Next Article in Journal
The Characterization of Silicone-Tungsten-Based Composites as Flexible Gamma-Ray Shields
Next Article in Special Issue
Transition Metal Dichalcogenides (TMDC)-Based Nanozymes for Biosensing and Therapeutic Applications
Previous Article in Journal
Effect of Defocused Nanosecond Laser Paint Removal on Mild Steel Substrate in Ambient Atmosphere
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Potentiality of Nanoenzymes for Cancer Treatment and Other Diseases: Current Status and Future Challenges

1
Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India
2
Department of Vegetable Crops and Medicinal Plants, University of Life Sciences in Lublin, 50A Doświadczalna St., 20-280 Lublin, Poland
3
Department of Botany, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan
4
Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
5
Institute of Soil Science and Environment Shaping, University of Life Sciences in Lublin, 7 Leszczyńskiego St., 20-069 Lublin, Poland
6
Department of Pharmacy, Southeast University, Banani, Dhaka 1213, Bangladesh
7
Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Wonju 26426, Gangwon-do, Korea
*
Authors to whom correspondence should be addressed.
Materials 2021, 14(20), 5965; https://doi.org/10.3390/ma14205965
Submission received: 18 August 2021 / Revised: 23 September 2021 / Accepted: 30 September 2021 / Published: 11 October 2021
(This article belongs to the Special Issue Nanozyme: Synthesis, Mechanisms, and Applications)

Abstract

:
Studies from past years have observed various enzymes that are artificial, which are issued to mimic naturally occurring enzymes based on their function and structure. The nanozymes possess nanomaterials that resemble natural enzymes and are considered an innovative class. This innovative class has achieved a brilliant response from various developments and researchers owing to this unique property. In this regard, numerous nanomaterials are inspected as natural enzyme mimics for multiple types of applications, such as imaging, water treatment, therapeutics, and sensing. Nanozymes have nanomaterial properties occurring with an inheritance that provides a single substitute and multiple platforms. Nanozymes can be controlled remotely via stimuli including heat, light, magnetic field, and ultrasound. Collectively, these all can be used to increase the therapeutic as well as diagnostic efficacies. These nanozymes have major biomedical applications including cancer therapy and diagnosis, medical diagnostics, and bio sensing. We summarized and emphasized the latest progress of nanozymes, including their biomedical mechanisms and applications involving synergistic and remote control nanozymes. Finally, we cover the challenges and limitations of further improving therapeutic applications and provide a future direction for using engineered nanozymes with enhanced biomedical and diagnostic applications.

Graphical Abstract

1. Introduction

Enzymes are considered natural biocatalysts which catalyze many biochemical reactions with good catalytic efficiency, biocompatibility, and substrate specificity. Recently, these reactions have been extensively used in various food industries and other biomedical applications. Their use in the agri-food industry promotes proper processing, storage activities and the functionalization of food products [1,2,3,4,5,6]. Enzymes play a significant role in enhancing the safety of food products [7]. Nanotechnology is believed to have a major part in advanced drug formulation, targeting a specific part of the body and controlled release of the drug. Nanotechnology is stated to communicate with the barrier of physical and organic sciences by putting forward nanospheres and structures in numerous scientific fields [8,9] other than nanomedicines and their delivery [10,11]. Nanotechnology engages the therapeutic agents at nanoscale levels for the development of medicines that are nano. Biomedicine including nanobiotechnology, biosensors, and tissue designing is done by the nanoparticles [12]. Recently, nanomedicines have become very much refreshing as nanostructures act as delivery agents by giving medication examples [13,14]. Using conveyance nano-drugs for the treatment depends upon various properties of targeted drugs such as biochemical functions [15]. Over the past few years, scientists have made an extraordinary attempt in developing artificial enzymes for various types of applications. Consider the examples that the chemical complexes based on porphyrin [16,17], hematin [18], cyclodextrin [19], hemin [20,21], and the specially designed biomolecules proteins successively imitate the function of the naturally occurring enzymes [22,23].The intrinsic limitations of the natural enzymes such as low stability, high cost, and storage difficulty have led to the introduction of artificial enzymes that imitate the activity of the naturally occurring enzymes [24]. As another sort of promising artificial enzyme, nanozymes have demonstrated a wide range of uses because of their evident favorable circumstances, including low cost, high stability, the large surface area for functionalization, high catalytic activity, and tuneable activity [25]. Various obstacles and constraints of further developing therapeutic applications are of significant interest, as well as a future direction for the usage of modified nanozymes with better biomedical and diagnostic applications. Nanozymes are defined as artificial nanomaterials possessing intrinsic enzyme-like activities. Scientists have worked toward their enhancing utility as they have many advantages over natural enzymes. Nanozymes are believed to act by mimicking the action of the natural enzymes [26,27,28]. The concept of nanozyme has reformed our essential comprehension of chemistry and biology, encouraging plenty of uses in the fields of biosensing, science, and medication [26]. Nanozyme synthesis is an innovative technology since it connects nanoparticles with biological activities and framework. Various assays have been implemented for the enzymes of proteins that also implement nanozymes, which could also have the potential for performing the catalysis of similar substrates. Due to such different functions of nanozymes, they are used for the treatment of the environment, biosensing, agents that act against microbes, cytoprotection of different cell biomolecules with management, diagnosis of diseases, etc. [29,30,31,32,33]. Various sources, properties, mimicking types, and analytical capabilities are shown in Figure 1 [1].

2. Types of Nanozymes

The nanozymes can be classified into different types (Table 1) based on the enzymes whose actions they mimic. It was mentioned in a 2013 review that there are generally four types of redox enzymes which include catalase, superoxide dismutase (SOD), oxidase, and peroxidase [34].

2.1. Peroxidasemimics

2.1.1. Iron-Based

The Fe3O4 NPs, which are magnetic, have functions such as imitating the intrinsic peroxidase, which was discovered in 2007 by the Yan group. It could lead to the oxidation of the three colorless peroxidase substrates involving ortho-phenylenediamine, TMB, and diazoamino benzene to the colored materials, which are comparable with H2O2. The MNP nanozymes have the occurring mechanism, which is known as ping pong catalysis, and is suggested in the kinetics studies. The higher as well as lower affinity of nanozymes towards TMB and H2O2, respectively, compared to the HRP, has been well indicated by the measured Michaelis–Menten constants [35]. Later on, peroxidase imitates linked with Fe3O4 MNP were applied for detecting glucose and hydrogen peroxide [35,36].

2.1.2. Vanadium-Based

The first demonstration regarding the V2O5 nanowire-based peroxidase mimics was done by a group named Tremel in 2011 [37]. After that, maximum attention was gained by the peculiar vanadium haloperoxidase imitating the functions of V2O5 wires which are nano along with their anti-biofouling and marine applications [38]. Various research on vanadium disulfides and peroxidase, which is similar to vanadium, has been reported since then [39,40,41,42,43].

2.1.3. Based on Noble Metal

There are various nanomaterials based on noble metals such as gold [44,45,46,47,48,49,50,51,52,53,54,55], silver [56,57,58,59,60,61], platinum [62,63,64,65,66,67,68,69,70,71,72,73,74,75], Pd [76,77,78], and multi-metallic NPs which are known as peroxidase imitates and are utilized for antibodies, therapy, and biosensing.

2.1.4. Carbon-Based

Carbon is another typical nanomaterial as peroxidase-like activities with pH, temp, and hydrogen peroxide concentration dependent functions have been possessed by nanotubes which have a single wall and oxides of graphene [79,80]. Propelled by these findings, there are various other carbon-based peroxidase mimics such as carbon dots [81,82,83,84,85,86,87,88], Fe/N doped carbon [89,90,91,92,93,94], carbon nitrides [95,96,97], etc., which have been explored.

2.1.5. Based on Metal–Organic Framework

This framework which has diverse porous structures has been used widely for biomedical applications. It can consist of coordinating ions or clusters of metals (e.g., Cu and iron) comprising the organic ligands. 2D MOFs are believed to exhibit high functions of catalysis as compared to the analogues of 3D, hence giving effective sensibility for the detection of biomolecules [98].

2.2. Oxidase Mimics

2.2.1. Gold-Based

Even though the nanomaterials, which are metals, are generally utilized for showing catalyzing responses, the disclosure of carbon-upheld Au or unbearable citrate-covered AuNPs (along with 3.5 nm of normal distance) and glucose oxidase-copying exercises were amazing and startling [99,100]. Further, according to the kinetics measurements, the mechanism of EleyRideal was suggested for AuNP-based imitates of oxidase [101].

2.2.2. Copper-Based

Nanoparticles that contain copper were have also been used as imitates of oxidase. For example, Goximitating composites of Cu2O or polypyrrole were accounted for the oxidative catalysis of glucose for creating hydrogen peroxidase in fundamental terms. Although oxidation of rising glucose exercises the composites of Cu2O or polypyrrole, which guarantee identification of glucose, the situation for this response ought to be additionally improved in terms of physiological conditions for more extensive utilizations [102].

2.2.3. Molybdenum-Based

It has been reported by Tremeland co-workers that the molybdenum trioxide nanoparticles can mimic sulfite oxidase for converting it into sulfate [103]. The high stability in water and serum led to the development of ultra-smallMoO3 NPs (with 2 nm average diameter).

2.2.4. Based on Platinum

An important part is played by ferroxidases generally in the transfer and storage of Fe in the cellular environment. Later on, some research linked with PtNPs as the imitates of ferroxidase for oxidizing them were noted. Examples such as Zhang, Knezand collaborators used apoferritin, which is a light chain as the platform for PtNPs to get ready. Nanozymes such as these organized one scan control the homeostasis of the iron cell, which is profited by the ferroxidase [104].

2.3. Catalase Mimics

Catalase is believed to decompose H2O2 into H2O and O2 effectively. Many nanomaterials such as metal oxides, metals and PB exhibit the type of activities linked with catalase [105,106,107,108,109,110,111,112]. It was noted that nanoparticles had activities similar to catalase with another catalyst-impersonating function, and pH or temp could have made the catalyst impersonating action predominant. Pt and Pd were demonstrated for possessing the good imitating functions of catalase compared to gold and silver [113]. Some metal oxide nanomaterials (such as ZrO3 and CoO4) and PB were also found to show catalase-mimicking properties at higher pH [114,115].

2.4. Superoxide Dismustase (SOD) Mimics

The damage that occurred through oxidation to the living organization may be caused by the species of dysregulated oxygen which is reactive. Naturally, SOD is believed to eliminate the anion which is superoxide O2, ROS, throughout the dismutation response of O2 to hydrogen peroxide and oxygen. For surviving constraints linked with SOD, which is common, an assortment linked with nanomaterials is utilized to imitate SOD [116,117,118,119,120,121,122,123,124].

2.4.1. Carbon-Based

C60[C(COOH)2]3, comprised of symmetry linked with C3, has been approved for possessing more properties such as acting against oxidation [125]. The catalytic elimination of the superoxide anion O2 resulted in the antioxidation activity. Later on, the non-change of C60-C3 and production of O2 and H2O2 from O2 was confirmed by the mechanism studies just like the SOD catalyzed reaction [126].

2.4.2. Cerium-Based

Nanoceria was classified as one of the first nanomaterials possessing SOD-mimicking activity. These have been allocated to the shuttle of electrons between the mixed states of oxidation [127,128]. The superoxide mechanism of action showing the cerium oxide’s ability to scavenge has yet not been verified, but there are some studies showing more SOD-mimicking activity by the high ratio of Ce3+/Ce4+ [129,130,131].

2.4.3. Melanin-Based

The nanoparticles which are melanin in nature comprise various free radicals which scavenged the activities which have been implemented by a group of Shi [132]. The mixture of the hydrochloride of dopamine along with NH3 in the ethanol and water led to the synthesis of melanin nanoparticles. Further, their stability was improved by functionalizing with amine-terminated PEG. Such types of PEGcMeNPs with an approximate diameter of 120 nm were shown to possess SOD-like activities for O2 scavenging.

2.5. Hydrolase Mimics

The hydrolysis of the chemical bond is catalyzed by hydrolase. For example, the bonds of nucleotides are hydrolyzed by nucleosidase. The phosphatase enzyme helps in the catalysis of phosphate cleavage from the molecules [24].

2.5.1. Carbon-Based

Other than the previously mentioned peroxidase and SOD-imitating activity, ultimately, nanozymes that are based on carbon are initially found to replicate the common nucleases [133]. Aqueous-solvent fullerene worked with the corrosive carboxylics, known as C60-1, and it was exhibited for catalyzing the phosphodiester cleavage obligation occurring in DNA and illuminated through the light. Hence, by formulating the fullerenes and the corresponding DNA, the effectiveness of a particular DNA’s cleavage site would be increased [134,135]. Notwithstanding fullerenes, the oxides of graphene were additionally utilized as hydrolase imitates [136,137,138].

2.5.2. Monolayer Functionalized AuNP-Based

AuNPs worked along with monolayers which are catalytic throughout the bonds of gold and silver are amongst the very first nanomaterials imitating as hydrolases that deserve acknowledgments. According to further studies, such performance has been allocated to increased common HPNP concentration, the cooperation between more than one center of metals and the stability being high [139,140,141].

2.5.3. Metal–Organic Framework-Based

Countless MOFs based on Zr are used as imitates of phosphor triesterase for the occurrence of cleavage of the bond of phosphate ester of CWA which is abbreviated as chemical warfare agent [142,143,144,145,146,147,148,149,150]. This was because of the similarity between their structures.

2.6. Other Enzyme Mimics

Other than hydrolysis and redox reactions, many reactions attained huge importance [151,152,153,154,155,156,157]. For example, other than peroxidase and the imitates of hydrolase discussed prior, an action like hydrogenase would likewise be figured out, insofar as giving MOFs photon ingestion specialists (porphyrin) and proton ingestion lessening operators (PtNPs) [158,159,160]. Moreover, the synthesis of MOFs with carbonic anhydrase limits the dangerous atmospheric deviation issue [161].
Furthermore, Chmielewski et al. revealed that the assembly of electrostatics, the peptide parts of trimethylammonium working AuNPs, could advance the ligating of peptides that are two in number, which resulted in inorganic implemented nanoparticles favorable in the biopolymers polymerization [162]. Morse et al. also illustrated the monolayer AuNPs functionalization which could mimic silicatein.

2.6.1. Single-Substrate Mechanism of Nanozymes

Such types of nanozymes only show a reaction with one substrate. Certain nanozymes were revealed by imitating the action of enzymes while giving a platform which is water-soluble. Basic functionalized groups are moored with cooperation onto different stages of nanoscale for synergist responses. Afterward, multivalent components such as metal ions came into consideration and have developed, expanding utilization in the biomedical field. Representative nanozymes along with the one-substrate mechanism would be sorted into a few types, dependent on responses and the sorts of enzymes occurring naturally.
This mechanism usually displays the kinetic profile which shows catalysis by Michaelis–Menten, in which catalysis has two stages, that is, the authoritative and response stages. While plotting the velocity of the reaction as a function of the concentration of substrate, kcatand KM are commonly determined to characterize nanozyme movement.
The nanozymes showing a single substrate mechanism generally include:
  • Hydrolase;
  • Peroxidase;
  • Superoxide dismutase;
  • Oxidase;
  • Catalase [26].

2.6.2. Nanozymes with the Multi-Substrate Mechanism

The advancement of nanotechnology and the comprehension of artificial enzymes have amassed, and their mechanism of multiple enzyme-like activities has been recognized. Representative nanozymes that follow one or more substrates or work differently under different situations, such as pH esteems, hydrogen peroxide or glutathione concentrations, and oxygenation levels, are listed in this section. These components may altogether impact the practices of nanozymes, which are particularly valid for the organic microenvironment at disease sites like cancer.
The multi-substrate mechanism can be depicted in:
  • Metal-based nanozymes;
  • Cu2O nanozymes;
  • Nanoceria;
  • Melanin nanoparticles;
  • Prussian blue nanoparticles [26].

3. Synthesis of Nanozymes

3.1. Nanozyme Production

The nanomaterials which are catalytic possess different properties in comparison with natural enzymes [163]. The activities of the nanozymes depend on the size of the particle, structure, and its shape which is affected by the coatings, charges, and external fields [164,165].

