ren J, Qu X. An enzyme‐mimicking single‐atom catalyst as an efficient multiple reactive oxygen and nitrogen species scavenger for sepsis management. Angewandte Chemie. 2020;132(13):5146–5153. doi:10.1002/ange.201912182

108. Yan R, Sun S, Yang J, et al. Nanozyme-based bandage with single-atom catalysis for brain trauma. ACS nano. 2019;13(10):11552–11560. doi:10.1021/acsnano.9b05075

109. Korsvik C, Patil S, Seal S, Self WT. Superoxide dismutase mimetic properties exhibited by vacancy engineered ceria nanoparticles. Chem Commun (Camb). 2007;(10):1056–1058. doi:10.1039/b615134e

110. Xi Z, Gao W, Xia X. Size effect in Pd-Ir core-shell nanoparticles as nanozymes. Chembiochem. 2020;21(17):2440–2444. doi:10.1002/cbic.202000147

111. Xu R, Wang D, Zhang J, Li Y. Shape-dependent catalytic activity of silver nanoparticles for the oxidation of styrene. Chem Asian J. 2006;1(6):888–893. doi:10.1002/asia.200600260

112. Ge C, Fang G, Shen X, et al. Facet energy versus enzyme-like activities: the unexpected protection of palladium nanocrystals against oxidative damage. ACS Nano. 2016;10(11):10436–10445. doi:10.1021/acsnano.6b06297

113. He W, Wu X, Liu J, et al. Design of AgM bimetallic alloy nanostructures (M = Au, Pd, Pt) with tunable morphology and peroxidase-like activity. Chem Mater. 2010;22(9):2988–2994. doi:10.1021/cm100393v

114. Wu J, Qin K, Yuan D, et al. Rational design of Au@Pt multibranched nanostructures as bifunctional nanozymes. ACS Appl Mater Interfaces. 2018;10(15):12954–12959. doi:10.1021/acsami.7b17945

115. Xia X, Zhang J, Lu N, et al. Pd–Ir core–shell nanocubes: a type of highly efficient and versatile peroxidase mimic. Acs Nano. 2015;9(10):9994–10004. doi:10.1021/acsnano.5b03525

116. Gao Z, Ye H, Tang D, et al. Platinum-decorated gold nanoparticles with dual functionalities for ultrasensitive colorimetric in vitro diagnostics. Nano Lett. 2017;17(9):5572–5579. doi:10.1021/acs.nanolett.7b02385

117. Lv F, Gong Y, Cao Y, et al. A convenient detection system consisting of efficient Au@PtRu nanozymes and alcohol oxidase for highly sensitive alcohol biosensing. Nanoscale Adv. 2020;2(4):1583–1589. doi:10.1039/d0na00002g

118. Mu X, Wang J, Li Y, et al. Redox trimetallic nanozyme with neutral environment preference for brain injury. ACS Nano. 2019;13(2):1870–1884. doi:10.1021/acsnano.8b08045

119. Wang Q, Cheng C, Zhao S, et al. A valence‐engineered self‐cascading antioxidant nanozyme for the therapy of inflammatory bowel disease. Angew Chem Int Ed Engl. 2022. doi:10.1002/anie.202201101

120. Wang C, Ren G, Yuan B, et al. Enhancing enzyme-like activities of Prussian blue analog nanocages by molybdenum doping: toward cytoprotecting and online optical hydrogen sulfide monitoring. Anal Chem. 2020;92(11):7822–7830. doi:10.1021/acs.analchem.0c01028

121. Liu Y-Q, Mao Y, Xu E, et al. Nanozyme scavenging ROS for prevention of pathologic α-synuclein transmission in Parkinson’s disease. Nano Today. 2021;36:101027. doi:10.1016/j.nantod.2020.101027

122. Liu B, Liu J. Surface modification of nanozymes. Nano Res. 2017;10(4):1125–1148. doi:10.1007/s12274-017-1426-5

123. Liu B, Huang Z, Liu J. Boosting the oxidase mimicking activity of nanoceria by fluoride capping: rivaling protein enzymes and ultrasensitive F detection. Nanoscale. 2016;8(28):13562–13567. doi:10.1039/c6nr02730j

124. Wang S, Chen W, Liu AL, Hong L, Deng HH, Lin XH. Comparison of the peroxidase-like activity of unmodified, amino-modified, and citrate-capped gold nanoparticles. Chemphyschem. 2012;13(5):1199–1204. doi:10.1002/cphc.201100906

