• Journal of Inorganic Materials
  • Vol. 38, Issue 1, 43 (2023)
Xuetong WU1、2, Ruofei ZHANG2, Xiyun YAN2, and Kelong FAN2、*
Author Affiliations
  • 11. Sino-Danish College, University of Chinese Academy of Sciences, Beijing 101408, China
  • 22. Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
  • show less
    DOI: 10.15541/jim20220578 Cite this Article
    Xuetong WU, Ruofei ZHANG, Xiyun YAN, Kelong FAN. Nanozyme: a New Approach for Anti-microbial Infections[J]. Journal of Inorganic Materials, 2023, 38(1): 43 Copy Citation Text show less

    Abstract

    Bacteria and viruses always posed a threat to human health. Most impressively, SARS-CoV-2 has raged around the world for almost three years, causing huge loss to human health. Facing increasing challenges of drug-resistance and poor treatment efficacy, new solutions are urgently needed to combat pathogenic microorganisms. Recently, nanozymes with intrinsic enzyme-like activities emerged as a promising new type of “antibiotics”. Nanozymes exhibit superior antibacterial and antiviral activities under physiological conditions by efficiently catalyzing generation of a large number of reactive oxygen species. Moreover, enhanced therapeutic effects are achieved in nanozyme-based therapy aided by the unique physicochemical properties of nanomaterials such as photothermal and photodynamic effects. This paper reviews the latest research progress in the field of anti-microbial nanozymes, systematically summarizes and analyzes the principles of nanozymes in the treatment of bacteria and viruses from a mechanistic point of view. An outlook on the future direction and the challenges of new anti-microbial infection nanomaterials are proposed to provide inspiration for developing next generation anti-microbial nanozymes.

    Bacteria and viruses have long been an important threat to human life and health. For example, SARS- CoV-2, which has caused a global storm of disease transmission[1], Staphylococcus aureus, which causes pneumonia, Helicobacter pylori, which causes gastritis, and Salmonella, which causes enteritis. To illustrate, pneumonia due to bacteria accounts for about one third of the causes of pneumonia[2]. These hazardous pathogens are even more damaging to vulnerable people such as children and patients. Pneumonia is estimated to account for 21% of deaths among children, based on an analysis of epidemiological data from 42 countries. This is similar to the findings of 19% of the WHO in 2000[3]. WHO indicates that pneumonia is in the top three factors contributing to death in children under five years of age. Acquired pneumonia has been a very common and threatening occurrence of nosocomial infections[4-5], with methicillin-resistant Staphylococcus aureus (MRSA) evading antimicrobial drugs and causing people to death[6]. The growth of resistant bacteria has also posed a significant challenge to the development of antimicrobial drugs. The dose of antibiotics increases with the increase of behavior of drug-resistant bacteria, and the overuse of antibiotics leading to the emergence of more resistant bacteria[7-8]. Moreover, a huge public health crisis is brewing along with the stagnation of antibiotic research and development. It is necessary to develop new antibacterial and antiviral drugs that are effective and do not generate bacterial resistance.

    The development of nanotechnology has brought new inspiration to human medicine, whether it is nanovaccines[9-10] or nanodrug delivery agents[11-12], giving hope that this new technology can be used to solve difficult problems[13]. Importantly, since their special properties were discovered in 2007[14], as a new role of nanomaterials, nanozymes have been given a new mission.

    Nanozymes are a class of nanomaterials with intrinsic enzyme-like activity. Since the first discovery of the intrinsic peroxidase-like activity of Fe3O4 nanoparticles by Gao et al. in 2007, nanozymes with other enzyme-like activities, including catalase-like[15], superoxide dismutase- like[16], oxidase-like[17] and glutathione peroxidase-like[18] activities have been subsequently revealed. The catalytic efficiency of nanozymes is close to or even better than natural enzymes, allowing them to be utilized in various catalysis-related aspects. Researchers have used nanozymes for various fields such as disease diagnosis[19], disease treatment[20-21] and detection[22-24]. In addition, many types of nanozymes have been used for antibacterial and antiviral treatments: metals[25], metal oxides[26], carbon dots[27], mesoporous silica[28], etc.

    These various effective nanozymes identified at present have some commonalities in the mechanisms of action. This review summarizes and analyzes the latest research advances in the field of antibacterial and antiviral nanozymes from a mechanistic point of view (Fig. 1). It is hoped that the introduction of antibacterial and antiviral strategies of nanozymes can provide inspiration for the design and development of novel antibacterial and antiviral drugs for challenging conditions.

    Mechanism of anti-microbial infections of nanozymes[25⇓⇓-28]

    Figure 1.Mechanism of anti-microbial infections of nanozymes[25-28]

    1 Mechanism of Anti-microbial Infections of Nanozymes

    1.1 Generation of reactive oxygen species

    Nanozymes are so called enzyme-mimics because the mechanism by which their function is primarily enzymatic catalysis. Nanozymes with peroxidase-like activity catalyze the generation of lethal hydroxyl radicals from H2O2, one of the most damaging reactive oxygen species (ROS) that destroys biological molecules and thus causes the death of the target organisms. Nanozymes with oxidase-like activity typically reduce O2 to H2O2 or H2O, and can also produce the toxic superoxide radical O2·-. Nanozymes with catalase-like activity catalyze the generation of O2 from H2O2 can improve the oxygen-depleted environment or be used in combination with other catalytic activities to enhance the target effect. Superoxide dismutase-like nanozymes can convert superoxide radicals into H2O2 and O2, which can be used as an important antioxidant to protect normal tissues from ROS attack[29-31]. In the treatment of anti-microbial infections, aggressive ROS generated by nanozymes generally play a key role.