3.2. Hydrothermal and Solvothermal Methods

The techniques which are very promising for synthesizing the nanomaterials are the hydrothermal and solvothermal methods. Nanocrystals of low cost with well-controlled dimensions can be obtained by utilizing the proposed methods [164,166].
A series of nano crystals which are spinel-type were synthesized by using the method of solvothermal, where the solvent used was ethylene glycol. The obtained nanozymes were utilized as enzyme mimics for the detection of hydrogen peroxide. For example, two types of carbon-based nano catalysts with a size of 100–150 nm were synthesized by utilizing a combination of two methods, a thermal method and a solid-state reaction, from the zeolitic imidazolate framework-8 (ZIF-8) [167,168]. The carbon cubic nanomaterial with the hollow structure was procured by chemically etching ZIF-8 along with tannic acid, stuck to it by a calcination process. The carbon cubic nanomaterial with the porous surface was acquired by direct pyrolysis [169].
Electrochemical observation of glucose and fructose formed on gold nanoparticles (AuNPs) placed onto graphene paper has lately been presented. These nanostructures were formed by two techniques: thermal and laser de-wetting processes [170]. Gold nanostructures acquired by both methods exhibited major differences in their particle morphology. Both types of AuNPs were investigated by their capacity to oxidize glucose and fructose [171].

3.3. Chemical Reduction

Chemical reduction is a method which is used very frequently because of its rapidity and simplicity. This tool enables the formation of NPs in which the morphology and the size of particle distribution are managed by changing the molar concentration of the reactants, the reductant type, and the reaction temperature [172]. The important factor in achieving very high chemical reduction is choosing the suitable reductants. The reduction of metal salts needs reactivity of the agent which causes reduction to the redox potential of the metal. The procured particles are small if the reaction rate during the synthesis procedure is too fast [173]. Nevertheless, if the reaction rate is too slow, particle aggregation may happen [174]. The synthesis of hollow copper sulfidenanocubes (h-CuS NCs) was done via the chemical reduction method [175]. This method has been utilized for the synthesis of peroxidase (PO)-like nanozyme-based AuNPs along with Pseudomonas aeruginosa-specific aptamer [176].

3.4. Sol–Gel Method

In the sol–gel method, a gel-like network containing liquid and solid phase is formed. The crystallinity, morphology, and magnetic properties of the nanozymes can be managed by choosing a suitable complexing agent, concentration and type of chemical additive, and temperature conditions [177]. The synthesis of PtNPs polyaniline (PAni) hydrogel heterostructures was produced with the sol–gel method [178]. Phytic acid was utilized as a complexing agent. The PtNPs loaded into the hydrogel matrix act as active catalysts for the oxidation of hydrogen peroxide. The acquired PtNP/PAni hydrogel had a 3D hierarchical structure consisting of connected PAni nanofibers with diameters of approximately 100 nm). The porous structure of the PAni hydrogel allows immobilization of concentrated enzyme solutions. Since water-soluble molecules can penetrate through the hydrogel, the PtNPs preserve their ability to catalyze glucose oxidation [179].

3.5. Co-Precipitation

Co-precipitation is a quick technique for the amalgamation of various sorts of nanocatalysts. Co-precipitation is a superb method to use when higher virtue and better stoichiometric control arerequired. Dashtestani et al. utilized a mix of two strategies for nanocompositeunion: reduction of HAuCl4 chemically and co-precipitation of the acquired gold nanoparticles with the copper (II) complex of cysteine (GNPs/Cu-Cys). The mix of GNPs and the Cu-Cyscomplex expanded the electrochemical sign toward O2 [180,181].

3.6. Electrochemical Deposition

Electrochemical deposition is a minimal-effort strategy for acquiring metalnanocatalysts. In any case, it is normally utilized less regularly than synthetic decrease strategies. The interaction is straightforward and incorporates a drenching of a conductive surface into an answer containing particles of the material to be saved and the use of a voltage across the strongelectrolyte interface. Throughout this strategy, a reaction of charge transfer causes the deposition of film [182,183].

3.7. Polymerization and Polycondensation

Nanozymes can be acquired either by utilizing insoluble polymers or by cross-connecting of a solvent polymer. Santhosh et al. blended composite center shell nanofibers comprising gold nanopartilces on poly (methylmethacrylate) by the mix of an electrospinning procedure, and furthermore, the in situ polymerization of aniline [184,185].

4. Nanozymes from Challenges to Opportunities

There has been a huge development observed in the field of nanozymes over the last few years, as shown in Figure 2. There has even been a considerable increase in the number of nanozymes along with the reaction. Enzymes are found to depict some of the basic characteristics such as high substrate specificity and excellent activity. Besides this, there are a variety of nanozymes which did not achieve the level of composure. Such limitations are served as a better opportunity for advanced development and research [186].
Types of Nanozymes
Many studies are describing different types of nanozymes (e.g., Fe3O4 nanoparticles were observed to possess peroxidase-like activity), indicating their ability to catalyze the substrate oxidation by using hydrogen peroxide [187,188]. Due to the biocompatible and magnetic qualities of Fe3O4, it is interesting and can be used in theragnostic in vivo procedures [189]. Later on, it was observed that many nanoparticles possess the same peroxidase-like activity [190]. Nanoceria consists of different types of enzyme mimics such as superoxidase dismutase (SOD), oxidase, and catalase [191]. Gold NPs can mimic glucose oxidase.

4.1. Biosensors

Electrochemical, calorimetric, and fluorescence detecting are the traditional procedures for the evaluation of estimated constituents through a corresponding difference in organic systemization and are further broadly utilized for the recognition of biomolecules [192]. Among these, the ELISA, conventional calorimetric discovery, is utilized for distinguishing exceptionally small amounts of the wanted substance [193]. The biosensors which are based on nanozymes are effectively created to recognize ions.

4.2. Detection of H2O2

It is linked generally along with transduction of single and cell growth. Excessive production of H2O2 may lead to an increased risk of many inflammatory infections such as lung diseases, atherosclerosis, hepatitis, etc. [194,195]. The detecting of H2O2 is important because of its significance in the field of biology and medicine. The iron oxide MNPs working as imitates of peroxidase were used to initially detect hydrogen peroxide with the chromogenic 2,2′-azino-bis (3-ethylbenzothiazoline-6-sulfonic acid) (ABTS) [196]. Various peroxidase nanozymes have been designed for calorimetric detection of H2O2 [197,198].

4.3. Detection of Glucose

These have attained a lot of consideration in the last several years due to their broad employments in clinical examination, biomedical sciences, food creation, and biology [199,200]. Glucose and comparative parts that can form hydrogen peroxide by the synergist response could be detected as per particular peroxidase and oxidase mimics [201]. A method for direct physical adsorption development of electrochemical biosensing of glucose was accomplished. This planned biosensing showed selectivity which was high and had sample feasibility [200].

4.4. Metal Ion Sensing

Many studies focus generally on the uses for sensing [201,202]. During the underlying work around there, a particular and sensitive sensor was created to identify Cu2+ utilizing Cu2+. The magnetic and nanotube silica NPs of carbon were used to create the extremely delicate sensors to discover the exceptionally low Cu2+ quantity [203]. In another examination, the platinum NPs (2 nm) with cow-like serum egg whites were created to examine imitating peroxidase action. This was useful for building up specific and touchy sensation for identification of Hg which has a straight identification of 0–120 nM of range [204].

4.5. Nucleic Acid Sensing

A few methodologies for the recognition of nucleic acids are created by the usage of nanozymes [205,206]. A test was created to identify bacterial DNA utilizing Fe3O4 MNPs to examine the checking of microorganisms in faucet water [207]. The proclivity towards different nanozymes is distinctive for abandoned DNA which is single as well as twofold abandoned DNA. Calorimetric technique is produced for the discovery of DNA through adjustment of peroxidase mimicking movement of “Au” nanoparticles on graphene sheets [208].

4.6. Aptasensors

These are used for building aptasensors for the little molecules which are bioactive and proteins [209,210]. Utilizing the AuNPs having peroxidase-like activity alongside high particularity and acetamiprid’s affinity explicit S-18 aptamers, a calorimetric assay was intended for pesticide quick checking [211]. The acetamiprid presence inside the sample could communicate along the aptamer which prevents binding and also helps in recovery.

4.7. Pollutant Detection

Melamine, a nitrogenous natural compound that becomes toxic when taken and has been illegally used in dairy products, was identified using a fast and efficient calorimetric approach [212]. The compound is found to work by repressing the reactant ABTS oxidation by NPs with hydrogen peroxide, yet it emphatically responds with it and creates compounds. The nanozyme-based strategies have been so far successful, straight forward, and cost-accommodating for mineralizing and debasing the natural colors of mechanical strategies. Most prominently, MNPs like peroxidase were considered to debase the natural pollutants. The degrading procedure gives a recognizable advantage on degradation methodology for extraordinary strength and diminished expense. The MNPs/H2O2 can effectively achieve the evacuation of 85% of phenolic mixes from the fluid arrangement in 3 h [213]. Degradation based on MNPs showed higher viability. It was discovered that 96% of this color could be degraded inside 15 min utilizing improved conditions [214].

4.8. Nanozyme-Based Immunoassays

Many considerable efforts have been made for designing immunoassaying with the help of nanozymes [215,216]. Various configurations were used for immunoassays utilizing the nanozymes as the signaling parts. For example, the sandwich immunoassay and antigen down (AD) immunoassay have been revealed [217]. Later on, various researchers executed the standard sandwich immunoassay for identification by using nanozyme mimics of peroxidase and oxidase [215]. For example, the preparation of imitates of peroxidase with enhanced activity was conducted [218].

5. Nano-Enzymes Role in Diseases

5.1. Cancer Diagnostics and Therapy

To detect cancer cells, nanozymes are being implicated [219]. Various substrates that are organic undergo catalytic oxidation which has been shown by cerium oxides NPs [220]. These were introduced for the immuno detection of cancer cells regarding the unique capability of folate conjugation. According to various studies, the folate receptor which is over-expressed onto the cancer cells is being detected selectively by folate. With the dissimilarity of ELISA, immunoassays based on the oxide of cerium manifested various benefits. However, these techniques which conventionally need the support of various antibodies may have some limitations when antibodies are denatured on the cancer cell surface. Secondarily, durability-like shortcomings are exhibited. Hence, when it is denatured, it may result in losing its original catalytic activity. Many researchers have said that NPs of cerium oxide are not considered as imitates of oxidase. Regardless of this, it is assisted as the catalyst of oxidation. Further, some people learned about this by nanoprobes [221]. Other than this, the nanocomposite was introduced by nanoparticle development which is gold [199]. Peroxidase can be mimicked by the nanocomposites which are formed.
The detection of cancer cells is implicated by nano-enzymes [219]. Figure 3 shows the detection of cancer cells with calorimetric strategy by using PtNPs/GO nanozymes. Regulation of gene process is majorly served by RNA interference [222,223]. A group introduced nanozymes which are similar to machinery, based on the structural characteristics and functional characteristics of this system which areused for the target RNA to be cleaved [224]. In this particular technique, gold nanoparticles act as the keystone of the nanozymes that offers a modification of DNA which are single-stranded oligonucleotides and endonucleases that are non-specific. The resulting nanocomposites indicated RNA-DNA nanoparticles. It was found that nanoparticles may inhibit virus replication and gene expression silencing. Diseases with expressions of proteins such as infections caused by viruses or cancers are associated with nanozymes. Photodynamic therapy employs nanozymes under hypoxia [225]. Singlet oxygen is formed from the tumor tissues by organic metal frameworks based on photodynamic therapy [226,227]. Tumor tissues are also having hypoxic conditions that are restricted by the therapeutic properties of photodynamic therapy [228]. The new technique was introduced for the photodynamic therapy through some modifications, and for this, oxygen was generated by the catalase mimics, which is PtNPs. On to the surface of organic–metal frameworks where Pt NPs are assembled, the production of oxygen which is single can be raised by the nanocomposites through the decomposition in the hypoxia’s tissue of hydrogen peroxide. Under the hypoxic environment, the cancer treatment is effectively served by nanozyme integrated organic–metal frameworks [229]. Malignant tumors are treated with certain approaches, and one of the promising approaches is apoptosis induced with nanozymes [230]. The treatment systems which are presently used require the support of ROS and oxygen under conditions of tumor. A successful nano-flower is biomimetic and was designed by assembly of various nanozymes [231]. The PtCo-NPs in the acidic tumor produce toxic ROS by oxidase mimics, whereas the MnO2 nanomaterials possess good catalase-like functions [232].

5.2. Neuroprotection

Various researchers have been developing the application for SOD imitates for guiding free radical destruction [233,234]. Afterwards, the C60[C(COOH)2]3 was introduced as an imitating agent of SOD, which shows therapeutic actions in knockout mice [235]. In SOD2 mice not able to express the manganese SOD in mitochondria, the period of life was increased. Nanoceria was introduced to imitate the SOD and also show various functions of neuroprotection [236]. Confirmation was done about nanoceria that it can do the aversion of the cells of the neurons present in the retina by the destruction of ROS [237]. In Alzheimer’s disease, RNS peptides and amyloid-beta are involved. The treatment for this disease is missing for the antioxidants. Microscopic studies suggested that the neurons internalize the nanoceria that is present. It can perform the RNS scavenging and hence results in the protection of neurons that are degenerated. Studies have suggested that nanoceria is used for the protection of the neurons from hyperphosphorylation or their death [238].

5.3. Antioxidation

The cell metabolism has certain by-products which are O2, OH, and H2O2 [239,240]. The ROS contributes to various signaling mechanisms and when the level of ROS is considered low, they act as second messengers which are significant [241]. On the other hand, if the ROS levels are exceeded, then they result in the damage of proteins, DNA, lipids, and other various molecules. Moreover, for activating the apoptosis of cells, the caspase can be induced by them [242]. Various pathological disorders are linked with ROS, for example: kidney diseases, diabetes, arthritis, cancer, and atherosclerosis [243]. It is known that ROS affects human health and life critically by participating in aging, human diseases, and death. Hence, for maintaining the intracellular redox homeostasis, the levels of ROS need to be regulated majorly. Within the cell system, various types of enzymes are antioxidants, for example: catalase, glutathione peroxidase, SOD, etc. An important function is being performed by the antioxidant enzymes which are maintaining the cellular redox balance. However, ROS overexpression can decrease the activities of enzymes which are under pathological situations [244].

5.4. Anti-Inflammatory

Inflammation is the state which involves two types of inflammation: acute and chronic inflammation [245]. The precursor to various diseases is certainly the response of inflammation. Hence, the emergence of various associated disorders can be treated successfully by treating inflammation, for example, diseases associated with heart and cancer. The quality of increase in the ROS is notable in the tissue of inflammation. The inflammation is alleviated by scavenging ROS along with their inhibition of diseases. From the time of the formation, the Prussian blue has been working as blue dye [246]. The Prussian blue has been employed as an antidote for thallotoxicosis in clinical trials because of its brilliant biosecurity along with the biocompatibility as recommended by the FDA [247]. For the treatment of cancer, the PB-NPs pay a role in photo thermal therapy, ultrasound imaging, and magnetic resonance imaging [248,249]. The nanoparticles PB have been discovered which possesses multi-enzyme-like functions, which can successively reduce the levels of ROS which are inside the cell and also attain the cytoprotecting efficacy [250]. It was noted that the feature of scavenging ROS was due to the attraction towards the hydroxide along with the mimicking of the enzymes. The altered immune response against the infection causes organ dysfunction, which is a great threat to life known as sepsis [251]. The systemic inflammation occurred by the microbe local infection which is further accompanied by fever, and because of host defense mechanisms, white blood cells increase in number [252]. If, within a short period, the treatment is not done, then the immune response becomes disordered which would cause multiorgan dysfunction, pro-inflammatory cytokines, and even death. Hence, if we inhabit the abnormal inflammatory response, then it would kill the bacteria [253].