125. Zhang XQ, Gong SW, Zhang Y, Yang T, Wang CY, Gu N. Prussian blue modified iron oxide magnetic nanoparticles and their high peroxidase-like activity. J Mater Chem. 2010;20(24). doi:10.1039/c0jm00174k

126. Liu X, Wei W, Yuan Q, et al. Apoferritin-CeO2 nano-truffle that has excellent artificial redox enzyme activity. Chem Commun (Camb). 2012;48(26):3155–3157. doi:10.1039/c1cc15815e

127. Asati A, Santra S, Kaittanis C, Nath S, Perez JM. Oxidase-like activity of polymer-coated cerium oxide nanoparticles. Angew Chem Int Ed Engl. 2009;48(13):2308–2312. doi:10.1002/anie.200805279

128. Pautler R, Kelly EY, Huang PJ, Cao J, Liu B, Liu J. Attaching DNA to nanoceria: regulating oxidase activity and fluorescence quenching. ACS Appl Mater Interfaces. 2013;5(15):6820–6825. doi:10.1021/am4018863

129. Fan K, Wang H, Xi J, et al. Optimization of Fe3O4 nanozyme activity via single amino acid modification mimicking an enzyme active site. Chem Commun (Camb). 2017;53(2):424–427. doi:10.1039/C6CC08542C

130. Sun Y, Zhao C, Gao N, Ren J, Qu X. Stereoselective nanozyme based on ceria nanoparticles engineered with amino acids. Chem Eur J. 2017;23(72):18146–18150. doi:10.1002/chem.201704579

131. Zhang R, Zhou Y, Yan X, Fan K. Advances in chiral nanozymes: a review. Mikrochim Acta. 2019;186(12):782. doi:10.1007/s00604-019-3922-7

132. Chen JLY, Pezzato C, Scrimin P, Prins LJ. Chiral nanozymes–gold nanoparticle‐based transphosphorylation catalysts capable of enantiomeric discrimination. Chem Eur J. 2016;22(21):7028–7032. doi:10.1002/chem.201600853

133. Zhan P, Wang ZG, Li N, Ding B. Engineering gold nanoparticles with DNA ligands for selective catalytic oxidation of chiral substrates. ACS Catal. 2015;5(3):1489–1498. doi:10.1021/cs5015805

134. Wang H, Yu D, Fang J, et al. Phenol-like group functionalized graphene quantum dots structurally mimicking natural antioxidants for highly efficient acute kidney injury treatment. Chem Sci. 2020;11(47):12721–12730. doi:10.1039/d0sc03246h

135. Sun X, Guo S, Chung CS, Zhu W, Sun S. A sensitive H2O2 assay based on dumbbell-like PtPd-Fe3O4 nanoparticles. Adv Mater. 2013;25(1):132–136. doi:10.1002/adma.201203218

136. Liu M, Li Z, Li Y, Chen J, Yuan Q. Self-assembled nanozyme complexes with enhanced cascade activity and high stability for colorimetric detection of glucose. Chin Chem Lett. 2019;30(5):1009–1012. doi:10.1016/j.cclet.2018.12.021

137. Wang Z, Dong K, Liu Z, et al. Activation of biologically relevant levels of reactive oxygen species by Au/g-C3N4 hybrid nanozyme for bacteria killing and wound disinfection. Biomaterials. 2017;113:145–157. doi:10.1016/j.biomaterials.2016.10.041

138. Zhang L, Pan J, Long Y, et al. CeO2-encapsulated hollow Ag-Au nanocage hybrid nanostructures as high-performance catalysts for cascade reactions. Small. 2019;15(43):e1903182. doi:10.1002/smll.201903182

139. Zhang S, Li H, Wang Z, et al. A strongly coupled Au/Fe3O4/GO hybrid material with enhanced nanozyme activity for highly sensitive colorimetric detection, and rapid and efficient removal of Hg2+ in aqueous solutions. Nanoscale. 2015;7(18):8495–8502. doi:10.1039/c5nr00527b

140. Huang Y, Liu Z, Liu C, et al. Self‐assembly of multi‐nanozymes to mimic an intracellular antioxidant defense system. Angew Chem Int Ed Engl. 2016;128(23):6758–6762. doi:10.1002/ange.201600868

141. He L, Huang G, Liu H, Sang C, Liu X, Chen T. Highly bioactive zeolitic imidazolate framework-8–capped nanotherapeutics for efficient reversal of reperfusion-induced injury in ischemic stroke. Sci Adv. 2020;6(12):eaay9751. doi:10.1126/sciadv.aay9751