    Huang et al.[32] have designed a bimetallic quasi-MOF nanozyme (Q-MOFCe0.5) with peroxidase-like activity. Such nanosheets featured by a layered heterogeneous junctional interface can continuously produce ROS to achieve effective killing of Escherichia coli (99.74%) and Staphylococcus aureus (99.35%) in vitro, and the morphology analysis clearly that the attacked bacteria lose their normal shapes (Fig. 2). Optimizing the catalytic activity of nanozymes by doping, modification, etc. can improve their antimicrobial properties. Pan et al.[33] compared the peroxidase-like activity of vigorous chitosan grafted Fe-doped carbon dots (CS@Fe/CDs) and carbon dots without Fe-doping (CS@CDs), found that nanozyme based on CS@Fe/CDs performs better catalytic activity. Authors pointed out that the mechanism of the peroxidase-like activity is the increased electron density in the active center and the reduced resistance of the steric sites due to the abundant carboxyl groups on the surface of the nanozyme, leading to an increased Fenton- like reaction. In antimicrobial experiments, CS@Fe/CDs showed satisfying killing effect on Staphylococcus aureus and Pseudomonas aeruginosa in the presence of H2O2.

    Antibacterial killing effect of a nanozyme, Q-MOFCe0.5[32](a, b) Plate count results of the antibacterial effects of Q-MOFCe0.5 on E. coli (a) and S. aureus (b); (c, d) Statistical results of the corresponding bacterial viability rates of E. coli (c) and S. aureus (d); (e) The live (SYTO-9, green)/dead (PI, red) staining results of E. coli and S. aureus; (f) Scanning electron microscope (SEM) results of bacterial samples treated by nanozymesColorful figures are available on website

    Figure 2.Antibacterial killing effect of a nanozyme, Q-MOFCe0.5[32](a, b) Plate count results of the antibacterial effects of Q-MOFCe0.5 on E. coli (a) and S. aureus (b); (c, d) Statistical results of the corresponding bacterial viability rates of E. coli (c) and S. aureus (d); (e) The live (SYTO-9, green)/dead (PI, red) staining results of E. coli and S. aureus; (f) Scanning electron microscope (SEM) results of bacterial samples treated by nanozymesColorful figures are available on website

    Through rational design, nanozymes with diverse enzymatic activities may achieve the protection of normal tissues while killing microorganisms. Cao et al.[26] successfully constructed an iron phosphate nanozyme- hydrogel (FePO4-HG) with multiple enzymatic activities. This nanozyme exhibited peroxidase-like activity under acidic conditions to kill bacteria, but showed superoxide dismutase-like and catalase-like activity under neutral to weak alkaline conditions to protect normal tissues from exogenous H2O2 damage. In addition, the nanozyme possessing positive charges and large pores is able to trap bacteria within a certain range, thereby enhancing its killing effect, which was confirmed in animal experiments.

    In order to cope with SARS-CoV-2, researchers have also found inspiration from nanozymes. Wang et al.[34] have designed a highly effective anti-viral TiO2 supported Ag atoms single atom nanozyme (Ag-TiO2 SAN) (99.65%) by loading atomically dispersed Ag atoms onto TiO2 nanoparticles. This single atom nanozyme can be phagocytosed by macrophages and co-localized with lysosomes through strong interaction of Ag atoms to the cysteine and asparagine of viral spike proteins, and has strong adsorption to SARS-CoV-2 (Fig. 3). In acidic environment, the high peroxidase-like activity exhibited by Ag-TiO2 SAN gives it the ability to generate ROS and to kill SARS-CoV-2.

    Characterization and ROS generation ability of a nanozyme, Ag-TiO2 SAN[34](a, b) Transmission electron microscope (TEM) images of Ag-TiO2 SAN co-localized with lysosomes of mouse (RAM 264.7) and human (THP-1) macrophages; (c, d) Ag-TiO2 SAN enhanced ROS generation. THP-1: a kind of human monocytic leukemia cellColorful figures are available on website

    Figure 3.Characterization and ROS generation ability of a nanozyme, Ag-TiO2 SAN[34](a, b) Transmission electron microscope (TEM) images of Ag-TiO2 SAN co-localized with lysosomes of mouse (RAM 264.7) and human (THP-1) macrophages; (c, d) Ag-TiO2 SAN enhanced ROS generation. THP-1: a kind of human monocytic leukemia cellColorful figures are available on website

    Nanozymes also were applied to create anti- microbial fabrics based on their antibacterial and antiviral properties. Inspired by the structure of SARS-CoV-2, Ni et al.[35] proposed a new idea to fight superbugs with super viruses by designing a triple-shell porous graphitic carbon nitride (g-C3N4) embedded with cobalt nanoparticles based on the morphology of coronaviruses. This artificial virus has oxidase-like activity, can penetrate the bacterial membrane barrier and cause ROS damage to bacteria. The artificial virus also has photodynamic and photothermal properties triggered by light due to the good photothermal conversion properties of the material, and can also be enriched and recycled by using the magnetic properties of the material. This kind artificial nanozyme-based virus has been used for river water circulation disinfection and in vivo wound healing.

    1.2 Combination therapy based on multifunctional properties of nanozymes

    Nanozymes are a special class of materials that possess both the enzymatic activity and the properties of inorganic nanomaterials inherent in them. Many studies therefore applied nanozymes in the direction of synergistic therapy.

    1.2.1 LSPR combined enzyme-like activities therapy

    When the size of nanoparticles is much smaller than the wavelength of incident light, and the frequency of incident light is the same as the frequency of free electron oscillation, it can cause the collective oscillation of free electrons on the surface, and this phenomenon is named local surface plasmon resonance (LSPR)[36]. Metal nanoparticles such as Au, Ag, Cu, etc. have the property of absorbing light in a specific range. In a different interpretation of the mechanism, a special class of nanozymes, plasmonic nanoparticles, were found to have LSPR properties in addition to the good catalytic activity of nanoparticles[37]. Liao et al.[38] found that the prepared Au nanozymes and Cu metal-organic framework nanosheets (Cu-MOFNs) had faster reaction kinetics under LSPR excitation. The dark-field scattering spectra of this nanoparticle indicated that its LSPR promotes hot electron transfer, resulting in faster reaction kinetics, and it showed good results in antimicrobial therapy and wound healing. The LSPR method has also been used by researchers in studies for the treatment of tumors[39].