5.5. Anti-Aging

Data have suggested that aging is somewhat linked with major mechanisms of redox, such as ROS detoxifying and the response of cells to the macromolecules which are damaged oxidatively. Amice brain slice was set up for imaging, and the results showed that its treatment with nanozymes slowed down the age associated with the damage within the tested region. Using EPR techniques, it was known that the nanozymes were in action inside the powerhouse of the cell, as the primary cellular source for ROS is reported to be mitochondria. Nanozymes have an impact on age-linked loss of memory which was determined, exhibiting nanozymes’ potential for securing the age-linked cognitive damage in the mice [254].

6. Biomedical Application of Nanozymes

Nanozymes are found to show a greater significance in the field of biomedicine as well as industry. Numerous nanozymes have just demonstrated serious adversaries to the enzymes which are naturally occurring and which they imitate. The developing disclosure and more profound comprehension of nanozyme systems have empowered various applications linked with the biomedical industry.

6.1. Nanozymes Acting by Themselves

They were found in calorimetric and biosensing assays for immediate substitutes of natural enzymes. They comprised extra functions regularly which were not offered by the natural enzymes. As it is among the most utilized strategies for the detection of biomarkers, ELISA ordinarily utilizes horseradish peroxidase (HRP) for oxidizing 3,3′,5,5′-tetramethylbenzidinel (TMB) for color improvement and ensuring measurement. In any case, HRP is restricted for a scope of pH, concentration, and temp because of its instability in worse conditions and significant expense. HRP in the biosensing processes can be challenged by various categories of the nanozymes which include nanoceria, (MNPs), (GO), and other peroxidase-imitating nanozymes. Furthermore, they have invaluable natural functions that would additionally encourage research [26]. Nanozymes permit biosensing applications custom-fitted for their particular properties, giving essential refinement using attraction [255]. MoO3 nanozymes were discovered for the detoxification of cells done valuably by imitates of sulfite oxidase. The absence of this imitate was related to the neurological harm and youth demise [256].

6.2. Synergistic Nanozymes

Although nanozymes were originally found for the free action of enzyme, attempts in recent times have already been conducted more profoundly. Revising basic attributes linked with an environment, for example, H2O2 concentration, pH, and oxygenation levels, may shed light on nanozyme exhibitions. Therefore, the subsequent therapeutic and diagnostic role of nanozymes can be better adjusted.
In light of the inherent peroxidase-imitating functions of GO, Qu, and other members, a method was set up for the estimation of the concentration of glucose [80]. Zhong and others furthermore built up the catalytic cascade utilizing AuNP nanosheets; hence, the previous includes Goximitating function and can go about as a natural peroxidase [257]. MOF fills in for layout to develop and accept hydrogen peroxide.
Xia and co-workers built up the system of nanozyme which is 3 in 1, which includes detection and catalysis utilizing CD/AUNP [258]. Further, in 2017 Shi, Chan, and co-workers led to the development of therapy by using graphene oxidase for the in vitro treatment of neoplasm [259].

6.3. Remote Control Nanozymes

These are the types of nanozymes which are controlled and are responsible for synergistic effect.
Thus, these functions were present with accentuation in techniques for improvement concerned with the future with superb transience and exactness. Light has been described as the utilized strategy for controlling the synergist response. It may very well be delivered with high precision and controllability. Different effects of the nanozymes can be triggered by tuning the wavelength. Prinsand co-workers led to the development of light-regulating AUNP nanozymes for the hydrolysis of RNA [260].
Inflammation is commonly linked with reactive oxygen species (ROS). Propelled by common photosynthesis and the way by which hydrogen gas may lead to the reduction of ●OH to H2O, a multi component framework has been gathered by Chia, Sung, and co-workers to create hydrogen gas in nearness to the site of inflammation in mice [261]. Heat is considered to be another stimulant for triggering them. Nieand others built up the method of amplifying signal-free enzymes. They used Au capsules as imitates for calorimetric assay of the disease [262]. Further, Qu and co-workers designed a heat-recovering rationale entryway by utilizing nanoceria as a signal transducer [263]. Other than heat and light, nanozymes utilized ultrasound as the boost. Yeh and others detailed a hydrogen peroxide-encapsulated Fe3O4-PLGA polymer nanozyme framework for malignancy treatment [264].

7. Future Perspectives for Nanozymes

Intending to peruse nanozymes, one has to have a vital source of innovation through productively conquering disadvantages of enzymes which are natural, and accompanying proposals are offered. There is a requirement of the advancement of fresh nanozymes comprised of high movement and customary examination functions; further has exploration followed a technique of screening of sound action dependent on the nuclear arrangements which were conceived for catalyzing the response of enzyme. The process to prepare normal composites for identifying the present significant limitations by adjusting synergistic effects for facilitating electron transfer between composite materials during redox reaction has also been started. Bioinspired synthesis of nanozymes additionally gives an alternative to prepare non-toxic nanozymes by successfully going around the utilization of poisonous synthetic compounds in traditional substance combination, accordingly quickening their use in therapeutic application. At last, the turn of events of novel surface designing innovation could specifically target the substrates by nanozymes and would be of great significance [23]. More of these developments would open up new avenues for single-stage sensors and theragnostic, which could be helpful in various biosensing and biomedical applications. The vast majority of the nanozymes are accounted for to show their synergist movement by redox action by surface iotas. Be that as it may, the reactant movement might be additionally improved by controlling the center of the nanozymes by doping with some uncommon earth components. Such procedures would add more redox “problem areas” for synergist action and along these lines upgrade the action of nanozymes. In contrast to characteristic catalysts, the size and synthesis of most nanozymes are not uniform, except for fullerene-based nanozymes. Further, group to-cluster variety fit as a fiddle of nanoparticles/nanozymes, and consequently adjustments in physicochemical properties, requires expanded spotlight on improving the union convention to create the monodispersed nanozymes with molecularly exact designs [264].

8. Conclusions

The enzyme-mimicking properties of nanoparticles have proved to be significant in medicine, industry, and healthcare. Certain types of nanozymes such as peroxidase mimics, superoxide mimics, catalase mimics, etc., have contributed to various applications and emerging opportunities. Different mechanisms of nanozymes such as single substrate and multi substrate have been studied. Nanozymes were found to have various options in the field of biosensors, apt sensors, glucose and hydrogen peroxide detection, nanozyme based immunoassay, etc. Other than this, nanozymes have also played a great role in the healthcare system in cancer diagnosis and treatment, anti-aging, neuroprotection, etc. Further, biomedical applications such as self-acting nanozymes, synergistic nanozymes, etc., have also been considerably studied, which lead to various therapeutic effects.

Author Contributions

Conceptualization, R.K.S., A.N. and P.K. (Prabhjot Kaur); writing—original draft preparation, R.K.S., M.S., H.S., P.K. (Prabhjot Kaur) and M.H.R.; investigation, P.K. (Parneet Kaur), P.K. (Prabhjot Kaur) and M.J.-S.; resources, M.S., S.C., A.N. and P.K. (Parneet Kaur); writing—review and editing, M.H.R., R.K.S. and S.C.; supervision, A.N. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

We wish to confirm that there are no known conflicts of interest associated with this publication and there has been no significant financial support for this work that could have influenced its outcome.