142. Zhang K, Kaufman RJ. From endoplasmic-reticulum stress to the inflammatory response. Nature. 2008;454(7203):455–462. doi:10.1038/nature07203

143. Guo H, Callaway JB, Ting JP. Inflammasomes: mechanism of action, role in disease, and therapeutics. Nat Med. 2015;21(7):677–687. doi:10.1038/nm.3893

144. Ferrucci L, Fabbri E. Inflammageing: chronic inflammation in ageing, cardiovascular disease, and frailty. Nat Rev Cardiol. 2018;15(9):505–522. doi:10.1038/s41569-018-0064-2

145. Simmonds N, Rampton D. Inflammatory bowel disease--a radical view. Gut. 1993;34(7):865. doi:10.1136/gut.34.7.865

146. Qin Z, Li Y, Gu N. Progress in applications of Prussian blue nanoparticles in biomedicine. Adv Healthc Mater. 2018;7(20):1800347. doi:10.1002/adhm.201800347

147. Qiu H, Gong H, Bao Y, Jiang H, Tong W. Reactive oxygen species-scavenging hollow MnO2 nanozymes as carriers to deliver budesonide for synergistic inflammatory bowel disease therapy. Biomater Sci. 2022;10(2):457–466. doi:10.1039/d1bm01525g

148. Zhong G, Yang X, Jiang X, et al. Dopamine-melanin nanoparticles scavenge reactive oxygen and nitrogen species and activate autophagy for osteoarthritis therapy. Nanoscale. 2019;11(24):11605–11616. doi:10.1039/c9nr03060c

149. Kumar S, Adjei IM, Brown SB, Liseth O, Sharma B. Manganese dioxide nanoparticles protect cartilage from inflammation-induced oxidative stress. Biomaterials. 2019;224:119467. doi:10.1016/j.biomaterials.2019.119467

150. Bao X, Zhao J, Sun J, Hu M, Yang X. Polydopamine nanoparticles as efficient scavengers for reactive oxygen species in periodontal disease. ACS Nano. 2018;12(9):8882–8892. doi:10.1021/acsnano.8b04022

151. Zerna C, Thomalla G, Campbell BC, Rha J-H, Hill MD. Current practice and future directions in the diagnosis and acute treatment of ischaemic stroke. The Lancet. 2018;392(10154):1247–1256. doi:10.1016/S0140-6736(18)31874-9

152. Rzigalinski BA, Meehan K, Davis RM, Xu Y, Miles WC, Cohen CA. Radical nanomedicine. Nanomedcine. 2006;1(4):399–412. doi:10.2217/17435889.1.4.399

153. Liu Y, Ai K, Ji X, et al. Comprehensive insights into the multi-antioxidative mechanisms of melanin nanoparticles and their application to protect brain from injury in ischemic stroke. J Am Chem Soc. 2017;139(2):856–862. doi:10.1021/jacs.6b11013

154. Huang G, Zang J, He L, et al. Bioactive nanoenzyme reverses oxidative damage and endoplasmic reticulum stress in neurons under ischemic stroke. ACS Nano. 2021. doi:10.1021/acsnano.1c07205

155. Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science. 2002;297(5580):353–356. doi:10.1126/science.1072994

156. Polanco JC, Li C, Bodea LG, Martinez-Marmol R, Meunier FA, Götz J. Amyloid-β and tau complexity—towards improved biomarkers and targeted therapies. Nat Rev Neurol. 2018;14(1):22–39. doi:10.1038/nrneurol.2017.162

157. Cheignon C, Tomas M, Bonnefont-Rousselot D, Faller P, Hureau C, Collin F. Oxidative stress and the amyloid beta peptide in Alzheimer’s disease. Redox Biol. 2018;14:450–464. doi:10.1016/j.redox.2017.10.014

158. Savelieff MG, Nam G, Kang J, Lee HJ, Lee M, Lim MH. Development of multifunctional molecules as potential therapeutic candidates for Alzheimer’s disease, Parkinson’s disease, and amyotrophic lateral sclerosis in the last decade. Chem Rev. 2019;119(2):1221–1322. doi:10.1021/acs.chemrev.8b00138