    Doping affects the LSPR property of nanozymes. The impurities or holes introduced by doping can change the LSPR properties of the nanozymes due to the changed carrier density. Doping with plasma metals such as Au enhances the LSPR field, which enhances the light absorption properties of the material and enhances the thermal transport rate so that the nanozymes can have higher photothermal conversion efficiency. Meanwhile, the hot electron driving of plasma provides higher generation rate of energetic electron-hole pairs for the catalytic properties of the nanozymes, which improves the catalytic efficiency of the nanozymes[40-41]. Compared to MoO3-x, the Au-doped MoO3-x (Au/MoO3-x) plasmonic- semiconductor hybrid was 3.6 times more effective in terms of peroxidase-like activity. In addition, the Au/MoO3-x exhibited a better photothermal conversion efficiency (52.40%) than the undoped MoO3-x (41.11%) due to the enhanced LSPR effect of the doped material. Through higher peroxidase-like activity and better photothermal property, the material achieved effective removal of MRSA bacteria (99.76%)[26].

    1.2.2 Photothermal combination therapy

    The photothermal effect refers to the interaction of photon energy with the lattice when the material is irradiated by incident light, and the absorbed light energy is converted into thermal motion energy of the lattice, causing the rise of temperature. Materials with high photothermal conversion efficiency usually have complex microstructure and high light absorption capacity[42]. Taking advantage of the good photothermal properties of the nanomaterials, Chen et al.[43] designed 4-mercaptophenylboronic acid (MPBA)-functionalized CuSe nanoprobes (CuSeNPs@MPBA) based on chemiluminescence (CL) for the construction of a multifunctional platform for the effective inactivation of bacteria. The nanozyme have peroxidase-like activity to catalyze the conversion of flash CL to luminescent CL, improving the stability and accuracy of the assay, as well as excellent and stable photothermal performance, with outstanding photothermal conversion efficiency (37%) that can significantly increase the temperature under NIR irradiation to cause inactivation of E. coli (99.2%) and S. aureus (99.8%).

    However, photothermal therapy is often accompanied by concerns about whether the heat causes damage to normal tissues. Li et al.[44] have synthesized hollow mesoporous Prussian Blue nanoparticles (HMPBNPs) with peroxidase-like activity for photothermal therapy, which requires not only a lower effective temperature (48 ℃) than the usual effective temperature for photothermal therapy (>53 ℃), but also a lower concentration of H2O2. These properties allow for the effective killing of both E. coli which represent Gram-negative bacteria and S. aureus which represent Gram-positive bacteria under mild conditions.

    Similarly, Lin et al.[45] constructed Bacitracin-modified dextran-MoSe2 nanosheets (AMP/dex-MoSe2 NSs) for low-temperature photothermal antimicrobial activity in the NIR irradiation. The lower degree of photothermal heating also increases the efficiency of the production of hydroxyl radicals by peroxidase-like catalysis which is one of the most active bactericidal components. The nanozyme can also bind to bacterial membranes by electrostatic adsorption to achieve a multimodal antimicrobial effect.

    1.2.3 Photodynamic combination therapy

    As a material with catalytic activity and good modifiable properties, the nanozymes coupled with photosensitizers can endow them with photodynamic properties that lead to photosensitization of oxygen to produce ROS, enhancing their catalytic activity and achieving the killing effect on microorganisms[46]. The shortcomings of nanozymes for anti-infection therapy mainly stem from their inadequate production of lethal ROS, both in terms of their yield and the time they can be sustained release. To overcome this limit, Gao et al.[47] optimized a new photoactivated nanozyme, Ag/Bi2MoO6 (Ag/BMO), by charge separation engineering, which not only exhibits catalytically enhanced photodynamic properties under NIR (II) irradiation, but also peroxidase-like properties on a sustained level. This is due to the fact that the appropriate light and acidity make it easier to separate the light-triggered electron-hole pairs to produce ROS (Fig. 4), while the electron transfer generated by laser irradiation at specific wavelengths makes it easier for the reversible Mo5+/Mo6+ change to take place, thus exhibiting enhanced catalytic activity. To improve the antibacterial activity of nanozymes, He et al.[48] have designed a multifunctional nanozyme using polydopamine (PDA)- modified copper oxide (CuxO-PDA). The peroxidase-like activity of the nanozyme can be enhanced under near infrared irradiation (NIR). The negative charge on the surface of this nanozyme changes to a positive charge as the neutral or basic conditions change to acidic conditions. This property allows bacteria with a negative surface charge to be trapped by electrostatic interactions. Moreover, after the nanozyme adhering to the bacterial membrane, the bacteria aggregate under the driving of the nanozyme under NIR irradiation, and this ability to lock and capture the bacteria allows the nanozyme to trap bacteria within the killing range of ROS, thus enhancing the antibacterial effect.

    Characterization, Ag+ release, and ROS generation ability of Ag/BMO NPs through photodynamic combination therapy[47](a) SEM image of Ag/BMO NPs; (b) Percentage release of Ag+ at pH 7.35 and pH 6.75 showing photo-enhanced ROS generation ability of Ag/BMO; (c) 1O2 detection result by probe of single oxygen sensor green; (d) ESR spectra of 1O2; (e) ·OH detection result by MB indicator; (f) ESR spectra of ·OH. L: 1064 nm laser (1 W·cm-2, 10 min) Colorful figures are available on website

    Figure 4.Characterization, Ag+ release, and ROS generation ability of Ag/BMO NPs through photodynamic combination therapy[47](a) SEM image of Ag/BMO NPs; (b) Percentage release of Ag+ at pH 7.35 and pH 6.75 showing photo-enhanced ROS generation ability of Ag/BMO; (c) 1O2 detection result by probe of single oxygen sensor green; (d) ESR spectra of 1O2; (e) ·OH detection result by MB indicator; (f) ESR spectra of ·OH. L: 1064 nm laser (1 W·cm-2, 10 min) Colorful figures are available on website

    Using different catalytic activities, Loukanov et al.[49] found that the histidine-modified carbon nanodots exhibited ideal inhibition of bacterial proliferation under visible light irradiation using their oxidase-like catalytic properties.

    The positively charged carbon nanodots tend to attract negatively charged bacteria, thus causing severe damage to bacterial membranes and achieving antibacterial effects.

    1.3 Nanozymes based on cascaded reaction

    Many nanozymes have multi-enzyme activities, such as peroxidase-like, catalase-like, and oxidase-like activities. These multi-enzyme activities can be combined to perform various functions, such as killing pathogens while protecting normal tissues from harsh environments. Secondly, nanozymes can be cascaded with natural enzymes to achieve a triggered reaction or amplify the signal. Cascade reactions can be used to solve complex problems when the signal is insufficient or the concentration of the reaction substrate is inadequate.