References

  1. Sindhu, R.K.; Kaur, H.; Kumar, M.; Sofat, M.; Yapar, E.A.; Esenturk, I.; Kara, B.A.; Kumar, P.; Keshavarzi, Z. The ameliorating approach of nanorobotics in the novel drug delivery systems: A mechanistic review. J. Drug Target 2021, 29, 822–833. [Google Scholar] [CrossRef]
  2. Cheng, M.H.; Rosentrater, K.A.; Sekhon, J.; Wang, T.; Jung, S.; Johnson, L.A. Economic feasibility of soybeanoil production by enzyme-assisted aqueous extraction processing. Food Bioprocess Technol. 2019, 12, 539–550. [Google Scholar] [CrossRef]
  3. Kasar, S.S.; Giri, A.P.; Pawar, P.K.; Maheshwari, V.L. A Protein α-amylase inhibitor from Withania somnifera and its role in overall quality and nutritional valuei mprovement of potato chips during processing. Food Bioprocess Technol. 2019, 12, 636–644. [Google Scholar] [CrossRef]
  4. Osete-Alcaraz, A.; Bautista-Ortín, A.B.; Ortega-Regules, A.E.; Gómez-Plaza, E. Combined use of pectolytic enzymes and ultra sounds for improving the extraction of phenolic compounds during vinification. Food Bioprocess Technol. 2019, 12, 1330–1339. [Google Scholar] [CrossRef]
  5. Wubshet, S.G.; Wold, J.P.; Afseth, N.K.; Böcker, U.; Lindberg, D.; Ihunegbo, F.N.; Måge, I. Feed-Forward Prediction of Product Qualities in Enzymatic Protein Hydrolysis of Poultry By-products: A Spectroscopic Approach. Food Bioprocess Technol. 2018, 11, 2032–2043. [Google Scholar] [CrossRef]
  6. Zhang, L.; Li, C.-Q.; Jiang, W.; Wu, M.; Rao, S.-Q.; Qian, J.-Y. Pulsed Electric Field as a Means to Elevate Activity and Expression of α-Amylase in Barley (Hordeum vulgare L.) Malting. Food Bioprocess Technol. 2019, 12, 1010–1020. [Google Scholar] [CrossRef]
  7. Sun, D.W. (Ed.) Handbook of Food Safety Engineering; Wiley Blackwell: Hoboken, NJ, USA, 2011. [Google Scholar]
  8. Patra, J.K.; Das, G.; Fraceto, L.F.; Campos, E.V.R.; del Pilar Rodriguez-Torres, M.; Acosta-Torres, L.S.; Diaz-Torres, L.A.; Grillo, R.; Swamy, M.K.; Sharma, S.; et al. Nano based drug delivery systems: Recent developments and future prospects. J. Nanobiotechnol. 2018, 16, 71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Liu, Z.; Tabakman, S.; Welsher, K.; Dai, H. Carbon nanotubes in biology and medicine: In vitro and in vivo detection, imaging and drug delivery. Nano Res. 2009, 2, 85–120. [Google Scholar] [CrossRef] [Green Version]
  10. Orive, G.; Gascon, A.R.; Hernández, R.M.; Domínguez-Gil, A.; Pedraz, J.L. Techniques: New approaches to the delivery of biopharmaceuticals. Trends Pharmacol. Sci. 2004, 25, 382–387. [Google Scholar] [CrossRef]
  11. Razzacki, S.Z.; Thwar, P.K.; Yang, M.; Ugaz, V.M.; Burns, M.A. Integrated microsystems for controlled drug delivery. Adv. Drug Deliv. Rev. 2004, 56, 185–198. [Google Scholar] [CrossRef]
  12. Mirza, A.Z.; Siddiqui, F.A. Nanomedicine and drug delivery: A mini review. Int. Nano Lett. 2014, 4, 94. [Google Scholar] [CrossRef] [Green Version]
  13. Jahangirian, H.; Lemraski, E.G.; Webster, T.J.; Rafiee-Moghaddam, R.; Abdollahi, Y. A review of drug delivery systems based on nanotechnology and green chemistry: Green nanomedicine. Int. J. Nanomed. 2017, 12, 2957–2978. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Lam, P.-L.; Wong, W.-Y.; Bian, Z.; Chui, C.-H.; Gambari, R. Recent advances in green nanoparticulate systems for drug delivery: Efficient delivery and safety concern. Nanomedicine 2017, 12, 357–385. [Google Scholar] [CrossRef]
  15. Antonescu (Mintas), A.-I.; Miere (Groza), F.; Fritea, L.; Ganea, M.; Zdrinca, M.; Dobjanschi, L.; Antonescu, A.; Vicas, S.I.; Bodog, F.; Sindhu, R.K.; et al. Perspectives on the Combined Effects of Ocimumbasilicum and Trifolium pratense Extracts in Terms of Phytochemical Profile and Pharmacological Effects. Plants 2021, 10, 1390. [Google Scholar] [CrossRef] [PubMed]
  16. Rodrigues, T.; Reker, D.; Schneider, P.; Schneider, G. Counting on natural products for drug design. Nat. Chem. 2016, 8, 531–541. [Google Scholar] [CrossRef] [PubMed]
  17. Siddiqui, A.A.; Iram, F.; Siddiqui, S.; Sahu, K. Role of natural products in the drug discovery process. Int. J. Drug Dev. Res. 2014, 6, 172–204. [Google Scholar]
  18. Silva, P.; Bonifácio, B.; Ramos, M.; Negri, K.; Bauab, T.M.; Chorilli, M. Nanotechnology-based drug delivery systems and herbal medicines: A review. Int. J. Nanomed. 2013, 9, 1–15. [Google Scholar] [CrossRef] [Green Version]
  19. Mohanty, S.K.; Swamy, M.K.; Sinniah, U.R.; Anuradha, M. Leptadeniareticulata (Retz.) Wight & Arn. (Jivanti): Botanical, agronomical, phytochemical, pharmacological, and biotechnological aspects. Molecules 2017, 22, 1019. [Google Scholar]
  20. Beutler, J.A. Natural products as a foundation for drug discovery. Curr. Prot. Pharmacol. 2009, 46, 9–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Thilakarathna, S.H.; Rupasinghe, H.P.V. Flavonoid Bioavailability and Attempts for Bioavailability Enhancement. Nutrients 2013, 5, 3367–3387. [Google Scholar] [CrossRef]
  22. Watkins, R.; Wu, L.; Zhang, C.; Davis, R.M.; Xu, B. Natural product-based nanomedicine: Recent advances and issues. Int. J. Nanomed. 2015, 10, 6055. [Google Scholar]
  23. Shin, H.Y.; Park, T.J.; Kim, M.I. Recent Research Trends and Future Prospects in Nanozymes. J. Nanomater. 2015, 2015, 1–11. [Google Scholar] [CrossRef] [Green Version]
  24. Liu, X.; Wu, J.; Liu, Q.; Lin, A.; Li, S.; Zhang, Y.; Wang, Q.; Li, T.; An, X.; Zhou, Z.; et al. Synthesis temperature regulated multi enzyme mimicking activities of ceria nanozymes. J. Mater. Chem. B 2021, 9, 7238. [Google Scholar] [CrossRef] [PubMed]
  25. Wang, P.; Wang, T.; Hong, J.; Yan, X.; Liang, M. Nanozymes: A New Disease Imaging Strategy. Front. Bioeng. Biotechnol. 2020, 8, 15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Jiang, D.; Ni, D.; Rosenkrans, Z.T.; Huang, P.; Yan, X.; Cai, W. Nanozyme: New horizons for responsive biomedical applications. Chem. Soc. Rev. 2019, 48, 3683–3704. [Google Scholar] [CrossRef]
  27. Zhang, X.; Li, G.; Chen, G.; Wu, D.; Wu, Y.; James, T.D. Enzyme Mimics for Engineered Biomimetic Cascade Nanoreactors: Mechanism, Applications, and Prospects. Adv. Funct. Mater. 2021, 2106139. [Google Scholar] [CrossRef]
  28. Wu, J.; Wang, X.; Wang, Q.; Lou, Z.; Li, S.; Zhu, Y.; Qin, L.; Wei, H. Nanomaterials with enzyme like characteristics (nanozymes): Next-generation artificial enzymes(II). Chem. Soc. Rev. 2019, 48, 1004–1076. [Google Scholar] [CrossRef]
  29. Munir, S.; Shah, A.A.; Rahman, H.; Bilal, M.; Rajoka, M.S.R.; Khan, A.A.; Khurshid, M. Nanozymes for medical biotechnology and its potential applications in biosensing and nanotherapeutics. Biotechnol. Lett. 2020, 42, 357–373. [Google Scholar] [CrossRef]
  30. Li, J.; Zhang, C.; Lin, J.; Yin, J.; Xu, J.; Chen, Y. Evaluating the bioavailability of heavy metals in natural-zeolite-amended aquatic sediments using thin-film diffusive gradients. Aquac. Fish. 2018, 3, 122–128. [Google Scholar] [CrossRef]
  31. Mutharaian, V.N.; Kamalakannan, R.; Mayavel, A.; Makesh, S.; Kwon, S.H.; Kang, K.-S. DNA polymorphisms and genetic relationship among populations of Acacia leucophloea using RAPD markers. J. For. Res. 2017, 29, 1013–1020. [Google Scholar] [CrossRef]
  32. Qiu, H.; Pu, F.; Ran, X.; Liu, C.; Ren, J.; Qu, X. Nanozyme as Artificial Receptor with Multiple Readouts for Pattern Recognition. Anal. Chem. 2018, 90, 11775–11779. [Google Scholar] [CrossRef] [Green Version]
  33. Yan-Yan, H.; You-Hui, L.; Fang, P.; Jin-Song, R.; Xiao-Gang, Q. The current progress of nanozymes indisease treatments. Prog. Biochem. Biophys. 2018, 45, 256–267. [Google Scholar]
  34. Huang, L.; Sun, D.W.; Pu, H.; Wei, Q. Development of Nanozymes for Food Quality and Safety Detection: Principles and Recent Applications. Compr. Rev. Food Sci. Food Saf. 2019, 18, 1496–1513. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Song, S.; Liu, Y.; Song, A.; Zhao, Z.; Lu, H.; Hao, J. Peroxidase mimetic activity of Fe3O4 nanoparticle prepared based on magnetic hydrogels for hydrogen peroxide and glucose detection. J. Colloid Interface Sci. 2017, 506, 46–57. [Google Scholar] [CrossRef] [PubMed]
  36. Wei, H.; Wang, E. Fe3O4 Magnetic Nanoparticles as Peroxidase Mimetics and Their Applications in H2O2 and Glucose Detection. Anal. Chem. 2008, 80, 2250–2254. [Google Scholar] [CrossRef] [PubMed]
  37. André, R.; Natálio, F.; Humanes, M.; Leppin, J.; Heinze, K.; Wever, R.; Schröder, H.-C.; Müller, W.E.G.; Tremel, W. V2O5 Nanowires with an Intrinsic PeroxidaseLike Activity. Adv. Funct. Mater. 2011, 21, 501–509. [Google Scholar] [CrossRef]
  38. Natalio, F.; Andre, R.; Hartog, A.F.; Stoll, B.; Jochum, K.P.; Wever, R.; Tremel, W. Vanadium pentoxide nanoparticles mimic vanadium halo peroxidases and thwart biofilm formation. Nat. Nanotechnol. 2012, 7, 530–535. [Google Scholar] [CrossRef] [PubMed]
  39. Nie, G.; Zhang, L.; Lei, J.; Yang, L.; Zhang, Z.; Lu, X.; Wang, C. Monocrystalline VO2 (B) nanobelts: Largescale synthesis, intrinsic peroxidase like activity and application in biosensing. J. Mater. Chem. A 2014, 2, 2910–2914. [Google Scholar] [CrossRef]
  40. Xie, J.; Zhang, X.; Jiang, H.; Wang, S.; Liu, H.; Huang, Y. V2O5 nanowires as a robust and efficient peroxidase mimic at high temperature in aqueous media. RSC Adv. 2014, 4, 26046–26049. [Google Scholar] [CrossRef]
  41. Niu, X.; He, Y.; Li, X.; Song, H.; Zhang, W.; Peng, Y.; Pan, J.; Qiu, F. Trace Iodide Dramatically Accelerates the Peroxidase Activity of VOx at ppb Concentration Levels. Chem. Sel. 2017, 2, 10854–10859. [Google Scholar] [CrossRef]
  42. Tian, R.; Sun, J.; Qi, Y.; Zhang, B.; Guo, S.; Zhao, M. Influence of VO2 Nanoparticle Morphology on the Colorimetric Assay of H2O2 and Glucose. Nanomaterials 2017, 7, 347. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Huang, L.; Zhu, W.; Zhang, W.; Chen, K.; Wang, J.; Wang, R.; Yang, Q.; Hu, N.; Suo, Y.; Wang, J. Layered vanadium(IV) disulfide nanosheets as a peroxidase like nanozyme for colorimetric detection of glucose. Microchim. Acta 2017, 185, 7. [Google Scholar] [CrossRef] [PubMed]
  44. Wu, Y.S.; Huang, F.F.; Lin, Y.W. Fluorescent Detection of Lead in Environmental Water and Urine Samples Using Enzyme Mimics of Catechin Synthesized Au Nanoparticles. ACS Appl. Mater. Interfaces 2013, 5, 1503–1509. [Google Scholar] [CrossRef] [PubMed]
  45. Zhu, R.; Zhou, Y.; Wang, X.-L.; Liang, L.-P.; Long, Y.-J.; Wang, Q.-L.; Zhang, H.-J.; Huang, X.-X.; Zheng, H.-Z. Detection of Hg2+ based on the selective inhibition of peroxidase mimetic activity of BSAAu clusters. Talanta 2013, 117, 127–132. [Google Scholar] [CrossRef] [PubMed]
  46. Deng, H.H.; Li, G.W.; Hong, L.; Liu, A.-L.; Chen, W.; Lin, X.-H.; Xia, X.-H. Colorimetric sensor based on dual-functional gold nanoparticles: Analyte-recognition and peroxidase-like activity. Food Chem. 2014, 147, 257–261. [Google Scholar] [CrossRef]
  47. Han, T.H.; Khan, M.M.; Lee, J.; Cho, M.H. Optimization of positively charged gold nanoparticles synthesized using a stainless-steel mesh and its application for colorimetric hydrogen peroxide detection. J. Ind. Eng. Chem. 2014, 20, 2003–2009. [Google Scholar] [CrossRef]
  48. Liu, Y.; Wang, C.; Cai, N.; Long, S.; Yu, F. Negatively charged gold nanoparticles as an intrinsicper oxidase mimic and their applications in the oxidation of dopamine. J. Mater. Sci. 2014, 49, 7143–7150. [Google Scholar] [CrossRef]
  49. Kaur, B.; Chaterjee, J.G.; Bruno, T.K. Sharma, Defining Target Product Profiles (TPPs) for AptamerBased Diagnostics. Adv. Biochem. Eng. Biotechnol. 2020, 174, 195–209. [Google Scholar] [PubMed]
  50. Drozd, M.; Pietrzak, M.; Parzuchowski, P.; Mazurkiewicz-Pawlicka, M.; Malinowska, E. Peroxidase like activity of gold nanoparticles stabilized by hyperbranched poly glycidol derivatives over a wide pH range. Nanotechnology 2015, 26, 495101. [Google Scholar] [CrossRef]
  51. Jiang, X.; Sun, C.; Guo, Y.; Nie, G.; Xu, L. Peroxidase like activity of apoferritin paired gold clusters for glucose detection. Biosens. Bioelectron. 2015, 64, 165–170. [Google Scholar] [CrossRef]
  52. Drozd, M.; Pietrzak, M.; Parzuchowski, P.; Malinowska, E. Pitfalls and capabilities of various hydrogen donors in evaluation of peroxidase-like activity of gold nanoparticles. Anal. Bioanal. Chem. 2016, 408, 8505–8513. [Google Scholar] [CrossRef] [Green Version]
  53. Jiang, C.; Zhu, J.; Li, Z.; Luo, J.; Wang, J.; Sun, Y. Chitosan–gold nanoparticles as peroxidase mimic and their application in glucose detection in serum. RSC Adv. 2017, 7, 44463–44469. [Google Scholar] [CrossRef] [Green Version]
  54. Zhu, X.; Mao, X.; Wang, Z.; Feng, C.; Chen, G.; Li, G. Fabrication of nanozymeDNA hydrogel and its application in biomedical analysis. Nano Res. 2017, 10, 959–970. [Google Scholar] [CrossRef]
  55. Singh, R.; Belgamwar, R.; Dhiman, M.; Polshettiwar, V. Dendritic fibrous nano-silica supported gold nanoparticles as an artificial enzyme. J. Mater. Chem. B 2018, 6, 1600–1604. [Google Scholar] [CrossRef]
  56. Sun, Z.; Zhang, N.; Si, Y.; Li, S.; Wen, J.; Zhu, X.; Wang, H. High-throughput colorimetric assays for mercury(ii) in blood and wastewater based on the mercury-stimulated catalytic activity of small silver nanoparticles in a temperature-switchable gelatin matrix. Chem. Commun. 2014, 50, 9196–9199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Priyadarshini, N.E. Pradhan, Gold nanoparticles as efficient sensors incolorimetric detection of toxic metal ions: A review. Sens. Actuators B Chem. 2017, 238, 888–902. [Google Scholar] [CrossRef]
  58. Hu, J.; Ni, P.; Dai, H.; Sun, Y.; Wang, Y.; Jiang, S.; Li, Z. Aptamer-based color imetric biosensing of abrinusing catalytic gold nanoparticles. Analyst 2015, 140, 3581–3586. [Google Scholar] [CrossRef] [PubMed]