159. Jenner P. Oxidative stress in Parkinson’s disease. Ann Reurol. 2003;53(S3):S26–S38. doi:10.1002/ana.10483

160. Spillantini MG, Schmidt ML, Lee VM-Y, Trojanowski JQ, Jakes R, Goedert M. α-Synuclein in Lewy bodies. Nature. 1997;388(6645):839–840. doi:10.1038/42166

161. Ma X, Hao J, Wu J, Li Y, Cai X, Zheng Y. Prussian blue nanozyme as a pyroptosis inhibitor alleviates neurodegeneration. Adv Mater. 2022;34(15):2106723. doi:10.1002/adma.202106723

162. Ni D, Jiang D, Kutyreff CJ, et al. Molybdenum-based nanoclusters act as antioxidants and ameliorate acute kidney injury in mice. Nat Commun. 2018;9(1):5421. doi:10.1038/s41467-018-07890-8

163. Zhang J, Pei W, Xu Q, Jiang H, Chen J. Desolvation-induced formation of recombinant camel serum albumin-based nanocomposite for glutathione colorimetric determination. Sens Actuators B Che. 2022;1;357.

164. Zhang J, Xu Q, Pei W, et al. Self-assembled recombinant camel serum albumin nanoparticles-encapsulated hemin with peroxidase-like activity for colorimetric detection of hydrogen peroxide and glucose. Int J Biol Macromol. 2021;193:2103–2112. doi:10.1016/j.ijbiomac.2021.11.042

165. Ryan JJ, Bateman HR, Stover A, et al. Fullerene nanomaterials inhibit the allergic response. J Immunol. 2007;179(1):665–672. doi:10.4049/jimmunol.179.1.665

166. Jacevic V, Djordjevic A, Srdjenovic B, et al. Fullerenol nanoparticles prevents doxorubicin-induced acute hepatotoxicity in rats. Exp Mol Pathol. 2017;102(2):360–369. doi:10.1016/j.yexmp.2017.03.005

167. Zhao S, Duan H, Yang Y, Yan X, Fan K. Fenozyme protects the integrity of the blood-brain barrier against experimental cerebral malaria. Nano Lett. 2019;19(12):8887–8895. doi:10.1021/acs.nanolett.9b03774

168. Kalashnikova I, Chung SJ, Nafiujjaman M, et al. Ceria-based nanotheranostic agent for rheumatoid arthritis. Theranostics. 2020;10(26):11863–11880. doi:10.7150/thno.49069

169. Bailey ZS, Nilson E, Bates JA, et al. Cerium oxide nanoparticles improve outcome after in vitro and in vivo mild traumatic brain injury. J Neurotrauma. 2020;37(12):1452–1462. doi:10.1089/neu.2016.4644

170. Cheng Y, Cheng C, Yao J, et al. Mn3O4 nanozyme for inflammatory bowel disease therapy. Adv Ther. 2021;4(9). doi:10.1002/adtp.202100081

171. Zhang X, Zhang S, Yang Z, Wang Z, Tian X, Zhou R. Self-cascade MoS2 nanozymes for efficient intracellular antioxidation and hepatic fibrosis therapy. Nanoscale. 2021;13(29):12613–12622. doi:10.1039/d1nr02366g

172. Onizawa S, Aoshiba K, Kajita M, Miyamoto Y, Nagai A. Platinum nanoparticle antioxidants inhibit pulmonary inflammation in mice exposed to cigarette smoke. Pulm Pharmacol Ther. 2009;22(4):340–349. doi:10.1016/j.pupt.2008.12.015

173. Katsumi H, Fukui K, Sato K, et al. Pharmacokinetics and preventive effects of platinum nanoparticles as reactive oxygen species scavengers on hepatic ischemia/reperfusion injury in mice. Metallomics. 2014;6(5):1050–1056. doi:10.1039/c4mt00018h

174. Zhao H, Zeng Z, Liu L, et al. Polydopamine nanoparticles for the treatment of acute inflammation-induced injury. Nanoscale. 2018;10(15):6981–6991. doi:10.1039/c8nr00838h

175. Liu Y, Cheng Y, Zhang H, et al. Integrated cascade nanozyme catalyzes in vivo ROS scavenging for anti-inflammatory therapy. Sci Adv. 2020;6(29):eabb2695. doi:10.1126/sciadv.abb2695

176. Huang Y, Ren J, Qu X. Nanozymes: classification, catalytic mechanisms, activity regulation, and applications. Chem Rev. 2019;119(6):4357–4412. doi:10.1021/acs.chemrev.8b00672

Creative Commons License © 2022 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.