    Du et al.[50] used iron oxide nanoparticles coated with a glucose oxidase shell (Fe3O4-GOx) to implement a cascade reaction to treat difficult-to-heal wounds. Fe3O4-GOx behaves glucose oxidase (GOx), catalase (CAT) and peroxidase (POD)-like activities (Fig. 5). Under acidic conditions, the GOx/POD cascade produces hydroxyl radicals, which inhibit bacterial growth and aid wound healing. Under neutral conditions, the GOx/CAT cascade produces oxygen, which addresses hypoxia and oxidative stress in difficult-to-heal wounds and accelerates normal tissue proliferation. This approach allows regrowth of difficult-to-heal wounds with minimal toxicity to normal tissues.

    In vitro antimicrobial performance of Fe3O4-GOx based on cascaded reaction[50](a) 2',7'-Dichlorofluorescein staining images of E. coli and MRSA; (b) Growth curves of MRSA; (c) Representative SEM and live/dead staining images of MRSA; (d) Crystal violet staining image and its absorbance for integrated MRSA biofilmColorful figures are available on website

    Figure 5.In vitro antimicrobial performance of Fe3O4-GOx based on cascaded reaction[50](a) 2',7'-Dichlorofluorescein staining images of E. coli and MRSA; (b) Growth curves of MRSA; (c) Representative SEM and live/dead staining images of MRSA; (d) Crystal violet staining image and its absorbance for integrated MRSA biofilmColorful figures are available on website

    In addition, the low substrate concentration requirement of the cascade reaction also gives it some advantages. For example, Zhang et al.[51] developed a cascade nanozyme (Fe2(MoO4)3@GOx) by loading glucose oxidase on the outside of a nanozyme with peroxidase-like activity. The nanozyme can solve the problem of poor therapeutic efficacy due to unstable H2O2 levels in the environment. GOx loaded on the surface digests glucose to produce gluconic acid and H2O2, which is then converted into hydroxyl radicals using the peroxidase-like activity of the nanozyme. This in vivo harmless cascade of nanozyme acts synergistically by causing starvation and generating ROS for antibacterial purposes against two typically drug-resistant bacteria, extended-spectrum beta- lactamases producing E. coli and methicillin- resistant S. aureus, both showed ideal antimicrobial efficacy (98.396% and 98.776%, respectively).

    1.4 Nanozymes based on bio-orthogonal catalysis

    Bio-orthogonal catalysis refers to catalytic reactions that can be carried out in living cells or tissues without interfering with the organism's inherent biochemical reactions. Bio-orthogonal catalysis makes it possible for catalytic processes in situ in cells or tissues to produce effective drugs without affecting the surrounding environment[52-53]. As a class of inorganic nanomaterials with catalytic activity, the good biosafety of nanozymes allows them to be used as bio-orthogonal catalytic processes. Microorganism intracellular infections a difficult problem to solve, due to their complex environment and the difficulty in adjusting the dose of antibiotics. Nanozymes can be used to solve this problem, as their stability and modifiability ensures that they are not degraded intracellularly and that the orthogonal catalysis does not have effective impact on normal cellular tissues due to high antibiotic doses.

    Joseph Hardie et al.[54] designed a nanozyme consist of mannose-functionalized gold nanoparticle and iron tetraphenyl porphyrin (FeTPP) that can be used as bio-orthogonal catalyst to treat intracellular bacterial infections of Lactobacillus sp. The nanozyme, which was used for bio-orthogonal catalysis, could be taken up by macrophages via the cell surface mannose receptor (CD206) and subsequently convert the inactive antibiotic precursor (pro-ciprofloxacin) to ciprofloxacin inside the cell, and showed better killing of Salmonella relative to non-pathogenic Lactobacillus. As a new strategy, it could also be taken as next step in typhoid and tuberculosis.

    Similarly, Akash Gupta et al.[55] used gold nanoparticles as a backbone and FeTPP as a catalyst to synthesize nanozymes (Man-NZ). The researchers modified gold nanoparticles on the surface of the RBCs, which have good biocompatibility and are susceptible to hemolyzed by bacterial toxins, through electrostatic interactions. The stability and the catalytic activity of the nanozyme was checked to be not influenced by the functionalized RBCs. Afterwards, the authors chose arylazide protected moxifloxacin (pro-Mox) as an antibiotic precursor, which was found to be well activated and acted upon by studying MRSA with E. coli, increasing the specificity of the treatment. The nanozyme can catalyze this antibiotic precursor in situ into effective antibiotic and then kill bacteria.

    Jing et al.[56] designed shape-based bio-orthogonal catalytic nanozyme that performs antibacterial activity. They fabricated a shell on the surface of the bacteria, following the shape of the bacteria, and then separated the bacteria and the shell by calcination to obtain the designed bio-orthogonal catalytic nanozyme. This nanozyme can selectively recognize the corresponding template pathogenic bacteria. As a bio-orthogonal catalyst, the nanozyme can catalyze the precursor into an active antimicrobial molecule 6 in situ (Fig. 6) and act as a bacterial killer. And this nanozyme have been used in treatment of E. coli and S. aureus infections in vivo. However, since the recognition is based on shape, it is still a great challenge to distinguish bacteria of the same shape or different subtypes of bacteria.

    S-Ab can be used as bio-orthogonal catalyst to complete shape-based selective recognition of bacteria and catalyze precursors into active antibacterial molecule[56]Colorful figure is available on website

    Figure 6.S-Ab can be used as bio-orthogonal catalyst to complete shape-based selective recognition of bacteria and catalyze precursors into active antibacterial molecule[56]Colorful figure is available on website

    2 Summary and Prospect

    As newly discovered nanomaterials with special functions, nanozymes are showing great potential for various applications (Table 1). In the fight against microbial infections, nanozymes work by relying on their enzyme-like catalytic properties, cleverly exploiting the environment to inhibit microorganisms through the production of lethal ROS. However, some individual studies indicated that antibacterial active nanozymes have different killing effects on Gram-negative and Gram- positive bacteria. The designed nanozyme kills Pseudomonas aeruginosa more effectively than S. aureus[33]. Therefore, the mechanism behind this needs to be carefully investigated.