  59. Sloan-Dennison, S.; Laing, S.; Shand, N.C.; Graham, D.; Faulds, K. A novel nanozyme assay utilizing the catalytic activity of silver nanoparticles and SERRS. Analyst 2017, 142, 2484–2490. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Karim, M.N.; Anderson, S.R.; Singh, S.; Ramanathan, R.; Bansal, V. Nanostructured silver fabricasa free-standing Nano Zyme for colorimetric detection of glucose inurine. Biosens. Bioelectron. 2018, 110, 8–15. [Google Scholar] [CrossRef] [PubMed]
  61. Fu, Y.; Zhang, H.; Dai, S.; Zhi, X.; Zhang, J.; Li, W. Glutathione-stabilized palladium nanozyme for colorimetric assay of silver(i) ions. Analyst 2015, 140, 6676–6683. [Google Scholar] [CrossRef]
  62. Chansuvarn, W.; Tuntulani, T.; Imyim, A. Colorimetric detection of mercury (II) based on gold nanoparticles, fluorescent gold nanoclusters and other gold-based nanomaterials. TRAC Trend Anal Chem. 2015, 65, 83–96. [Google Scholar] [CrossRef]
  63. Li, W.; Zhang, J.; Fu, Y. Synthesis and sensing application of glutathione-capped platinum nanoparticles. Anal. Methods 2015, 7, 4464–4471. [Google Scholar] [CrossRef]
  64. Sindhu, R.K.; Chitkara, M.; Sandhu, I.S. Nanotechnology: Principles and Applications, 1st ed.; Jenny Stanford Publishing: Singapore, 2021; pp. 41–70. [Google Scholar]
  65. Lin, X.-Q.; Deng, H.-H.; Wu, G.-W.; Peng, H.-P.; Liu, A.-L.; Lin, X.-H.; Xia, X.-H.; Chen, W. Platinum nanoparticles/grapheneoxide hybrid with excellent peroxidaselike activity and its application for cysteine detection. Analyst 2015, 140, 5251–5256. [Google Scholar] [CrossRef] [PubMed]
  66. Cai, K.; Lv, Z.; Chen, K.; Huang, L.; Wang, J.; Shao, F.; Wang, Y.; Han, H.-Y. Aqueous synthesis of porous platinum nanotubes at room temperature and their intrinsic peroxidaselike activity. Chem. Commun. 2013, 49, 6024–6026. [Google Scholar] [CrossRef]
  67. Gao, Z.; Xu, M.; Hou, L.; Chen, G.; Tang, D. Irregular-shaped platinum nanoparticles as peroxidase mimics for highly efficient colorimetric immunoassay. Anal. Chim. Acta 2013, 776, 79–86. [Google Scholar] [CrossRef]
  68. He, S.B.; Deng, H.H.; Liu, A.L.; Li, G.W.; Lin, X.H.; Chen, W.; Xia, X.H. Synthesis and Peroxidase Like Activity of Salt Resistant Platinum Nanoparticles by Using Bovine Serum Albumin as the Scaffold. ChemCatChem 2014, 6, 1543–1548. [Google Scholar] [CrossRef]
  69. Liu, Y.; Wu, H.; Li, M.; Yin, J.J.; Nie, Z. pH dependent catalytic activities of platinum nanoparticles with respect to the decomposition of hydrogen peroxide and scavenging of superoxide and singlet oxygen. Nanoscale 2014, 6, 11904–11910. [Google Scholar] [CrossRef] [PubMed]
  70. Wang, Z.; Yang, X.; Feng, J.; Tang, Y.; Jiang, Y.; He, N. Label-free detection of DNA by combining gated mesoporous silica and catalytic signal amplification of platinum nanoparticles. Analyst 2014, 139, 6088–6091. [Google Scholar] [CrossRef]
  71. Ju, Y.; Kim, J. Dendrimerencapsulated Pt nanoparticles with peroxidase-mimetic activity as biocatalytic labels for sensitive colorimetric analyses. Chem. Commun. 2015, 51, 13752–13755. [Google Scholar] [CrossRef]
  72. Raynal, M.; Ballester, P.; VidalFerran, A.; van Leeuwen, P.W.N.M. Supramolecular catalysis. Part 2: Artificial enzyme mimics. Chem Soc Rev. 2014, 43, 1734–1787. [Google Scholar] [CrossRef]
  73. Wang, Z.; Yang, X.; Yang, J.; Jiang, Y.; He, N. Peroxidase-like activity of mesoporous silica encapsulated Pt nanoparticle and its application in colorimetric immunoassay. Anal. Chim. Acta 2015, 862, 53–63. [Google Scholar] [CrossRef]
  74. Jin, L.; Meng, Z.; Zhang, Y.; Cai, S.; Zhang, Z.; Li, C.; Shang, L.; Shen, Y. Ultrasmall Pt Nanoclusters as Robust Peroxidase Mimics for Colorimetric Detection of Glucose in Human Serum. ACS Appl. Mater. Interfaces 2017, 9, 10027–10033. [Google Scholar] [CrossRef]
  75. Ye, H.; Liu, Y.; Chhabra, A.; Lilla, E.; Xia, X. Poly vinylpyrrolidone (PVP) Capped Ptnanocubes with superior peroxidase-Like activity. ChemNanoMat 2017, 3, 33–38. [Google Scholar] [CrossRef]
  76. Lan, J.; Xu, W.; Wan, Q.; Zhang, X.; Lin, J.; Chen, J.; Chen, J. Colorimetric determination of sarcosine in urine samples of prostatic carcinoma by mimic enzyme palladium nanoparticles. Anal. Chim. Acta 2014, 825, 63–68. [Google Scholar] [CrossRef] [PubMed]
  77. Liu, Y.; Purich, D.L.; Wu, C.; Wu, Y.; Chen, T.; Cui, C.; Zhang, L.; Cansiz, S.; Hou, W.; Wang, Y.; et al. Ionic function alization of hydrophobic colloidal nanoparticles to formionic nanoparticles with enzymelike properties. J. Am. Chem. Soc. 2015, 137, 14952–14958. [Google Scholar] [CrossRef] [PubMed]
  78. Wei, J.; Chen, X.; Shi, S.; Mo, S.; Zheng, N. An investigation of the mimetic enzyme activity of two-dimensional Pd-based nanostructures. Nanoscale 2015, 7, 19018–19026. [Google Scholar] [CrossRef] [PubMed]
  79. Hu, L.Z.; Liao, H.; Feng, L.Y.; Wang, M.; Fu, W.S. Accelerating the peroxidase-like activity of gold nanoclusters at neutral pH for colorimetric detection of heparin and heparinase activity. Anal Chem. 2018, 90, 6247–6252. [Google Scholar] [CrossRef]
  80. Song, Y.; Qu, K.; Zhao, C.; Ren, J.; Qu, X. Graphene oxide: Intrinsic per oxidase catalytic activity and its application to glucose detection. Adv. Mater. 2010, 22, 2206–2210. [Google Scholar] [CrossRef]
  81. Wu, D.; Deng, X.; Huang, X.; Wang, K.; Liu, Q. Low-cost preparation of photoluminescent carbon nanodots and application as peroxidase mimetics in colorimetric detection of H2O2and glucose. J. Nanosci. Nanotechnol. 2013, 13, 6611–6616. [Google Scholar] [CrossRef]
  82. Mohammadpour, Z.; Safavi, A.; Shamsipur, M. A new label free colorimetric chemosensor for detection of mercury ion with tunable dynamic range using carbon nanodots as enzyme mimics. Chem. Eng. J. 2014, 255, 1–7. [Google Scholar] [CrossRef]
  83. Shamsipur, M.; Safavi, A.; Mohammadpour, Z. Indirect colorimetric detection of glutathione based on its radical restoration ability using carbon nanodots as nanozymes. Sens. Actuators B Chem. 2014, 199, 463–469. [Google Scholar] [CrossRef]
  84. Zhu, W.; Zhang, J.; Jiang, Z.; Wang, W.; Liu, X. High-quality carbon dots: Synthesis, peroxidase-like activity and their application in the detection of H2O2, Ag+ and Fe3+. RSC Adv. 2014, 4, 17387–17392. [Google Scholar] [CrossRef]
  85. Garg, B.; Bisht, T. Carbon Nanodots as Peroxidase Nanozymes for Biosensing. Molecules 2016, 21, 1653. [Google Scholar] [CrossRef] [PubMed]
  86. Tang, D.; Liu, J.; Yan, X.; Kang, L. Graphene oxide derived graphene quantum dots with different photoluminescence properties and peroxidase-like catalytic activity. RSC Adv. 2016, 6, 50609–50617. [Google Scholar] [CrossRef]
  87. Nirala, N.R.; Khandelwal, G.; Kumar, B.; Prakash, R.; Kumar, V. One step electro-oxidative preparation of graphene quantum dots from wood charcoal as a peroxidase mimetic. Talanta 2017, 173, 36–43. [Google Scholar] [CrossRef]
  88. Vázquez-González, M.; Liao, W.C.; Cazelles, R.; Wang, S.; Yu, X.; Gutkin, V.; Willner, I. Mimicking Horseradish Peroxidase Functions Using Cu2+-Modified Carbon Nitride Nanoparticles or Cu2+-Modified Carbon Dots as Heterogeneous Catalysts. ACS Nano 2017, 11, 3247–3253. [Google Scholar] [CrossRef]
  89. Dong, Y.; Li, J.; Shi, L.; Guo, Z. Iron impuritiesas theactive sites for peroxidase like catalytic reaction on graphene and its derivatives. ACS Appl. Mater. Interfaces 2015, 7, 15403–15413. [Google Scholar] [CrossRef]
  90. Dong, Y.; Li, J.; Shi, L.; Xu, J.; Wang, X.; Guo, Z.; Liu, W. Grapheneoxide–iron complex: Synthesis, characterization and visible-light-driven photo catalysis. J. Mater. Chem. A 2013, 1, 644–650. [Google Scholar] [CrossRef]
  91. Gayathri, P.; Kumar, A.S. An Iron Impurity in Multiwalled Carbon Nanotube Complexes with Chitosan that Biomimics the Heme-Peroxidase Function. Chem.-A Eur. J. 2013, 19, 17103–17112. [Google Scholar] [CrossRef]
  92. Lin, L.; Song, X.; Chen, Y.; Rong, M.; Zhao, T.; Wang, Y.; Jiang, Y.; Chen, X. Intrinsic peroxidase-like catalytic activity of nitrogen-doped graphene quantum dots and their application in the colorimetric detection of H2O2 and glucose. Anal. Chim. Acta 2015, 869, 89–95. [Google Scholar] [CrossRef]
  93. Zhang, R.; He, S.; Zhang, C.; Chen, W. Three-dimensional Fe- and N-incorporated carbon structures as peroxidase mimics for fluorescence detection of hydrogen peroxide and glucose. J. Mater. Chem. B 2015, 3, 4146–4154. [Google Scholar] [CrossRef]
  94. Yang, W.; Huang, T.; Zhao, M.; Luo, F.; Weng, W.; Wei, Q.; Lin, Z.; Chen, G. High peroxidase-like activity of iron and nitrogen co-doped carbon dots and its application in immunosorbent assay. Talanta 2017, 164, 1–6. [Google Scholar] [CrossRef] [Green Version]
  95. Tian, J.; Liu, Q.; Asiri, A.M.; Qusti, A.H.; Al-Youbi, A.O.; Sun, X. Ultrathin graphitic carbon nitride nanosheets: A novel peroxidase mimetic, Fe doping-mediated catalytic performance enhancement and application to rapid, highly sensitive optical detection of glucose. Nanoscale 2013, 5, 11604–11609. [Google Scholar] [CrossRef] [Green Version]
  96. Lin, T.; Zhong, L.; Wang, J.; Guo, L.; Wu, H.; Guo, Q.; Fu, F.; Chen, G. Graphite-like carbon nitrides as peroxidase mimetics and their applications to glucose detection. Biosens. Bioelectron. 2014, 59, 89–93. [Google Scholar] [CrossRef]
  97. Qiao, F.; Wang, J.; Ai, S.; Li, L. As an ewperoxidasemimetic: The synthesis of selenium doped graphitic carbon nitride nanosheets and applications on colorimetric detection of H2O2 and xanthine. Sens. Actuators B Chem. 2015, 216, 418–427. [Google Scholar] [CrossRef]
  98. Cheng, H.; Liu, Y.; Hu, Y.; Ding, Y.; Lin, S.; Cao, W.; Wang, Q.; Wu, J.; Muhammad, F.; Zhao, X.; et al. Monitoring of Heparin Activity in Live Rats Using Metal–Organic Framework Nanosheets as Peroxidase Mimics. Anal. Chem. 2017, 89, 11552–11559. [Google Scholar] [CrossRef] [PubMed]
  99. Biella, S.; Prati, L.; Rossi, M. Selective Oxidation of D-Glucose on Gold Catalyst. J. Catal. 2002, 206, 242–247. [Google Scholar] [CrossRef]
  100. Comotti, M.; Della Pina, C.; Matarrese, R.; Rossi, M. The Catalytic Activity of Naked Gold Particles. Angew. Chem. Int. Ed. 2004, 43, 5812–5815. [Google Scholar] [CrossRef] [PubMed]
  101. Beltrame, P.; Comotti, M.; Della Pina, C.; Rossi, M. Aerobic oxidation of glucose: II. Catalysis by colloidal gold. Appl. Catal. A Gen. 2006, 297, 1–7. [Google Scholar] [CrossRef]
  102. Periasamy, A.P.; Roy, P.; Wu, W.P.; Huang, Y.H.; Chang, H.T. Glucose Oxidase and Horseradish Peroxidase Like Activities of Cuprous Oxide/Polypyrrole Composites. Electrochim. Acta 2016, 215, 253–260. [Google Scholar] [CrossRef]
  103. Lewandowska, H.; Wójciuk, K.; Karczmarczyk, U. Metal Nanozymes: New Horizons in Cellular Homeostasis Regulation. Appl. Sci. 2021, 11, 9019. [Google Scholar] [CrossRef]
  104. Li, L.; Zhang, L.; Carmona, U.; Knez, M. Semi-artificial and bioactive ferroxidase with nanoparticles as the active sites. Chem. Commun. 2014, 50, 8021–8023. [Google Scholar] [CrossRef]
  105. Mu, J.; Zhang, L.; Zhao, M.; Wang, Y. CO3O4 nanoparticles as an efficient catalase mimic: Properties, mechanism and its electro catalytic sensing application for hydrogen peroxide. J. Mol. Catal. A Chem. 2013, 378, 3037. [Google Scholar] [CrossRef]
  106. Wang, X.; Zhang, Y.; Li, T.; Tian, W.; Zhang, Q.; Cheng, Y. Generation poly amidoamine dendrimeren capsulated platinum nanoparticle mimics catalase size, shape, and catalytic activity. Langmuir 2013, 29, 5262–5270. [Google Scholar] [CrossRef]
  107. Zhang, W.; Zhang, Y.; Gu, N. Prussian Blue Modified Ferritin Nanoparticlesas Peroxidase and Catalase Mimetics and Their Application in Glucose Detection. In Key Engineering Materials; Trans Tech Publications Ltd.: Bäch, Switzerland, 2013; Volume 562, pp. 1333–1339. [Google Scholar]
  108. Zhu, Z.; Guan, Z.; Jia, S.; Lei, Z.; Lin, S.; Zhang, H.; Ma, Y.; Tian, Z.-Q.; Yang, C.J. Au@Pt Nanoparticle Encapsulated Target-Responsive Hydrogel with Volumetric Bar-Chart Chip Readout for Quantitative Point-of-Care Testing. Angew. Chem. Int. Ed. 2014, 53, 12503–12507. [Google Scholar]
  109. Nicolini, V.; Gambuzzi, E.; Malavasi, G.; Menabue, L.; Menziani, M.C.; Lusvardi, G.; Pedone, A.; Benedetti, F.; Luches, P.; D’Addato, S.; et al. Evidence of Catalase Mimetic Activity in Ce3+/Ce4+ Doped Bioactive Glasses. J. Phys. Chem. B 2015, 119, 4009–4019. [Google Scholar] [CrossRef]
  110. Aneesh, K.; Vusa, C.S.; Berchmans, S. Dualenzyme mimic ryexhibited by ITO nanocubes and their application inspector photometric and electro chemical sensing. Analyst 2016, 141, 4024–4028. [Google Scholar] [CrossRef] [PubMed]
  111. Hu, M.; Korschelt, K.; Daniel, P.; Landfester, K.; Tremel, W.; Bannwarth, M.B. Fibrous Nanozyme Dressings with Catalase-Like Activity for H2O2 Reduction to Promote Wound Healing. ACS Appl. Mater. Interfaces 2017, 9, 38024–38031. [Google Scholar] [CrossRef] [PubMed]
  112. Kim, M.Y.; Kim, J. Chitosan Microgels Embedded with Catalase Nanozyme-Loaded Mesocellular Silica Foam for Glucose-Responsive Drug Delivery. ACS Biomater. Sci. Eng. 2017, 3, 572–578. [Google Scholar] [CrossRef] [PubMed]
  113. Li, J.; Liu, W.; Wu, X.; Gao, X. Mechanism of pH-switchable peroxidase and catalase-like activities of gold, silver, platinum and palladium. Biomaterials 2015, 48, 37–44. [Google Scholar] [CrossRef]
  114. Zhang, W.; Dong, J.; Wu, Y.; Cao, P.; Song, L.; Ma, M.; Gu, N.; Zhang, Y. Shape-dependent enzyme-like activity of CO3O4 nanoparticles and their conjugation with his-tagged EGFR single-domain antibody. Colloids Surfaces B Biointerfaces 2017, 154, 55–62. [Google Scholar] [CrossRef]
  115. Sobańska, K.; Pietrzyk, P.; Sojka, Z. Generation of reactive oxygen species via electro proticinter action of H2O2 with ZrO2 gel: Ionic sponge effect and pH switchable peroxidase and catalase like activity. ACS Catal. 2017, 7, 2935–2947. [Google Scholar] [CrossRef]
  116. Ma, X.; Hu, W.; Guo, C.; Yu, L.; Gao, L.; Xie, J.; Li, C.M. DNA-Templated Biomimetic Enzyme Sheets on Carbon Nanotubes to Sensitively In Situ Detect Superoxide Anions Released from Cells. Adv. Funct. Mater. 2014, 24, 5897–5903. [Google Scholar] [CrossRef]
  117. Yuan, L.; Liu, S.; Tu, W.; Zhang, Z.; Bao, J.; Dai, Z. Biomimetic Superoxide Dismutase Stabilized by Photopolymerization for Superoxide Anions Biosensing and Cell Monitoring. Anal. Chem. 2014, 86, 4783–4790. [Google Scholar] [CrossRef] [PubMed]
  118. Grace, A.N.; Pandian, K. Organically Dispersible Gold and Platinum Nanoparticles Using Aromatic Amines as Phase Transfer and Reducing Agent and Their Applications in Electro-Oxidation of Glucose. Colloids Surf. A Physicochem. Eng. Asp. 2007, 302, 113–120. [Google Scholar] [CrossRef]
  119. Kamada, K.; Soh, N. Enzyme-Mimetic Activity of Ce-Intercalated Titanate Nanosheets. J. Phys. Chem. B 2015, 119, 5309–5314. [Google Scholar] [CrossRef]
  120. Liu, T.; Niu, X.; Shi, L.; Zhu, X.; Zhao, H.; Lana, M. Electro catalytic analysis of super oxideanion radical using nitrogen-doped graphene supported Prussian Blueasa biomimetic superoxide dismutase. Electrochim. Acta. 2015, 176, 1280–1287. [Google Scholar] [CrossRef]
  121. Shen, X.; Liu, W.; Gao, X.; Lu, Z.; Wu, X.; Gao, X. Mechanisms of Oxidase and Superoxide Dismutation-like Activities of Gold, Silver, Platinum, and Palladium, and Their Alloys: A General Way to the Activation of Molecular Oxygen. J. Am. Chem. Soc. 2015, 137, 15882–15891. [Google Scholar] [CrossRef]
  122. Mu, J.; Zhao, X.; Li, J.; Yang, E.-C.; Zhao, X.-J. Novel hierarchical NiO nanoflowers exhibiting intrinsic superoxide dismutase-like activity. J. Mater. Chem. B 2016, 4, 5217–5221. [Google Scholar] [CrossRef] [PubMed]
  123. Shen, X.; Wang, Q.; Liu, Y.; Xue, W.; Ma, L.; Feng, S.; Wan, M.; Wang, F.; Mao, C. Manganese Phosphate Self-Assembled Nanoparticle Surface and Its application for Superoxide Anion Detection. Sci. Rep. 2016, 6, 28989. [Google Scholar] [CrossRef] [PubMed]
  124. Wang, M.-Q.; Ye, C.; Bao, S.-J.; Xu, M.-W.; Zhang, Y.; Wang, L.; Ma, X.-Q.; Guo, J.; Li, C.M. Nanostructured cobalt phosphates as excellent biomimetic enzymes to sensitively detect superoxide anions released from living cells. Biosens. Bioelectron. 2017, 87, 998–1004. [Google Scholar] [CrossRef]
  125. Liguori, I.; Russo, G.; Curcio, F.; Bulli, G.; Aran, L.; Della-Morte, D.; Gargiulo, G.; Testa, G.; Cacciatore, F.; Bonaduce, D. Oxidative Stress, Aging, and Diseases. Clin. Interv. Aging 2018, 13, 757. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Huang, F.; Wang, J.; Chen, W.; Wan, Y.; Wang, X.; Cai, N.; Liu, J.; Yu, F. Synergistic Peroxidase-like Activity of CeO2 -Coated Hollow Fe3O4 Nanocomposites as an Enzymatic Mimic for Low Detection Limit of Glucose. J. Taiwan Inst. Chem. Eng. 2018 83, 40–49. [CrossRef]
  127. Tarnuzzer, R.W.; Colon, J.; Patil, S.; Seal, S. Vacancy Engineered Ceria Nanostructures for Protection from Radiation-Induced Cellular Damage. Nano Lett. 2005, 5, 2573–2577. [Google Scholar] [CrossRef] [PubMed]
  128. Korsvik, C.; Patil, S.; Seal, S.; Self, W.T. Superoxide dismutase mimetic proper ties exhibited by vacancy engineer edceria nanoparticles. Chem. Commun. 2007, 10, 1056–1058. [Google Scholar] [CrossRef] [PubMed]
  129. Heckert, E.G.; Karakoti, A.S.; Seal, S.; Self, W.T. The role of cerium redox state in the SOD mimetic activity of nanoceria. Biomaterials 2008, 29, 2705–2709. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Yang, Z.-Y.; Luo, S.-L.; Li, H.; Dong, S.-W.; He, J.; Jiang, H.; Li, R.; Yang, X.-C. Alendronate as a robust anchor for ceria nanoparticle surface coating: Facile binding and improved biological properties. RSC Adv. 2014, 4, 59965–59969. [Google Scholar] [CrossRef]
  131. Pulido-Reyes, G.; Rodea-Palomares, I.; Das, S.; Sakthivel, T.S.; Leganes, F.; Rosal, R.; Seal, S.; Fernández-Piñas, F. Untangling the biological effects of ceriumoxide nanoparticles: The role of surface valence states. Sci. Rep. 2015, 5, 1–4. [Google Scholar] [CrossRef] [Green Version]
  132. Liu, Y.; Ai, K.; Ji, X.; Askhatova, D.; Du, R.; Lu, L.; Shi, J. 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, 856–862. [Google Scholar] [CrossRef] [Green Version]
  133. Tokuyama, H.; Yamago, S.; Nakamura, E.; Shiraki, T.; Sugiura, Y. Photoinduced biochemical activity of fullerene carboxylic acid. J. Am. Chem. Soc. 1993, 115, 7918–7919. [Google Scholar] [CrossRef]
  134. Boutorine, A.S.; Takasugi, M.; Hélène, C.; Tokuyama, H.; Isobe, H.; Nakamura, E. Fullerene Oligonucleotide Conjugates: Photoinduced Sequence Specific DNA Cleavage. Angew. Chem. Int. Ed. Engl. 1995, 33, 2462–2465. [Google Scholar] [CrossRef]
  135. Yamakoshi, Y.N.; Yagami, T.; Sueyoshi, S.; Miyata, N. Acridine adduct of fullerene witen hanced DNA cleaving activity. J. Org. Chem. 1996, 61, 7236–7237. [Google Scholar] [CrossRef] [PubMed]
  136. Hostert, L.; Blaskievicz, S.; Fonsaca, J.; Domingues, S.; Zarbin, A.J.; Orth, E. Imidazole-derived graphene nanocatalysts for organophosphate destruction: Powder and thin film heterogeneous reactions. J. Catal. 2017, 356, 75–84. [Google Scholar] [CrossRef]
  137. Ma, X.; Zhang, L.; Xia, M.; Li, S.; Zhang, X.; Zhang, Y. Mimicking the Active Sites of Organophosphorus Hydrolase on the Backbone of Graphene Oxide to Destroy Nerve Agent Simulants. ACS Appl. Mater. Interfaces 2017, 9, 21089–21093. [Google Scholar] [CrossRef] [PubMed]
  138. Wang, T.; Wang, J.; Yang, Y.; Su, P.; Yang, Y. CO3O4/reduced grapheneoxide nanocomposites as effective phosphotriest erase mimetics for degradation and detection of paraoxon. Ind. Eng. Chem. Res. 2017, 56, 9762–9769. [Google Scholar] [CrossRef]
  139. Manea, F.; Houillon, F.B.; Pasquato, L.; Scrimin, P. Nanozymes: Gold-Nanoparticle-Based Transphosphorylation Catalysts. Angew. Chem. 2004, 116, 6291–6295. [Google Scholar] [CrossRef]
  140. Baldim, V.; Bedioui, F.; Mignet, N.; Margaill, I.; Berret, J.-F. The enzyme-like catalytic activity of cerium oxide nanoparticles and its dependency on Ce3+ surface area concentration. Nanoscale 2018, 10, 6971–6980. [Google Scholar] [CrossRef] [Green Version]
  141. Zaupa, G.; Mora, C.; Bonomi, R.; Prins, L.J.; Scrimin, P. Catalytic Self-Assembled Monolayers on Au Nanoparticles: The Source of Catalysis of a Transphosphorylation Reaction. Chem.-A Eur. J. 2011, 17, 4879–4889. [Google Scholar] [CrossRef]
  142. Li, P.; Klet, R.C.; Moon, S.-Y.; Wang, T.C.; Deria, P.; Peters, A.W.; Klahr, B.M.; Park, H.-J.; Al-Juaid, S.S.; Hupp, J.T.; et al. Synthesis of nanocrystals of Zr-based metal–organic frameworks with csq-net: Significant enhancement in the degradation of a nerve agent simulant. Chem. Commun. 2015, 51, 10925–10928. [Google Scholar] [CrossRef]
  143. Mondloch, J.E.; Katz, M.; Iii, W.C.I.; Ghosh, P.; Liao, P.; Bury, W.; Wagner, G.W.; Hall, M.G.; DeCoste, J.B.; Peterson, G.; et al. Destruction of chemical warfare agents using metal–organic frameworks. Nat. Mater. 2015, 14, 512–516. [Google Scholar] [CrossRef]
  144. Moon, S.-Y.; Wagner, G.W.; Mondloch, J.E.; Peterson, G.W.; Decoste, J.B.; Hupp, J.T.; Farha, O.K. Effective, Facile, and Selective Hydrolysis of the Chemical Warfare Agent VX Using Zr6-Based Metal–Organic Frameworks. Inorg. Chem. 2015, 54, 10829–10833. [Google Scholar] [CrossRef] [PubMed]
  145. Nunes, P.; Gomes, A.C.; Pillinger, M.; Gonçalves, I.S.; Abrantes, M. Promotion of phosphor ester hydrolysis by the ZrIV based metal organic framework UiO-67. Microporous Mesoporous Mater. 2015, 208, 21–29. [Google Scholar] [CrossRef]
  146. López-Maya, E.; Montoro, C.; Rodríguez-Albelo, L.M.; Aznar Cervantes, S.D.; Lozano-Pérez, A.A.; Cenís, J.L.; Barea, E.; Navarro, J.A. Textile/Metal Organic Framework Composites as Self Detoxifying Filters for Chemical Warfare Agents. Angew. Int. Ed. 2015, 54, 6790–6794. [Google Scholar] [CrossRef] [PubMed]
  147. Nath, I.; Chakraborty, J.; Verpoort, F. Metal organic frameworks mimicking natural enzymes: A structural and functional analogy. Chem. Soc. Rev. 2016, 45, 4127–4170. [Google Scholar] [CrossRef] [PubMed]
  148. Lee, D.T.; Zhao, J.; Peterson, G.W.; Parsons, G.N. A Catalytic ‘MOF-Cloth’ Formed via Directed Supramolecular Assembly of UiO-66-NH2 Crystals on ALD-coated Textiles for Rapid Degradation of Chemical Warfare Agent Simulants. Chem. Mater. 2017, 29, 4894–4903. [Google Scholar] [CrossRef]
  149. McCarthy, D.L.; Liu, J.; Dwyer, D.B.; Troiano, J.L.; Boyer, S.M.; De Coste, J.B.; Bernier, W.E.; Jones, W.E., Jr. Electrospunmetal organic framework polymer composites for the catalytic degradation of methylparaoxon. New J. Chem. 2017, 41, 8748–8753. [Google Scholar] [CrossRef]
  150. Park, H.J.; Jang, J.K.; Kim, S.-Y.; Ha, J.-W.; Moon, D.; Kang, I.-N.; Bae, Y.-S.; Kim, S.; Hwang, D.-H. Synthesis of a Zr-Based Metal–Organic Framework with Spirobifluorenetetrabenzoic Acid for the Effective Removal of Nerve Agent Simulants. Inorg. Chem. 2017, 56, 12098–12101. [Google Scholar] [CrossRef]
  151. Najafpour, M.M.; Madadkhani, S.; Zand, Z.; Hołyńska, M.; Allakhverdiev, S.I. Engineered poly peptidearound nano-sized manganese calciumoxideas an artificial water oxidizing enzyme mimic king natural photosynthesis: Toward artificial enzymes with highly active site densities. Int. J. Hydrog. Energy 2016, 41, 17826–17836. [Google Scholar] [CrossRef]
  152. Saeed, M.; Deng, L. Carbon nanotube enhanced PVA-mimic enzyme membrane for post-combustion CO2 capture. Int. J. Greenh. Gas Control. 2016, 53, 254–262. [Google Scholar] [CrossRef]
  153. Nandhakumar, P.; Kim, B.; Lee, N.-S.; Yoon, Y.H.; Lee, K.; Yang, H. Nitrosoreductase-Like Nanocatalyst for Ultrasensitive and Stable Biosensing. Anal. Chem. 2017, 90, 807–813. [Google Scholar] [CrossRef]
  154. Xu, K.; Zhong, Z.; Xu, H.; Wang, X.; Zhao, M.; Wu, C. Highly Efficient Aerobic Oxidation of Arylalkanes with a Biomimetic Catalyst Platform. Chin. J. Appl. Chem. 2017, 34, 1079–1085. [Google Scholar]
  155. Xu, J.J.; Wang, M.M.; Liu, L.Z.; Li, F.; Tian, J.S. Analysis of alkaline phosphatase activity of magnetosome. J. China Agric. Univ. 2018, 23, 8–13. [Google Scholar]
  156. Murugan, C.; Murugan, N.; Sundramoorthy, A.K.; Sundaramurthy, A. Nanoceria Decorated Flower-like Molybdenum Sulphide Nanoflakes: An Efficient Nanozyme for Tumour Selective ROS Generation and Photo Thermal Therapy. Chem. Commun. 2019, 55, 8017–8020. [Google Scholar] [CrossRef]
  157. Xue, T.; Peng, B.; Xue, M.; Zhong, X.; Chiu, C.-Y.; Yang, S.; Qu, Y.; Ruan, L.; Jiang, S.; Dubin, S.; et al. Integration of molecular and enzymatic catalysts on graphene for biomimetic generation of antithrombotic species. Nat. Commun. 2014, 5, 1–6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Fateeva, A.; Chater, P.A.; Ireland, C.P.; Tahir, A.A.; Khimyak, Y.Z.; Wiper, P.V.; Darwent, J.R.; Rosseinsky, M.J. A Water-Stable Porphyrin-Based Metal-Organic Framework Active for Visible-Light Photocatalysis. Angew. Chem. Int. Ed. 2012, 51, 7440–7444. [Google Scholar] [CrossRef]
  159. Sasan, K.; Lin, Q.; Mao, C.; Feng, P. Incorporation of iron hydrogenase active sites into a highly stable metal–organic framework for photocatalytic hydrogen generation. Chem. Commun. 2014, 50, 10390–10393. [Google Scholar] [CrossRef] [PubMed]
  160. Pullen, S.; Fei, H.; Orthaber, A.; Cohen, S.M.; Ott, S. Enhanced Photochemical Hydrogen Production by a Molecular Diiron Catalyst Incorporated into a Metal–Organic Framework. J. Am. Chem. Soc. 2013, 135, 16997–17003. [Google Scholar] [CrossRef] [PubMed]
  161. Sahoo, P.C.; Jang, Y.N.; Lee, S.W. Enhanced biomimetic CO2 sequestration and CaCO3 crystallization using complex encapsulated metal organic framework. J. Cryst. Growth 2013, 373, 96–101. [Google Scholar] [CrossRef]
  162. Fillon, Y.; Verma, A.; Ghosh, P.; Ernenwein, D.; Rotello, V.M.; Chmielewski, J. Peptideligation catalyzed by functionalized gold nanoparticles. J. Am. Chem. Soc. 2007, 129, 6676–6677. [Google Scholar] [CrossRef] [Green Version]
  163. Qin, L.; Hu, Y.; Wei, H. Nanozymes: Preparation and Characterization. In Nanostructure Science and Technology; Yan, X., Ed.; Springer: Singapore, 2020. [Google Scholar]
  164. Li, J.; Wu, Q.; Wu, J. Synthesis of Nanoparticlesvia Solvothermal and Hydrothermal Methods. In Handbook of Nanoparticles; Aliofkhazraei, M., Ed.; Springer International Publishing: Cham, Switzerland, 2016; pp. 295–328. [Google Scholar]
  165. Wang, H.; Wan, K.; Shi, X. Recent Advances in Nanozyme Research. Adv. Mater. 2019, 31, e1805368. [Google Scholar] [CrossRef]
  166. Luo, L.; Zhang, Y.; Li, F.; Si, X.; Ding, Y.; Deng, D.; Wang, T. Enzyme mimics of spinel-type CoxNi1−xFe2O4 magnetic nanomaterial for eletroctrocatalytic oxidation of hydrogen peroxide. Anal. Chim. Acta 2013, 788, 46–51. [Google Scholar] [CrossRef] [PubMed]
  167. Wu, Q.; He, L.; Jiang, Z.W.; Li, Y.; Zheng, M.C.; Cheng, Z.H.; Li, Y.F. CuO Nanoparticles Derived from Metal-Organic Gelwith Excellent Electrocatalytic and Peroxidase-Mimicking Activities for Glucose and Cholesterol Detection. Biosens. Bioelectron. 2019, 145, 111704. [Google Scholar] [CrossRef] [PubMed]
  168. Zheng, S.; Li, B.; Tang, Y.; Li, Q.; Xue, H.; Pang, H. Ultrathin Nanosheet Assembled [Ni3(OH)2(PTA)2(H2O)4]2H2O Hierarchical Flowers for High Performance Electrocatalysis of Glucose Oxidation Reactions. Nanoscale 2018, 10, 13270–13276. [Google Scholar] [CrossRef]
  169. Li, Y.; Zhang, H.; Cai, X.; Zhao, H.; Magdassi, S.; Lan, M. Electrochemical detection of superoxide anions in HeLa cells by using two enzyme-free sensors prepared from ZIF-8-derived carbon nanomaterials. Microchim. Acta 2019, 186, 370. [Google Scholar] [CrossRef] [PubMed]
  170. Wang, C.; Liu, C.; Li, J.; Sun, X.; Shen, J.; Han, W.; Wang, L. Electrospun metal–organic framework derived hierarchical carbon nanofibers with high performance for supercapacitors. Chem. Commun. 2017, 53, 1751–1754. [Google Scholar] [CrossRef] [PubMed]