    MechanismNanozymeCatalytic activityMechanismPathogensRef.
    Generation ROSCu-MOFOXDROSE. coli; S. aureus[25]
    Cu-CDPODROSE. coli; S. aureus[27]
    Chitosan grafted Fe-doped-carbon dots (CS@Fe/CDs)PODROSP. aeruginosa; S. aureus.[33]
    Ag-TiO2 SANPODROSSARS-CoV-2[34]
    Fmoc-diphenylalanine hydrogel-eEncapsulated PtOXD; PODROSE. coli; S. aureus[57]
    CFO@BFO nanozyme-eelCATROSE. coli; MRSA[58]
    Cu2O@CuO; Cu@Cu2S nanodotOXD; PODROSE. coli; S. aureus[59]
    FeCo@PDA NPsPODROSE. coli; S. aureus[60]
    Fe3O4@SiO2@dendritic mesoporous silica@small-Fe3O4 nanoparticlesPODROSE. coli[61]
    Mesoporous vanadium oxide nanospheresPODROSE. coli; S. aureus[62]
    Au3+-UiO-67 NMOFsOXD; PODROSE. coli; S. aureus[63]
    CS@Fe3O4POD; SOD; CATROSA. baumannii[64]
    N/Cl-CDs + Ag NPsOXDROSE. coli; S. aureus; MRSA[65]
    Cu-N-COXD; PODROSE. coli; S. aureus; B. cereus; C. albicans; MRSA[66]
    PEGMA-co-GMA-co-AAm-HBPL-MnO2 hydrogelCAT; PODROSP. aeruginosa; S. aureus; E. coli[67]
    GOx-MOF hydrogelGOx; PODROSE. coli; S. aureus[68]
    Taurine-Cu-3(PO4)(2) hybrid nanoflowerPODROSE. coli; S. aureus;B. cereus; C.albicans[69]
    FePO4-HGPOD; SOD; CATROSE. coli; S. aureus[70]
    Combination therapyAu/MoO3-xPODPhotothermal; ROS(O2•-MRSA[26]
    Cu-MOFN nanosheetPODHot electron transferred ROSS. aureus[38]
    CuSeNPs@MPBAPODPhotothermal; ROSE. coli; S. aureus[43]
    Hollow mesoporous Prussian blue nanoparticles (HMPBNPs)PODROS; PhotothermalE. coli; S. aureus[44]
    Bacitracin-functionalized dextran-MoSe2(AMP/dex-MoSe2 NSs)PODPhotothermal; ROSE. coli[45]
    MechanismNanozymeCatalytic activityMechanismPathogensRef.
    Combination therapyAg/Bi2MoO6PODPhotodynamic; ROSS. aureus[47]
    CuxO-PDAPODPhotothermal; ROSE. coli; S. aureus[48]
    Histidine-containing carbon nanodotsOXDPhotodynamic; ROSE. coli[49]
    VOx-artificial enzymeOXD, PODElectron enhanced ROSS. aureus[71]
    EM@MoS2PODPhotothermal; ROSS. aureus[72]
    LS-CuS@PVAPODPhotothermal; Photodynamic; ROSE. coli; S. aureus[73]
    D-A-conjugated COFPODPhotothermal; Photodynamic; ROSE. coli; S. aureus[74]
    Cu3SnS4 NSsPODPhotothermal; ROSE. coli; S. aureus[75]
    Mn3O4HNSs@ICGOXDPhotothermal; Photodynamic; ROSE. faecalis; E. coli; P. aeruginosa[76]
    Au NCs@PCN MOFPODPhotothermal; ROSE. coli; S. aureus[77]
    Ag (8.5%)@NiS2-xPODPhotothermal; ROSE. coli[78]
    Cascaded reactionFe3O4-GOxGOx; CAT; PODROSE. coli; S. aureus[50]
    Fe2(MoO4)3@GOxGOx; PODROSE. coli; S. aureus[51]
    ODex/gC/MoS2@Au@BSA HydrogelPODROSE. coli; S. aureus[79]
    CuO nanospheresPOD; CATROSE. coli; S. aureus[80]
    MnFe2O4@MIL/Au&GOx(MMAG)GOx; PODROSE. coli; S. aureus[81]
    Bio-orthogonal catalysisMan-NZs(Au, FeTPP)Bio-orthogonal catalysisSalmonella; Lactobacillus sp.[54]
    Man-NZBio-orthogonal catalysisE. coli; MSRA[55]
    E-Ab and S-AbBio-orthogonal catalysisE. coli; S. aureus[56]
    OthersCeO2@ZrO2HaloperoxidaseHBr-E. coli; S. aureus[82]

    Table 1.

    Nanozymes for anti-microbial infections

    In addition to the properties of enzyme-mimic, as functional inorganic nanomaterials, nanozymes can also perform anti-microbial infection functions by combining their intrinsic physiochemical properties and catalytic properties. As materials with surprisingly special properties at the nanoscale, nanozymes can possess good photothermal conversion efficiency inherently or after modification for photothermal combination therapy; nanozymes can be designed photo-triggered or composed of photosensitive materials with photodynamic properties to enhance catalytic effects. The LSPR properties of nanozymes can also be exploited to give nanozymes superior photothermal properties and enhanced catalytic activity. In addition, nanozymes can be carefully designed to achieve specific functions in complex environments. For example, cascade-based nanozymes can perform different functions at different pH or overcome the problem of unstable catalytic substrate H2O2 levels in the environment. Due to their good security, biocompatibility and catalytic properties, nanozymes can also be used as orthogonal catalysts to achieve precise in situ cell killing effects.

    Unfortunately, the mechanism of ROS production of nanozymes against microbial infections is not well elaborated and classified by researchers. In addition, there are two issues that need to be addressed in the field of anti-microbial infection application of nanozymes as follows.

    1) Antimicrobial specificity. The principle of nanozymes functioning as antibiotics in vivo is mainly to produce ROS to kill pathogens, but it needs to be discussed whether the recognition between pathogens and normal tissues by nanozymes is so good that ROS does not attack normal tissues. Moreover, whether the specificity of the nanozymes for the target pathogen is adequate to avoid the normal survival of non-target bacteria also needs to be further studied.