  171. Scandurra, A.; Ruffino, F.; Sanzaro, S.; Grimaldi, M.G. Laserand Thermal Dewetting of Gold Layer onto Graphene Paperforn on Enzymatic Electrochemical Detection of Glucose and Fructose. Sens. Actuators B Chem. 2019, 301, 127113. [Google Scholar] [CrossRef]
  172. Chou, K.-S.; Ren, C.-Y. Synthesis of nanosized silver particles by chemical reduction method. Mater. Chem. Phys. 2000, 64, 241–246. [Google Scholar] [CrossRef]
  173. Rane, A.V.; Kanny, K.; Abitha, V.K.; Sabu, T.C. Methods for Synthesis of Nanoparticles and Fabrication of Nanocomposites. In Synthesis of Inorganic Nanomaterials Advances and Key Technologies Micro and Nano Technologies, 1st ed.; Woodhead Publishing Company: Sawston, UK, 2018; pp. 121–139. [Google Scholar]
  174. Suriati, G.M.; Mariatti, M.; Azizan, A. Synthesis of silver nanoparticles buchemical reduction method: Effect of reducing agent and surfactant concentration. Int. J. Automot. Mech. Eng. 2014, 10, 1920–1927. [Google Scholar] [CrossRef]
  175. Zhu, J.; Peng, X.; Nie, W.; Wang, Y.; Gao, J.; Wen, W.; Wang, S. Hollow copper sulfide nanocubesas multifunctional nanozymes for colorimetric detection of dopamine and electrochemical detection of glucose. Biosens. Bioelectron. 2019, 141, 111450. [Google Scholar] [CrossRef]
  176. DAS, R.; Dhiman, A.; Kapil, A.; Bansal, V.; Sharma, T.K. Aptamer-mediated colorimetric and electrochemical detection of Pseudomonas aeruginosa utilizing peroxidase-mimic activity of gold NanoZyme. Anal. Bioanal. Chem. 2019, 411, 1229–1238. [Google Scholar] [CrossRef]
  177. Han, R.; Lu, Y.; Mingjun, L.; Yanbo, W.; Xuan, L.; Chongyang, L.; Kun, L.; Lingxing, Z.; Aihua, L. Green tide biomass templated synthesis of molybdenumoxide nanorods supported on carbon as efficient nanozyme for sensitive glucose colorimetric assay. Sens. Actuators B Chem. 2019, 296, 126517. [Google Scholar]
  178. Han, L.; Zhang, H.; Li, F. Bioinspired Nanozymes with pH Independent and MetalIons-Controllable Activity: Field Programmable Logic Conversion of Sole Logic Gate System. Part. Part. Syst. Char. 2018, 35, 1800207. [Google Scholar] [CrossRef]
  179. Zhai, D.; Liu, B.; Shi, Y.; Pan, L.; Wang, Y.; Li, W.; Zhang, R.; Yu, G. Highly Sensitive Glucose Sensor Based on Pt Nanoparticle/Polyaniline Hydrogel Heterostructures. ACS Nano 2013, 7, 3540–3546. [Google Scholar] [CrossRef]
  180. Chen, H.-I.; Chang, H.-Y. Synthesis of nanocrystalline cerium oxide particles by the precipitation method. Ceram. Int. 2005, 31, 795–802. [Google Scholar] [CrossRef]
  181. Dashtestani, F.; Ghourchian, H.; Eskandari, K.; RafieePour, H.A. A superoxide dismutase mimic nanocomposite for ampere metric sensing of superoxide anions. Microchim. Acta 2015, 82, 1045–1053. [Google Scholar] [CrossRef]
  182. Tonelli, D.; Scavetta, E.; Gualandi, I. Electrochemical Deposition of Nanomaterials for Electrochemical Sensing. Sensors 2019, 19, 1186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Al-Bat’hi, S.A.M. Electrode position of nanostructure materials. In Electroplating of Nanostructures; Aliofkhazraei, M., Ed.; InTech: Rijeka, Croatia, 2015; pp. 3–25. [Google Scholar]
  184. Wurm, F.R.; Weiss, C.K. Nanoparticles from renewable polymers. Front. Chem. 2014, 2, 49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Santhosh, P.; Manesh, K.M.; Lee, S.-H.; Uthayakumar, S.; Gopalan, A.I.; Lee, K.-P. Sensitive electrochemical detection of superoxide anion using gold nanoparticles distributed poly(methyl methacrylate)–polyaniline core–shell electrospun composite electrode. Analyst 2011, 136, 1557–1561. [Google Scholar] [CrossRef] [PubMed]
  186. Wei, H.; Wang, E. Nanomaterials with enzyme-like characteristics (nanozymes): Next-generation artificial enzymes. Chem. Soc. Rev. 2013, 42, 6060–6093. [Google Scholar] [CrossRef]
  187. Wang, X.; Hu, Y.; Wei, H. Nanozymes in bionanotechnology: From sensing to therapeutics and beyond. Inorg. Chem. Front. 2016, 3, 41–60. [Google Scholar] [CrossRef]
  188. Gao, L.; Fan, K.; Yan, X. Iron Oxide Nanozyme: A Multifunctional Enzyme Mimetic for Biomedical Applications. Theranostics 2017, 7, 3207–3227. [Google Scholar] [CrossRef] [PubMed]
  189. Liu, B.; Han, X.; Liu, J. Iron oxide nanozyme catalyzed synthesis of fluorescent polydopamine for light-up Zn2+ detection. Nanoscale 2016, 8, 13620–13626. [Google Scholar] [CrossRef] [PubMed]
  190. Celardo, I.; Pedersen, J.Z.; Traversa, E.; Ghibelli, L. Pharmacological potential of cerium oxide nanoparticles. Nanoscale 2011, 3, 1411–1420. [Google Scholar] [CrossRef]
  191. Feng, L.; Musto, C.J.; Suslick, K.S. A Simple and Highly Sensitive Colorimetric Detection Method for Gaseous Formaldehyde. J. Am. Chem. Soc. 2010, 132, 4046–4047. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Butler, J.E. Enzyme-linked immune sorbent assay. J. Immunoass. 2000, 21, 165–209. [Google Scholar] [CrossRef]
  193. Deng, J.; Wang, K.; Wang, M.; Yu, P.; Mao, L. Mitochondria Targeted Nanoscale Zeolitic Imidazole Framework-90 for ATP Imaging in Live Cells. J. Am. Chem. Soc. 2017, 139, 5877–5882. [Google Scholar] [CrossRef]
  194. Fu, P.P.; Xia, Q.; Hwang, H.-M.; Ray, P.C.; Yu, H. Mechanisms of nanotoxicity: Generation of reactive oxygen species. J. Food Drug Anal. 2014, 22, 64–75. [Google Scholar] [CrossRef] [Green Version]
  195. Ge, S.; Liu, W.; Liu, H.; Liu, F.; Yu, J.; Yan, M.; Huang, J. Colorimetric detection of the flux of hydrogen peroxide released from living cells based on the high peroxidase-like catalytic performance of porous PtPd nanorods. Biosens. Bioelectron. 2015, 71, 456–462. [Google Scholar] [CrossRef]
  196. Chi, M.; Nie, G.; Jiang, Y.; Yang, Z.; Zhang, Z.; Wang, C.; Lu, X. Self-Assembly Fabrication of Coaxial Tepoly(3,4-ethylenedioxythiophene) Nanocables and Their Conversion to Pdpoly(3,4-ethylenedioxythiophene) Nanocables with a High Peroxidase-like Activity. ACS Appl. Mater. Interfaces 2016, 8, 1041–1049. [Google Scholar] [CrossRef]
  197. Ariga, K.; Ji, Q.; Mori, T.; Naito, M.; Yamauchi, Y.; Abe, H.; Hill, J. Enzyme nanoarchitectonics: Organization and device application. Chem. Soc. Rev. 2013, 42, 6322–6345. [Google Scholar] [CrossRef]
  198. Chen, H.C.; Tu, Y.M.; Hou, C.C.; Lin, Y.C.; Chen, C.H.; Yang, K.H. Direct electron transfer of glucose oxidase and dual hydrogen peroxide and glucose detection based on water-dispersible carbon nanotubes derivative. Anal. Chim. Acta 2015, 867, 83–91. [Google Scholar] [CrossRef] [PubMed]
  199. Han, L.; Shi, J.; Liu, A. Novel biotemplated MnO2 1D nanozyme with controllable peroxidase-like activity and unique catalytic mechanism and its application for glucose sensing. Sens. Actuators B Chem. 2017, 252, 919–926. [Google Scholar] [CrossRef]
  200. Wang, Q.; Zhang, X.; Huang, L.; Zhang, Z.; Dong, S. Onepotsynthesis of Fe3O4 nanoparticle loaded 3D porous graphene nanocomposites with enhanced nanozyme activity for glucose detection. ACS Appl. Mater. Interfaces 2017, 9, 7465–7471. [Google Scholar] [CrossRef] [PubMed]
  201. Lien, C.-W.; Chen, Y.-C.; Chang, H.-T.; Huang, C.-C. Logical regulation of the enzyme-like activity of gold nanoparticles by using heavy metal ions. Nanoscale 2013, 5, 8227–8234. [Google Scholar] [CrossRef]
  202. Wang, G.L.; Jin, L.-Y.; Wu, X.M.; Dong, Y.M.; Li, Z.J. Label-free colorimetric sensor for mercury(II) and DNA on the basis of mercury(II) switched-on the oxidase-mimicking activity of silver nanoclusters. Anal. Chim. Acta 2015, 871, 1–8. [Google Scholar] [CrossRef] [PubMed]
  203. Song, Y.; Qu, K.; Xu, C.; Ren, J.; Qu, X. Visual and quantitative detection of copper ions using magnetic silica nanoparticles clicked on multiwalled carbon nanotubes. Chem. Commun. 2010, 46, 6572–6574. [Google Scholar] [CrossRef]
  204. Li, W.; Chen, B.; Zhang, H.; Sun, Y.; Wang, J.; Zhang, J.; Fu, Y. BSA-stabilized Pt nanozyme for peroxidase mimetics and its application on colorimetric detection of mercury(II) ions. Biosens. Bioelectron. 2015, 66, 251–258. [Google Scholar] [CrossRef] [PubMed]
  205. Guo, Y.; Deng, L.; Li, J.; Guo, S.; Wang, E.; Dong, S. Hemin−Graphene Hybrid Nanosheets with Intrinsic Peroxidase-like Activity for Label-free Colorimetric Detection of Single-Nucleotide Polymorphism. ACS Nano 2011, 5, 1282–1290. [Google Scholar] [CrossRef]
  206. Qu, K.; Shi, P.; Ren, J.; Qu, X. Nanocomposite Incorporating V2O5Nanowires and Gold Nanoparticles for Mimicking an Enzyme Cascade Reaction and Its Application in the Detection of Biomolecules. Chem. A Eur. J. 2014, 20, 7501–7506. [Google Scholar] [CrossRef]
  207. Thiramanas, R.; Jangpatarapongsa, K.; Tangboriboonrat, P.; Polpanich, D. Detection of Vibrio cholerae Using the Intrinsic Catalytic Activity of a Magnetic Polymeric Nanoparticle. Anal. Chem. 2013, 85, 5996–6002. [Google Scholar] [CrossRef]
  208. Liu, M.; Zhao, H.; Chen, S.; Yu, H.; Quan, X. Stimuli-responsive peroxidase mimicking at a smart graphene interface. Chem. Commun. 2012, 48, 7055. [Google Scholar] [CrossRef]
  209. Chen, L.; Sha, L.; Qiu, Y.; Wang, G.; Jiang, H.; Zhang, X. An amplified electrochemical aptasensor based on hybridization chain reactions and catalysis of silver nanoclusters. Nanoscale 2015, 7, 3300–3308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  210. Taghdisi, S.M.; Danesh, N.M.; Lavaee, P.; Emrani, A.S.; Ramezani, M.; Abnous, K. A novel colorimetric triple-helix molecular switch aptasensor based on peroxidase-like activity of gold nanoparticles for ultrasensitive detection of lead(ii). RSC Adv. 2015, 5, 43508–43514. [Google Scholar] [CrossRef]
  211. Weerathunge, P.; Ramanathan, R.; Shukla, R.; Sharma, T.K.; Bansal, V. Aptamer-Controlled Reversible Inhibition of Gold Nanozyme Activity for Pesticide Sensing. Anal. Chem. 2014, 86, 11937–11941. [Google Scholar] [CrossRef]
  212. Ding, N.; Yan, N.; Ren, C.; Chen, X. Colorimetric Determination of Melamine in Dairy Products by Fe3O4 Magnetic Nanoparticles−H2O2−ABTS Detection System. Anal. Chem. 2010, 82, 5897–5899. [Google Scholar] [CrossRef] [PubMed]
  213. Zhang, J.; Zhuang, J.; Gao, L.; Zhang, Y.; Gu, N.; Feng, J.; Yang, D.; Zhu, J.; Yan, X. Decomposing phenol by the hidden talent of ferromagnetic nanoparticles. Chemosphere 2008, 73, 1524–1528. [Google Scholar] [CrossRef]
  214. Jiang, J.; Zou, J.; Zhu, L.; Huang, L.; Jiang, H.; Zhang, Y. Degradation of Methylene Blue with H2O2 Activated by Peroxidase-Like Fe3O4 Magnetic Nanoparticles. J. Nanosci. Nanotechnol. 2011, 11, 4793–4799. [Google Scholar] [CrossRef]
  215. Gao, Z.; Xu, M.; Lu, M.; Chen, G.; Tang, D. Urchin-like (goldcore) & (platinumshell) nanohybrids: A highly efficient peroxidase mimetic system for insitu amplified colorimetric immune assay. Biosens. Bioelectron. 2015, 70, 194–201. [Google Scholar]
  216. Kim, M.; Kim, M.S.; Kweon, S.H.; Jeong, S.; Kang, M.H.; Kim, M.I.; Lee, J.; Doh, J. Simple and Sensitive Point-of-Care Bioassay System Based on Hierarchically Structured Enzyme-Mimetic Nanoparticles. Adv. Heal. Mater. 2015, 4, 1311–1316. [Google Scholar] [CrossRef]
  217. Gao, L.; Zhuang, J.; Nie, L.; Zhang, J.; Zhang, Y.; Gu, N.; Wang, T.; Feng, J.; Yang, D.; Perrett, S.; et al. Intrinsic peroxidase-like activity of ferromagnetic nanoparticles. Nat. Nanotechnol. 2007, 2, 577–583. [Google Scholar] [CrossRef]
  218. Kim, M.I.; Ye, Y.; Woo, M.-A.; Lee, J.; Park, H.G. A Highly Efficient Colorimetric Immunoassay Using a Nanocomposite Entrapping Magnetic and Platinum Nanoparticles in Ordered Mesoporous Carbon. Adv. Heal. Mater. 2014, 3, 36–41. [Google Scholar] [CrossRef] [PubMed]
  219. Tian, Z.; Li, J.; Zhang, Z.; Gao, W.; Zhou, X.; Qu, Y. Highly sensitive and robust peroxidase-like activity of porous nanorods of ceria and their application for breast cancer detection. Biomaterials 2015, 59, 116–124. [Google Scholar] [CrossRef]
  220. Asati, A.; Santra, S.; Kaittanis, C.; Nath, S.; Perez, J.M. Oxidase-Like Activity of Polymer-Coated Cerium Oxide Nanoparticles. Angew. Chem. 2009, 121, 2344–2348. [Google Scholar] [CrossRef]
  221. Tao, Y.; Lin, Y.; Huang, Z.; Ren, J.; Qu, X. Incorporating graphen eoxide and gold nanoclusters: A synergistic catalyst with surprisingly high peroxidase like activity over abroad pH range and its application for cancer cell detection. Adv. Mater. 2013, 25, 2594–2599. [Google Scholar] [CrossRef] [PubMed]
  222. Maji, S.K.; Mandal, A.K.; Nguyen, K.T.; Borah, P.; Zhao, Y. Cancer Cell Detection and Therapeutics Using Peroxidase-Active Nanohybrid of Gold Nanoparticle-Loaded Mesoporous Silica-Coated Graphene. ACS Appl. Mater. Interfaces 2015, 7, 9807–9816. [Google Scholar] [CrossRef]
  223. Pratt, A.; MacRae, I.J. The RNA-induced Silencing Complex: A Versatile Gene-silencing Machine. J. Biol. Chem. 2009, 284, 17897–17901. [Google Scholar] [CrossRef] [Green Version]
  224. Wang, Z.; Liu, H.; Yang, S.H.; Wang, T.; Liu, C.; Cao, Y.C. Nanoparticle-based artificial RNA silencing machinery for antiviral therapy. Proc. Natl. Acad. Sci. USA 2012, 109, 12387–12392. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Zhang, Y.; Wang, F.; Liu, C.; Wang, Z.; Kang, L.; Huang, Y.; Dong, K.; Ren, J.; Qu, X. Nanozyme Decorated Metal–Organic Frameworks for Enhanced Photodynamic Therapy. ACS Nano 2018, 12, 651–661. [Google Scholar] [CrossRef]
  226. Atilgan, A.; Islamoglu, T.; Howarth, A.J.; Hupp, J.T.; Farha, O.K. Detoxification of aS ulfur Mustard Simulant Using a BODIPY-Functionalized Zirconium Based Metal Organic Framework. ACS Appl. Mater. Interfaces 2017, 9, 24555–24560. [Google Scholar] [CrossRef]
  227. Chen, Z.-X.; Liu, M.-D.; Zhang, M.-K.; Wang, S.-B.; Xu, L.; Li, C.-X.; Gao, F.; Xie, B.-R.; Zhong, Z.-L.; Zhang, X.-Z. Interfering with Lactate-Fueled Respiration for Enhanced Photodynamic Tumor Therapy by a Porphyrinic MOF Nanoplatform. Adv. Funct. Mater. 2018, 28, 1803498. [Google Scholar] [CrossRef]