    2) Drug resistance. Drug resistance caused by antibiotics is one of the major issues of public healthcare nowadays. Due to the different mechanisms of effect, nanozymes mainly contribute to the removal of pathogens by means of adsorption and contact killing, ROS production, etc. These methods with no interference to biochemical and metabolic processes of bacteria have a low risk of resistance development. However, further studies are necessary support whether this property also leads to the development of drug resistance in pathogens.

    In the future, the wide range of properties of nanozymes as inorganic materials can be explored more deeply in order to make a better combination of various excellent properties including catalytic activity to make contribution in the field of anti-microbial infection.

    References

    [1] W J WIERSINGA, A RHODES, A C CHENG et al. Pathophysiology, transmission, diagnosis, and treatment of coronavirus disease 2019 (COVID-19): a review. JAMA-Journal of the American Medical Association, 782(2020).

    [2] S N GRIEF, J K LOZA. Guidelines for the evaluation and treatment of pneumonia. Primary Care, 485(2018).

    [3] R E BLACK, S S MORRIS, J BRYCE. Where and why are 10 million children dying every year?. Lancet, 2226(2003).

    [4] G A DENYS, R F RELICH. Antibiotic resistance in nosocomial respiratory infections. Clinics in Laboratory Medicine, 257(2014).

    [5] V YAP, D DATTA, M L METERSKY. Is the present definition of health care-associated pneumonia the best way to define risk of infection with antibiotic-resistant pathogens?. Infectious Disease Clinics of North America, 1(2013).

    [6] H W BOUCHER, G H TALBOT, J S BRADLEY et al. Bad bugs, no drugs: no eskape! An update from the infectious diseases society of America. Clinical Infectious Diseases, 1(2009).

    [7] J D D PITOUT, K B LAUPLAND. Extended-spectrum beta-lactamase-producing enterobacteriaceae: an emerging public-health concern. Lancet Infectious Diseases, 159(2008).

    [8] S Y C TONG, J S DAVIS, E EICHENBERGER et al. Staphylococcus aureus infections: epidemiology, pathophysiology, clinical manifestations, and management. Clinical Microbiology Reviews, 603(2015).

    [9] R YANG, J XU, L XU et al. Cancer cell membrane-coated adjuvant nanoparticles with mannose modification for effective anticancer vaccination. ACS Nano, 5121(2018).

    [10] G Z ZHU, G M LYNN, O JACOBSON et al. Albumin/vaccine nanocomplexes that assemble in vivo for combination cancer immunotherapy. Nature Communications, 8:, 1954(2017).

    [11] J LU, M LIONG, Z LI et al. Biocompatibility, biodistribution, and drug-delivery efficiency of mesoporous silica nanoparticles for cancer therapy in animals. Small, 1794(2010).

    [12] Y TIAN, S LI, J SONG et al. A doxorubicin delivery platform using engineered natural membrane vesicle exosomes for targeted tumor therapy. Biomaterials, 2383(2014).

    [13] Y YANG, Z Y LI, K YAMAGUCHI et al. Controlled fabrication of silver nanoneedles array for SERS and their application in rapid detection of narcotics. Nanoscale, 2663(2012).

    [14] L GAO, J ZHUANG, L NIE et al. Intrinsic peroxidase-like activity of ferromagnetic nanoparticles. Nature Nanotechnology, 577(2007).

    [15] B XU, H WANG, W WANG et al. A single-atom nanozyme for wound disinfection applications. Angewandte Chemie International Edition, 4911(2019).

    [16] H Q ZHAO, R F ZHANG, X Y YAN et al. Superoxide dismutase nanozymes: an emerging star for anti-oxidation. Journal of Materials Chemistry B, 6939(2021).

    [17] X HU, F Y LI, F XIA et al. Biodegradation-mediated enzymatic activity-tunable molybdenum oxide nanourchins for tumor-specific cascade catalytic therapy. Journal of the American Chemical Society, 1636(2020).

    [18] X MENG, D LI, L CHEN et al. High-performance self-cascade pyrite nanozymes for apoptosis-ferroptosis synergistic tumor therapy. ACS Nano, 5735(2021).

    [19] B JIANG, L YAN, J ZHANG et al. Biomineralization synthesis of the cobalt nanozyme in SP94-ferritin nanocages for prognostic diagnosis of hepatocellular carcinoma. ACS Applied Materials & Interfaces, 9747(2019).

    [20] D W JIANG, D L NI, Z T ROSENKRANS et al. Nanozyme: new horizons for responsive biomedical applications. Chemical Society Reviews, 3683(2019).

    [21] Z WANG, G LI, Y GAO et al. Trienzyme-like iron phosphates-based (FePOs) nanozyme for enhanced anti-tumor efficiency with minimal side effects. Chemical Engineering Journal, 125574(2021).

    [22] Y A ZHANG, Y N LI, J L ZHANG et al. Nanocage-based capture-detection system for the clinical diagnosis of autoimmune disease. Small, 2101(2021).

    [23] D DUAN, K FAN, D ZHANG et al. Nanozyme-strip for rapid local diagnosis of ebola. Biosensors & Bioelectronics, 134(2015).

    [24] X MENG, S ZOU, D LI et al. Nanozyme-strip for rapid and ultrasensitive nucleic acid detection of SARS-CoV-2. Biosensors & Bioelectronics, 114739(2022).

    [25] Z MAO, J CHEN, Y WANG et al. Copper metal organic framework as natural oxidase mimic for effective killing of gram- negative and gram-positive bacteria. Nanoscale, 9474(2022).

    [26] M CAO, Z CHANG, J TAN et al. Superoxide radical-mediated self-synthesized Au/MoO(3-x) hybrids with enhanced peroxidase-like activity and photothermal effect for anti-MRSA therapy. ACS Applied Materials & Interfaces, 13025(2022).

    [27] Y WANG, J YAO, Z CAO et al. Peroxidase-mimetic copper-doped carbon-dots for oxidative stress-mediated broad-spectrum and efficient antibacterial activity. Chemistry-a European Journal, e202(2022).

    [28] K M FUENTES, D ONNA, T RIOUAL et al. Copper upcycling by hierarchical porous silica spheres functionalized with branched polyethylenimine: antimicrobial and catalytic applications. Microporous and Mesoporous Materials, 111391(2021).

    [29] N SINGH, M A SAVANUR, S SRIVASTAVA et al. A redox modulatory Mn3O4 nanozyme with multi-enzyme activity provides efficient cytoprotection to human cells in a Parkinson's disease model. Angewandte Chemie International Edition, 14267(2017).

    [30] X MU, J WANG, Y LI et al. Redox trimetallic nanozyme with neutral environment preference for brain injury. ACS Nano, 1870(2019).

    [31] Y AI, J YOU, J GAO et al. Multi-shell nanocomposites based multienzyme mimetics for efficient intracellular antioxidation. Nano Research, 2644(2021).

    [32] L HUANG, D W SUN, H PU. Photosensitized peroxidase mimicry at the hierarchical 0D/2D heterojunction-like quasi metal-organic framework interface for boosting biocatalytic disinfection. Small, 2200178(2022).

    [33] T PAN, H CHEN, X GAO et al. Engineering efficient artificial nanozyme based on chitosan grafted Fe-doped-carbon dots for bacteria biofilm eradication. Journal of Hazardous Materials, 128996(2022).

    [34] D WANG, B ZHANG, H DING et al. TiO2 supported single Ag atoms nanozyme for elimination of SARS-COV2. Nano Today, 101243(2021).

    [35] Y NI, J WANG, M WANG et al. COVID-19-inspired "artificial virus" to combat drug-resistant bacteria by membrane-intercalation- photothermal-photodynamic multistage effects. Chemical Engineering Journal, 137322(2022).

    [36] S SARINA, E R WACLAWIK, H ZHU. Photocatalysis on supported gold and silver nanoparticles under ultraviolet and visible light irradiation. Green Chemistry, 1814(2013).

    [37] M MU, S WEN, S HU et al. Putting surface-enhanced Raman spectroscopy to work for nanozyme research: methods, materials and applications. Trac-Trends in Analytical Chemistry, 116603(2022).

    [38] X LIAO, Q XU, H SUN et al. Plasmonic nanozymes: localized surface plasmonic resonance regulates reaction kinetics and antibacterial performance. Journal of Physical Chemistry Letters, 312(2022).

    [39] L LI, J YANG, J WEI et al. SERS monitoring of photoinduced- enhanced oxidative stress amplifier on Au@carbon dots for tumor catalytic therapy. Light-Science & Applications, 286(2022).

    [40] D WANG, Y FANG, W YU et al. Significant solar energy absorption of mxene Ti3C2Tx nanofluids via localized surface plasmon resonance. Solar Energy Materials and Solar Cells, 110850(2021).

    [41] O ZANDI, A AGRAWAL, A B SHEARER et al. Impacts of surface depletion on the plasmonic properties of doped semiconductor nanocrystals. Nature Materials, 710(2018).

    [42] X GENG, D ZHANG, Z ZHENG et al. Integrated multifunctional device based on Bi2S3/Pd: localized heat channeling for efficient photothermic vaporization and real-time health monitoring. Nano Energy, 105700(2021).

    [43] X CHEN, X WANG, Y FANG et al. Long-lasting chemiluminescence- based poct for portable and visual pathogenic detection and in situ inactivation. Analytical Chemistry, 8382(2022).

    [44] Y LI, Y ZHU, C WANG et al. Mild hyperthermia induced by hollow mesoporous prussian blue nanoparticles in alliance with a low concentration of hydrogen peroxide shows powerful antibacterial effect. Molecular Pharmaceutics, 819(2022).

    [45] T LIN, G JIANG, D LIN et al. Bacitracin-functionalized dextran- mose 2 with peroxidase-like and near-infrared photothermal activities for low-temperature and synergetic antibacterial applications. ACS Applied Biomaterials, 2347(2022).

    [46] S P SONGCA, Y ADJEI. Applications of antimicrobial photodynamic therapy against bacterial biofilms. International Journal of Molecular Sciences, 3209(2022).

    [47] C GAO, T ZHANG, N YANG et al. POD nanozyme optimized by charge separation engineering for light/pH activated bacteria catalytic/photodynamic therapy. Signal Transduction and Targeted Therapy, 86(2022).

    [48] S HE, Y FENG, Q SUN et al. Charge-switchable CuxO nanozyme with peroxidase and near-infrared light enhanced photothermal activity for wound antibacterial application. ACS Applied Materials & Interfaces, 25042(2022).

    [49] A LOUKANOV, A KURIBARA, S NIKOLOVA et al. Light-activated oxidize-mimicking nanozyme for inhibition of pathogenic escherichia coli. Microscopy Research and Technique, 1949(2022).

    [50] X DU, B JIA, W WANG et al. pH-switchable nanozyme cascade catalysis: a strategy for spatial-temporal modulation of pathological wound microenvironment to rescue stalled healing in diabetic ulcer. Journal of Nanobiotechnology, 12(2022).

    [51] Y ZHANG, D LI, Y XU et al. Application of a cascaded nanozyme in infected wound recovery of diabetic mice. ACS Biomaterials Science & Engineering, 1522(2022).

    [52] S FEDELI, J IM, S GOPALAKRISHNAN et al. Nanomaterial-based bioorthogonal nanozymes for biological applications. Chemical Society Reviews, 13467(2021).

    [53] S S NGUYEN, J A PRESCHER. Developing bioorthogonal probes to span a spectrum of reactivities. Nature Reviews Chemistry, 476(2020).

    [54] J HARDIE, J M MAKABENTA, A GUPTA et al. Selective treatment of intracellular bacterial infections using host cell-targeted bioorthogonal nanozymes. Materials Horizons, 1489(2022).

    [55] A GUPTA, R DAS, J M MAKABENTA et al. Erythrocyte-mediated delivery of bioorthogonal nanozymes for selective targeting of bacterial infections. Materials Horizons, 3424(2021).

    [56] J NIU, L WANG, T CUI et al. Antibody mimics as bio-orthogonal catalysts for highly selective bacterial recognition and antimicrobial therapy. ACS Nano, 15841(2021).

    [57] J CHEN, S ZHANG, X CHEN et al. A self-assembled fmoc-diphenylalanine hydrogel-encapsulated Pt nanozyme as oxidase-and peroxidase-like breaking ph limitation for potential antimicrobial application. Chemistry-a European Journal, e2021(2022).

    [58] Q DENG, L ZHANG, X LIU et al. Magnetoelectrically ignited nanozyme-eel for combating bacterial biofilms. Chemical Communications, 7634(2022).

    [59] Y HE, M YIN, J SUN et al. Excellent catalytic properties of luminescent Cu@Cu2S nanozymes and their antibacterial applications. Chemical Communications, 2995(2022).

    [60] F KUANG, Y CHEN, W SHAN et al. Biomimetic FeCo@Pd a nanozyme platform with fenton catalytic activity as efficient antibacterial agent. Journal of Materials Chemistry B, 5582(2022).

    [61] Y HUANG, D LIU, R GUO et al. Magnetic-controlled dandelion-like nanocatalytic swarm for targeted biofilm elimination. Nanoscale, 6497(2022).

    [62] P LI, Y FENG, D CHENG et al. Self-template synthesis of mesoporous vanadium oxide nanospheres with intrinsic peroxidase-like activity and high antibacterial performance. Journal of Colloid and Interface Science, 435(2022).

    [63] M M PAN, Y OUYANG, Y L SONG et al. Au3+-functionalized UIO-67 metal-organic framework nanoparticles: O2-(center dot-) and center dot OH generating nanozymes and their antibacterial functions. Small, 2200548(2022).

    [64] W WANG, Z WU, P SHI et al. Antibacterial effect of chitosan-modified Fe3O4 nanozymes on Acinetobacter baumannii. Journal of Microbiology and Biotechnology, 263(2022).

    [65] J ZHU, Q LI, X LI et al. Simulated enzyme activity and efficient antibacterial activity of copper-doped single-atom nanozymes. Langmuir, 6860(2022).

    [66] J ZHU, X LI, X WU et al. Nanocomposite of Ag nanoparticles and deep eutectic solvent-derived carbon dots with oxidase mimicking activity as synergistic bactericidal agent. Letters in Applied Microbiology, 684(2022).

    [67] C TU, H LU, T ZHOU et al. Promoting the healing of infected diabetic wound by an anti-bacterial and nano-enzyme-containing hydrogel with inflammation-suppressing, ROS-scavenging, oxygen and nitric oxide-generating properties. Biomaterials, 121597(2022).

    [68] S ZHANG, F DING, Y LIU et al. Glucose-responsive biomimetic nanoreactor in bacterial cellulose hydrogel for antibacterial and hemostatic therapies. Carbohydrate Polymers, 119615(2022).

    [69] S G YILMAZ, A DEMIRBAS, Z KARAAGAC et al. Synthesis of taurine-Cu3(PO4)2 hybrid nanoflower and their peroxidase-mimic and antimicrobial properties. Journal of Biotechnology, 96(2022).

    [70] Z Y LIAO, W W GAO, N N SHAO et al. Iron phosphate nanozyme-hydrogel with multienzyme-like activity for efficient bacterial sterilization. ACS Applied Materials & Interfaces, 18170(2022).

    [71] L LI, S CAO, Z WU et al. Modulating electron transfer in vanadium-based artificial enzymes for enhanced ROS-catalysis and disinfection. Advanced Materials, 2108646(2022).

    [72] T SHI, Z RUAN, X WANG et al. Erythrocyte membrane- enveloped molybdenum disulfide nanodots for biofilm elimination on implants via toxin neutralization and immune modulation. Journal of Materials Chemistry B, 1805(2022).

    [73] Y XIE, C GAN, Z LI et al. Fabrication of a lignin-copper sulfide-incorporated PVA hydrogel with near-infrared-activated photothermal/photodynamic/peroxidase-like performance for combating bacteria and biofilms. ACS Biomaterials Science & Engineering, 560(2022).

    [74] G P YANG, X L MENG, S J XIAO et al. Construction of D-a-conjugated covalent organic frameworks with enhanced photodynamic, photothermal, and nanozymatic activities for efficient bacterial inhibition. ACS Applied Materials & Interfaces, 28289(2022).

    [75] Y YANG, C WANG, N WANG et al. Photogenerated reactive oxygen species and hyperthermia by Cu3SnS4 nanoflakes for advanced photocatalytic and photothermal antibacterial therapy. Journal of Nanobiotechnology, 195(2022).

    [76] X ZHANG, Y MIN, Q ZHANG et al. Functionalized Mn3O4 nanosheets with photothermal, photodynamic, and oxidase-like activities triggered by low-powered near-infrared light for synergetic combating multidrug-resistant bacterial infections. Advanced Healthcare Materials, 2200121(2022).

    [77] X ZHAO, L CHANG, Y HU et al. Preparation of photocatalytic and antibacterial MOF nanozyme used for infected diabetic wound healing. ACS Applied Materials & Interfaces, 18194(2022).

    [78] Y ZHANG, D YANG, W GUO et al. A multifunctional hydrogel dressing with antibacterial properties for effective wound healing. Dalton Transactions, 6817(2022).

    [79] Y LI, R FU, Z DUAN et al. Injectable hydrogel based on defect-rich multi-nanozymes for diabetic wound healing via an oxygen self-supplying cascade reaction. Small, 2200165(2022).

    [80] P WANG, L PENG, J LIN et al. Enzyme hybrid virus-like hollow mesoporous CuO adhesive hydrogel spray through glucose-activated cascade reaction to efficiently promote diabetic wound healing. Chemical Engineering Journal, 128901(2021).

    [81] X ZHOU, S ZHANG, Y LIU et al. Antibacterial cascade catalytic glutathione-depleting MOF nanoreactors. ACS Applied Materials & Interfaces, 11104(2022).

    [82] Q LUO, Y LI, X HUO et al. Stabilizing ultrasmall ceria-cluster nanozyme for antibacterial and antibiofouling applications. Small, 2107401(2022).

    Xuetong WU, Ruofei ZHANG, Xiyun YAN, Kelong FAN. Nanozyme: a New Approach for Anti-microbial Infections[J]. Journal of Inorganic Materials, 2023, 38(1): 43
    Download Citation