  228. Vaupel, P.; Harrison, L. Tumor Hypoxia: Causative Factors, Compensatory Mechanisms, and Cellular Response. Oncologist 2004, 9, 4–9. [Google Scholar] [CrossRef] [Green Version]
  229. Liang, M.; Fan, K.; Zhou, M.; Duan, D.; Zheng, J.; Yang, D.; Feng, J.; Yan, X. H-ferritinnanocaged doxorubicin nanoparticles specifically target and kill tumors with a single-dose injection. Proc. Natl. Acad. Sci. USA 2014, 111, 14900–14905. [Google Scholar] [CrossRef] [Green Version]
  230. Ziech, D.; Franco, R.; Pappa, A.; Panayiotidis, M.I. Reactive Oxygen Species (ROS)Induced genetic and epigenetic alterations in human carcinogenesis. Mutat. Res. Mol. Mech. Mutagen. 2011, 711, 167–173. [Google Scholar] [CrossRef]
  231. Wang, Z.; Zhang, Y.; Ju, E.; Liu, Z.; Cao, F.; Chen, Z.; Ren, J.; Qu, X. Biomimetic nanoflowers by self-assembly of nanozymes to induce intracellular oxidative damage against hypoxic tumors. Nat. Commun. 2018, 9, 1–14. [Google Scholar] [CrossRef]
  232. Cai, S.; Qi, C.; Li, Y.; Han, Q.; Yang, R.; Wang, C. PtCo bimetallic nanoparticles with high oxidase-like catalytic activity and their applications for magnetic-enhanced colorimetric biosensing. J. Mater. Chem. B 2016, 4, 1869–1877. [Google Scholar] [CrossRef]
  233. Dugan, L.L.; Gabrielsen, J.K.; Yu, S.P.; Lin, T.-S.; Choi, D.W. Buckminsterfuller enol Free Radical Scavengers Reduce Excitotoxic and Apoptotic Death of Cultured Cortical Neurons. Neurobiol. Dis. 1996, 3, 129–135. [Google Scholar] [CrossRef] [Green Version]
  234. Dugan, L.L.; Turetsky, D.M.; Du, C.; Lobner, D.; Wheeler, M.; Almli, C.R.; Shen, C.K.-F.; Luh, T.-Y.; Choi, D.W.; Lin, T.-S. Carboxyfullerenes as neuroprotective agents. Proc. Natl. Acad. Sci. USA 1997, 94, 9434–9439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  235. Ali, S.S.; Hardt, J.I.; Quick, K.L.; Kim-Han, J.S.; Erlanger, B.F.; Huang, T.-T.; Epstein, C.J.; Dugan, L.L. A biologically effective fullerene (C60) derivative with superoxide dismutase mimetic properties. Free. Radic. Biol. Med. 2004, 37, 1191–1202. [Google Scholar] [CrossRef] [PubMed]
  236. Kim, C.K.; Kim, T.; Choi, I.-Y.; Soh, M.; Kim, D.; Kim, Y.-J.; Jang, H.; Yang, H.-S.; Kim, J.Y.; Park, H.-K.; et al. Ceria Nanoparticles that can Protect against Ischemic Stroke. Angew. Chem. Int. Ed. 2012, 51, 11039–11043. [Google Scholar] [CrossRef]
  237. Chen, J.; Patil, S.; Seal, S.; McGinnis, J.F. Rare earth nanoparticles prevent retinal degeneration induced by intracellular peroxides. Nat. Nanotechnol. 2006, 1, 142–150. [Google Scholar] [CrossRef] [PubMed]
  238. Dowding, J.M.; Song, W.; Bossy, K.; Karakoti, A.; Kumar, A.; Kim, A.; Bossy, B.; Seal, S.; Ellisman, M.H.; Perkins, G.; et al. Ceriumoxide nanoparticles protect against Aβ-induced mitochondrial fragmentation and neuronal cell death. Cell Death Differ. 2014, 21, 1622–1632. [Google Scholar] [CrossRef] [PubMed]
  239. Hensley, K.; Robinson, K.A.; Gabbita, S.; Salsman, S.; Floyd, R.A. Reactive oxygen species, cell signaling, and cell injury. Free. Radic. Biol. Med. 2000, 28, 1456–1462. [Google Scholar] [CrossRef]
  240. Gechev, T.S.; Van Breusegem, F.; Stone, J.M.; Denev, I.; Laloi, C. Reactive oxygen species as signals that modulate plant stress responses and programmed cell death. Bioessays 2006, 28, 1091–1101. [Google Scholar] [CrossRef] [PubMed]
  241. Rhee, S.G. Redox signaling: Hydrogen peroxide as intracellular messenger. Exp. Mol. Med. 1999, 31, 53–59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  242. Berlett, B.S.; Stadtman, E.R. Protein Oxidation in Aging, Disease, and Oxidative Stress. J. Biol. Chem. 1997, 272, 20313–20316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  243. Barnham, K.J.; Masters, C.L.; Bush, A.I. Neurodegenerative diseases and oxidative stress. Nat. Rev. Drug Discov. 2004, 3, 205–214. [Google Scholar] [CrossRef]
  244. Matés, J.M.; Pérez-Gómez, C.; De Castro, I.N. Antioxidant enzymes and human diseases. Clin. Biochem. 1999, 32, 595–603. [Google Scholar] [CrossRef]
  245. Hotamisligil, G.S. Inflammation and metabolic disorders. Nature 2006, 444, 860–867. [Google Scholar] [CrossRef] [PubMed]
  246. Cordy, A.; Yeh, K.N. Blue dye identification on cellulosic fibers: Indigo, logwood and Prussian blue. J. Am. Inst. Conserv. 1984, 24, 33–39. [Google Scholar] [CrossRef]
  247. Thompson, D. Management of Thallium Poisoning. Clin. Toxicol. 1981, 18, 979–990. [Google Scholar] [CrossRef]
  248. Cheng, L.; Gong, H.; Zhu, W.; Liu, J.; Wang, X.; Liu, G.; Liu, Z. PEGylated Prussian blue nanocubes as a theragnostic agent for simultaneous cancer imaging and photothermal therapy. Biomaterials 2014, 35, 9844–9852. [Google Scholar] [CrossRef]
  249. Cai, X.; Gao, W.; Ma, M.; Wu, M.; Zhang, L.; Zheng, Y.; Chen, H.; Shi, J.A. Prussian Blue Based Core Shell Hollow Structured Mesoporous Nanoparticleasa Smart Theragnostic Agent with Ultrahigh pH Responsive Longitudinal Relativity. Adv. Mater. 2015, 27, 6382–6389. [Google Scholar] [CrossRef] [PubMed]
  250. Zhang, W.; Hu, S.; Yin, J.-J.; He, W.; Lu, W.; Ma, M.; Gu, N.; Zhang, Y. Prussian Blue Nanoparticles as Multienzyme Mimetics and Reactive Oxygen Species Scavengers. J. Am. Chem. Soc. 2016, 138, 5860–5865. [Google Scholar] [CrossRef] [PubMed]
  251. Bone, R.C.; Balk, R.A.; Cerra, F.B.; Dellinger, R.P.; Fein, A.M.; Knaus, W.A.; Schein, R.M.; Sibbald, W.J. Definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis. Chest 1992, 101, 1644–1655. [Google Scholar] [CrossRef] [Green Version]
  252. Lenz, A.; Franklin, G.A.; Cheadle, W.G. Systemic inflammation after trauma. Injury 2007, 38, 1336–1345. [Google Scholar] [CrossRef]
  253. Sharma, R.; Tepas, J.J.; Hudak, M.L.; Mollitt, D.L.; Wludyka, P.S.; Teng, R.J.; Premachandra, B.R. Neonatal gut barrier and multiple organ failure: Role of endotoxin and proinflammatory cytokines insepsis and necrotizing enterocolitis. J. Pediatric Surg. 2007, 42, 454–461. [Google Scholar] [CrossRef] [PubMed]
  254. Quick, K.L.; Ali, S.S.; Arch, R.; Xiong, C.; Wozniak, D.; Dugan, L.L. A carboxyfullerene SOD mimetic improves cognition and extends the lifespan of mice. Neurobiol. Aging 2008, 29, 117–128. [Google Scholar] [CrossRef]
  255. Song, Y.; Wang, X.; Zhao, C.; Qu, K.; Ren, J.; Qu, X. Label-Free Colorimetric Detection of Single Nucleotide Polymorphism by Using Single-Walled Carbon Nanotube Intrinsic Peroxidase-Like Activity. Chem. A Eur. J. 2010, 16, 3617–3621. [Google Scholar] [CrossRef]
  256. Ragg, R.; Natalio, F.; Tahir, M.N.; Janssen, H.; Kashyap, A.; Strand, D.; Strand, S.; Tremel, W. Molybdenum Trioxide Nanoparticles with Intrinsic Sulfite Oxidase Activity. ACS Nano. 2014, 8, 5182–5189. [Google Scholar] [CrossRef] [PubMed]
  257. Huang, Y.; Zhao, M.; Han, S.; Lai, Z.; Yang, J.; Tan, C.; Ma, Q.; Lu, Q.; Chen, J.; Zhang, X.; et al. Growth of Au Nanoparticles on 2D Metalloporphyrinic Metal-Organic Framework Nanosheets Used as Biomimetic Catalysts for Cascade Reactions. Adv. Mater. 2017, 29, 1700102. [Google Scholar] [CrossRef] [PubMed]
  258. Zhao, Y.; Huang, Y.; Zhu, H.; Zhu, Q.; Xia, Y. Three-in-One: Sensing, Self-Assembly, and Cascade Catalysis of Cyclodextrin Modified Gold Nanoparticles. J. Am. Chem. Soc. 2016, 138, 16645–16654. [Google Scholar] [CrossRef] [PubMed]
  259. Huo, M.; Wang, L.; Chen, Y.; Shi, J. Tumor-selective catalytic nanomedicine by nanocatalyst delivery. Nat. Commun. 2017, 8, 1–12. [Google Scholar] [CrossRef]
  260. Neri, S.; Martin, S.G.; Pezzato, C.; Prins, L.J. Photoswitchable Catalysis by a Nanozyme Mediated by a LightSensitive Cofactor. J. Am. Chem. Soc. 2017, 139, 1794–1797. [Google Scholar] [CrossRef] [Green Version]
  261. Wan, W.-L.; Lin, Y.-J.; Chen, H.-L.; Huang, C.-C.; Shih, P.-C.; Bow, Y.-R.; Chia, W.-T.; Sung, H.-W. In Situ Nanoreactor for Photosynthesizing H2 Gas to Mitigate Oxidative Stress in Tissue Inflammation. J. Am. Chem. Soc. 2017, 139, 12923–12926. [Google Scholar] [CrossRef]
  262. Ye, H.; Yang, K.; Tao, J.; Liu, Y.; Zhang, Q.; Habibi, S.; Nie, Z.; Xia, X. An Enzyme-Free Signal Amplification Technique for Ultrasensitive Colorimetric Assay of Disease Biomarkers. ACS Nano 2017, 11, 2052–2059. [Google Scholar] [CrossRef] [PubMed]
  263. Lin, Y.; Xu, C.; Ren, J.; Qu, X. Using Thermally Regenerable Cerium Oxide Nanoparticles in Biocomputing to Perform Label-free, Resettable, and Colorimetric Logic Operations. Angew. Chem. Int. Ed. 2012, 51, 12579–12583. [Google Scholar] [CrossRef]
  264. Li, W.-P.; Su, C.-H.; Chang, Y.-C.; Lin, Y.-J.; Yeh, C.-S. Ultrasound-Induced Reactive Oxygen Species Mediated Therapy and Imaging Using a Fenton Reaction Activable Polymersome. ACS Nano 2016, 10, 2017–2027. [Google Scholar] [CrossRef]
Figure 1. Sources, properties, mimicking types, and analytical capabilities of nanozymes.
Figure 1. Sources, properties, mimicking types, and analytical capabilities of nanozymes.
Materials 14 05965 g001
Figure 2. Opportunities in the field of nanozymes.
Figure 2. Opportunities in the field of nanozymes.
Materials 14 05965 g002
Figure 3. PtNPs/GO nanozymes for detecting cancer cells with calorimetric strategy.
Figure 3. PtNPs/GO nanozymes for detecting cancer cells with calorimetric strategy.
Materials 14 05965 g003
Table 1. Classification of nanozymes.
Table 1. Classification of nanozymes.
Sl. NoNanozymesSubtypesFeaturesReference
1.Peroxidase mimicsIron-basedThe Fe3O4 NPs, which are magnetic, have functions such as imitating the intrinsic peroxidase. The MNP nanozymes have the occurring mechanism which is known as ping pong catalysis and is suggested in the kinetics studies.[35]
Vanadium-basedV2O5 nanowire-based peroxidase mimics were the first demonstration done. Other research included the peculiar vanadium haloperoxidase imitating the functions of V2O5 wires which are nano, along with their anti-biofouling and marine applications.[37,38]
Based on noble metalMulti metallic NPs of the noble metals (Pd,Ag,Pt) which are known as peroxidase imitates and are utilized for antibodies, therapy, and biosensing.[44,45,46,47,48]
Carbon-basedThey possess pH, temp and hydrogen peroxide concentration dependent functions. These have been possessed by nanotubes which have a single wall and oxides of graphene.[79,80]
Based on metal–organic framework2D MOFs are believed to exhibit high functions of catalysis as compared to the analogs of 3D, hence giving effective sensibility for the detection of biomolecules.[98]
2.Oxidase mimicsGold-basedAccording to the kinetics measurements, mechanism of Eley–Rideal was suggested for AuNP-based imitates of oxidase.[101]
Copper-basedNanoparticles that contained copper were also used as imitates of oxidase. For example, Goximitating composites of Cu2O or polypyrrole were accounted for the oxidative catalysis of glucose for creating hydrogen peroxidase in fundamental terms.[102]
Molybdenum-basedIt has been reported that the molybdenum trioxidenanoparticles can mimic sulfite oxidase for converting it to sulfate beneath the terms of physiology.[103]
Based on platinumSome research linked with PtNPs as the imitates of ferroxidase for oxidizing them were noted. Examples such as, Zhang, Knez, and collaborators used apoferritin which is a light chain as the platform for PtNPs to get ready.[104]
3.Catalase mimics--------------There are many nanomaterials such as metal oxides, metals, and PB which exhibit the type of activities linked with catalase. Pt and Pd were demonstrated for possessing the good imitating functions of catalase compared to those of gold and silver.[105,106,107,108,109,110,111,112,113]
4.Superoxide dismutase (SOD) mimicsCarbon-basedC60[C(COOH)2]3 comprised of symmetry linked with C3 has been approved for possessing more properties such as acting against oxidation.[125]
Cerium-basedNanoceria was classified as one of the first nanomaterials possessing SOD mimicking activity. These have been allocated to the shuttle of electrons between the mixed states of oxidation.[127,128]
Melanin-basedThe mixture of the hydrochloride of dopamine along with NH3 in the ethanol and water led to the synthesis of melanin nanoparticles.[132]
5.Hydrolase mimicsCarbon-basedAqueous-solvent fullerene worked with the corrosive carboxylic, known as C 60-1, and it was exhibited for catalyzing the phosphodiester cleavage obligation occurring in DNA and illuminated through the light.[134,135]
Monolayer functionalized AuNP basedAuNPs worked along with monolayers, which are catalytic throughout the bonds of gold and silver, are amongst the very first nanomaterials
imitating as hydrolases that deserve acknowledgments.
[139,140,141]
MOF-basedMOFs based on Zr are used as imitates of phosphor triesterase for the occurrence of cleavage of the bond of phosphate ester of chemical warfare agent.[142,143,144,145,146,147,148,149,150]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Sindhu, R.K.; Najda, A.; Kaur, P.; Shah, M.; Singh, H.; Kaur, P.; Cavalu, S.; Jaroszuk-Sierocińska, M.; Rahman, M.H. Potentiality of Nanoenzymes for Cancer Treatment and Other Diseases: Current Status and Future Challenges. Materials 2021, 14, 5965. https://doi.org/10.3390/ma14205965

AMA Style

Sindhu RK, Najda A, Kaur P, Shah M, Singh H, Kaur P, Cavalu S, Jaroszuk-Sierocińska M, Rahman MH. Potentiality of Nanoenzymes for Cancer Treatment and Other Diseases: Current Status and Future Challenges. Materials. 2021; 14(20):5965. https://doi.org/10.3390/ma14205965

Chicago/Turabian Style

Sindhu, Rakesh K., Agnieszka Najda, Prabhjot Kaur, Muddaser Shah, Harmanpreet Singh, Parneet Kaur, Simona Cavalu, Monika Jaroszuk-Sierocińska, and Md. Habibur Rahman. 2021. "Potentiality of Nanoenzymes for Cancer Treatment and Other Diseases: Current Status and Future Challenges" Materials 14, no. 20: 5965. https://doi.org/10.3390/ma14205965

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop