Skip to main content

Single-atom nanozymes shines diagnostics of gastrointestinal diseases

Abstract

Various clinical symptoms of digestive system, such as infectious, inflammatory, and malignant disorders, have a profound impact on the quality of life and overall health of patients. Therefore, the chase for more potent medicines is both highly significant and urgent. Nanozymes, a novel class of nanomaterials, amalgamate the biological properties of nanomaterials with the catalytic activity of enzymes, and have been engineered for various biomedical applications, including complex gastrointestinal diseases (GI). Particularly, because of their distinctive metal coordination structure and ability to maximize atom use efficiency, single-atom nanozymes (SAzymes) with atomically scattered metal centers are becoming a more viable substitute for natural enzymes. Traditional nanozyme design strategies are no longer able to meet the current requirements for efficient and diverse SAzymes design due to the diversification and complexity of preparation processes. As a result, this review emphasizes the design concept and the synthesis strategy of SAzymes, and corresponding bioenzyme-like activities, such as superoxide dismutase (SOD), peroxidase (POD), oxidase (OXD), catalase (CAT), and glutathione peroxidase (GPx). Then the various application of SAzymes in GI illnesses are summarized, which should encourage further research into nanozymes to achieve better application characteristics.

Graphical abstract

Introduction

Hospitalizations for digestive system disorders have risen absolutely as a proportion of all hospitalizations over the past two decades, with the largest rises in disorders including, intestinal infections and pancreatitis, posing significant implications for the quality of life and overall health of patients [1, 2]. Tumors of the digestive system (e.g., liver cancer [3], pancreatic [4], and colorectal cancer [5]) also account for a large percentage of solid tumors. Despite the constant updating of therapeutic approaches, the incidence and mortality of certain diseases, such as gastric, esophageal, and colorectal cancers, are increasing annually [6]. Although significant progress has been made in the detection and treatment of digestive illnesses since the advent of endoscopic techniques [7], novel therapeutic approaches are still required to address the complexity and diversity of disease types. Catalytic nanomaterials known as nanozymes have features similar to those of enzymes and come in a variety of sizes, shapes, and surface arrangements. These artificial enzymes exhibit both great catalytic stability and high catalytic efficiency. It has been demonstrated that nanozymes possess the biocatalytic activity of naturally occurring enzymes, such as SOD [8,9,10], OXD [11, 12], POD [13], CAT [9, 14] and so on. The focus of nanozyme research is currently on single-atom nanozymes (SAzymes), a novel class of nanozymes with the benefits of high atom utilization, high catalytic activity, cheap production cost, and great selectivity. SAzymes are also gradually being developed for the treatment of digestive diseases, such as liver cancer and cirrhosis [15], pancreatitis [16], and the control of intestinal inflammation [17].

Herein, an emphasis on their unprecedent design concepts to optimize dispersion, stability, and enzyme activity has been made. Based on it, we give a brief summary of the function of the biocatalytic-like activity of SAzymes and the corresponding application innovation of theme in the field of gastrointestinal disorders. Finally, in response to the problems and controversies of SAzymes in terms of bioactivity and biological applications, we point out the future opportunities and challenges of SAzymes and propose potential solutions and future directions to accelerate the development of SAzymes in biomedical fields (Fig. 1). Finally, an overview of the biological uses of SAzymes, along with issues and disagreements surrounding them, is provided. The outlook and challenges confronting SAzymes in the forthcoming period are also highlighted along with potential solutions and future pathways to expedite their development within the biomedical industry.

Fig. 1
figure 1

Schematic overview. Recent developments in nanozymes in terms of material preparation, enzyme-like catalytic activity and biological applications of nanozymes in Gastrointestinal (GI) diseases

Design principles and synthesis strategy of SAzymes

The preparation of frameworks, such as metal–organic frameworks (MOFs), covalent organic frameworks (COFs), hydrogen-bonded organic frameworks (HOFs) frameworks with various active centers, has actually been linked to a wide range of multi-enzyme activities. Because of this, scientists have experimented with a number of design approaches in an effort to maximize the dispersion, stability, especially catalytic activity in recent years. This section focuses on the remarkable advancements made recently in engineering techniques for activity tuning, such as modifications to the SAzymes framework and optimizations for SAzymes active centers. Based on this, the particular synthesis processes and modalities of SAzymes are described in order to further analyze their synthesis strategy (Fig. 2).

Fig. 2
figure 2

Optimization strategies for single-atom nanozymes at both framework structure and active center levels

Optimization of framework structure

It is essential to use an appropriate supporting substrate while synthesizing SAzymes. This is because, as a result of aggregating during catalysis, instability in the framework may cause the core metal atom to become inactive. The atoms that comprise the framework have the ability to interact with the atom of the core metal, so influencing the metal’s activity and, consequently, the total catalytic activity of nanozymes. Consequently, in this section, we investigate how the activity of nanozymes is affected by the optimization of various kinds of framework materials. MOFs, COFs, and HOFs are examples of crystalline porous materials that typically have excellent porosity and structural composition designability. They are also thought to have controllable morphologies, modifiable backbones, flexible active site structures, tunable charge transfer pathways, and designable porosities [18, 19].

MOFs are unique because of their ability to fine-tune pore structure and their variety in design. They are created by coordinating metal ions with organic ligands [20]. They are typically made using a one-pot self-assembly method that creates metal-containing nodes on the spot. Ma et al. [21] introduced Zr-MOF nanozymatic coatings into natural bacterial cellulose (BC) nanofibers, and this design enabled the nanozymes to have multilayered macro–micro pores, which led to the full exposure of catalytic active sites and exhibited excellent enzyme-mimicking catalytic activity (Fig. 3A). The same is true for CMZM, which has multi-enzymatic activity and can be utilized to reverse immunosuppressive TME. Additionally, the enzyme-like activities of CMZM improve the effectiveness of multimodal imaging-guided CDT and PDT treatments [14] (Fig. 3B). The bimetallic MOF pathway has been applied to the optimization of MOFs. The Cu, Mn bimetallic nanozymes (Cu-TCPP-Mn) prepared from MOFs have combined high catalytic properties for SOD and CAT, and can synergistically scavenge ROS (Fig. 3C) [22]. Another crystalline porous materials, COF, constructed through covalently linked organic units, show excellent properties for synthesizing SAzymes [23,24,25]. The one-dimensional iron porphyrin covalent organic skeleton (COF-CNT) coated on carbon nanotubes nanozymes were able to produce reactive oxygen species (ROS). Furthermore, the peroxidase-like activity of COF-CNT was much enhanced in the presence of an electric field, suggesting that the COF framework had a discernible impact on the enzymatic activity of the nanozymes [26]. When loaded with enzymes, pore congestion or partial clogging prevents COF from being used. HOFs are less stable than MOFs and COFs because they rely on hydrogen bonding interactions to stabilize their structure. However, because hydrogen bonding is typically weaker than ionic or covalent bonding, HOFs have an additional deformability that can be used to create flexible porous HOF materials [27].

Fig. 3
figure 3

Copyright 2023. Wiley–VCH. B Illustration of CMZM preparation process. Reproduced with permission [14]. Copyright 2023. Wiley–VCH. C Schematic illustration of the design and synthesis of Cu-TCPP-Mn nanozyme for myocardial injury treatment. a) The bimetallic Cu-TCPP-Mn nanozyme was fabricated by embedding manganese and copper into the porphyrin via solvothermal method, followed by sonication into small MOF nanodots. b) Cu-TCPP-Mn nanozyme retained cascade activity that has been shown to scavenge ROS, inhibit inflammation, reduce myocardium fibrosis and promote constructive remodeling and vascularization in MI and I/R injury animal models. Reproduced with permission [22]. Copyright 2023. Ivyspring International Publisher. D Schematic illustration of OxgeMCC-r. a) Schematic illustration of OxgeMCC-r. OxgeMCC-r consists of catalytically active single-atom Ru site anchored in MCC with outer PVP protection layer. b) Partial molecular structure of OxgeMCC-r with active single-atom Ru site serving as catalase-like nanozyme for oxygen generation. c) Multicomponent coordination interactions within the OxgeMCC-r SAE. Reproduced with permission [28]. Copyright 2020. Springer Nature

Optimization scheme for MOF structures. A Illustration of the differences in diffusion between the less macroporous MOF nanozyme xerogel (left) and the microporous MOF nanozyme aerogel (right). Reproduced with permission [21].

Increasing the catalytic activity of SAzymes through optimization of these frameworks has become a major area of research interest. An essential technique for creating N-doped carbon materials for stabilizing and spreading substrates supported by enzymes is the pyrolysis of MOFs. The nanozymes prepared either by doping Ru into the framework [28] or by encapsulating ferritin in ZIF-8 [29] have excellent stability and catalytic activity (Fig. 3D). It is also possible to tune the Fe–N coordination by optimizing the precursor type and pyrolysis temperature. Triple peroxidase-like activity and triple catalytic sites were added by Li et al. [30] to complement the synthesis of a unique core–shell nanocomposite, Prussian blue@Fe-covalent organic framework@Au (PB@Fe-COF@Au) (Fig. 4A). On the other hand, the strategy of synthesizing porous N-doped carbon nano-enzymes using COF as a precursor increased the exposure of the catalytic site and exhibited stronger peroxidase-like activity [31]. Innovatively grown platinum nanoparticles and ultramicro rhodium nanoparticles on the surface of COF NPs, Gao et al. [32] and Zhang et al. [33], respectively, served as catalase mimics in situ. The optimized nanozymes demonstrated high affinity for the catalytic substrates and excellent peroxidase-mimicking activity (Fig. 4B). In addition, the functional groups in the COF can act as Lewis acid–base sites within the porous skeleton to mimic the functions of amino acid residues and tailor the pore microenvironment around the active center, thus enhancing the catalytic activity of MOF-based nanozymes [34]. Constructing ionic HOFs is one of the strategies for HOF framework optimization. PFC-33 [35], the first anionic HOF synthesized, exhibits synergistic photodynamic and chemical antimicrobial efficiencies (Fig. 4C). Based on this, Tong et al. [36] created three light-responsive HOFs nanomaterials with various pore structures. Next, they mimicked light-responsive oxidative enzymes using structurally well-defined hydrogen-bonded organic frameworks (HOFs), and by creating isostructured HOFs, they were able to demonstrate the importance of pore environments in mediating the activity of oxidative enzyme-like enzymes. This demonstrated that, in addition to active centers, pore environments have a significant impact on the activity of the nanozymes (Fig. 4D). They suggested that the difference in pore channels could regulate the activity of isotope-like oxidase in structured HOFs. Ionic HOFs with anionic or cationic backbones were designed and synthesized to attract noncovalently bonded counterions in the pore channels. Currently, another optimization strategy for HOF frameworks is to add modifying substances to HOFs to improve the porosity and photodynamic efficiency. Yin et al. [37] performed surface modification by adding rabies virus glycoprotein (RVG)29. Not coincidentally, the final shell nanostructures were created by Liu et al. [38] utilizing a progressive ligand grafting technique (Fig. 4E). It is evident that altering the morphology or spatial arrangement of HOFs affects the catalytic activity of the nanocomplexes built using them as frameworks; this may be due to the catalytic sites being completely exposed through physical or chemical mechanisms. Similar to the dual action of COF-MOFs, the combination of HOFs with MOFs also showed superior biological and chemical activities to those of MOFs alone [39].

Fig. 4
figure 4

Copyright 2023. American Chemical Society. B Schematic illustration showing a) the synthesis of COF and COF-Rh, and b the strategy for Cys detection based on regulating the POD-like activity of COF-Rh. Reproduced with permission [33]. Copyright 2023. Elsevier. C Preparation of polyHOF by PSM followed by polymerization. Reproduced with permission [35]. Copyright 2020. Wiley–VCH. D a) Schematic of hydrogen bonding assembly of HOFs nanozymes. b The oxidase-like catalysis using TMB as the substrate. The colors used are: red for Oatom; blue for Natom; gray for Catom; white for Hatom. Reproduced with permission [36]. Copyright 2023. Wiley–VCH. E Fabrication of core–shell UCNPs@PFC-55. a) Fabrication of core–shell UCNPs@PFC-55. b, e TEM images of as-synthesized oleate-stabilized β-NaYF4:Yb,Er UCNPs; c, f PDI-C anchoring UCNPs-L; d, g the final products UCNPs@PFC-55 with inorganic UCNPs “cores” and organic PFC-55 “shells”. Reproduced with permission [38]. Copyright 2021. Wiley–VCH

Optimization scheme for COF and HOF structures. A Procedure for the Synthesis of PB@Fe-COF@Au. Reproduced with permission [30].

Carbon dots, a new type of carbon nanomaterial with luminescent properties, possess a rich source of raw materials, facile surface modification, low toxicit [40] and high biocompatibility [41]. Doping or loading individual metal atoms can boost the catalytic activity of carbon dots, including peroxidase activity, oxidase mimetic activity, and catalytic activity [10, 42]. To enhance the biosafety, controllability, and catalytic activity of single-atom nanozymes, carbon dots were introduced during the synthesis of SAzymes [43, 44]. By using a solvothermal technique, Han et al. [45] created carbon dots (CDs)-loaded single-atom iron nanozymes (ph-CDs-Fe SAzymes), which showed high POD activity (Fig. 5A). Yu et al. [46] used polyethyleneimine (PEI) to modify carbon dots, receiving better optical characteristics (Fig. 5B). Furthermore, by varying the reaction temperature, it was possible to optimize the various coordination structures of CD, thereby altering the bioactivity of the composites with SAzymes. For example, CD-loaded copper single-atom nanozymes with various coordination structures and peroxidase-like characteristics could be synthesized at various temperatures (Fig. 5C) [47]. The aforementioned research has demonstrated a connection between the carbon dot form and the catalytic site variation, which in turn influences the biological activity of the composites.

Fig. 5
figure 5

Copyright 2023. Wiley–VCH. B The schematic diagram delineates how CD-PEI-DOX-siMRP1 delivers doxorubicin to tumors and antagonizes chemoresistance by hindering drug efflux through knocking down MRP1 expression. Conversion of GSH to GSSG and subsequent ROS increase by CD-PEI oxidase and peroxidase activity further impairs MRP1 function. Collectively, CD-PEI-DOX-siMRP1 was capable of delivering drugs efficiently to tumor entities and retaining them in cells by hindering outflux of MRP1 through synergistic delivery of siRNA and perturbation of GSH-ROS balance. Reproduced with permission [46]. Copyright 2023. Dove Medical Press Ltd. C Synthesis and morphologies of Cu-CDs-300. a) Scheme of the calcining procedure for Cu-CDs-300 as an example. b) TEM and HRTEM images of Cu-CDs-300. c) Typical HAADF-STEM image of distributed single Cu atoms (orange circles) in Cu-CDs-300. Reproduced with permission [47]. Copyright 2023. Wiley–VCH

Optimization scheme for carbon dots structures. A Peroxidase-like activity of ph-CDs-Fe SAzyme. a) Schematic illustration of POD-like activity of CDs, Fe-CDs SAzyme, and ph-CDs-Fe SAzyme. b) POD-like activity of CDs, CDs-Fe, and ph-CDs-Fe SAzyme. c) Degradation of methylene blue (MB, 12.5 µg mL−1, pH 5.0) by CDs, Fe-CDs SAzyme, and ph-CDs-Fe SAzyme in the presence of H2O2 (5 mM). Reproduced with permission [45].

Because of their atomic characteristics and spatial arrangement, we found that materials that consist of crystalline frameworks, such as MOFs, COFs, and HOFs, are crucial for optimizing single-atom nanozyme complexes. MOFs structures possess a tendency to collapse in high-temperature environments, which is not conducive to a high degree of dispersion, and lead to changes in the oxidation state, phase purity and atomic level characterization of the metal, leaving the stability of the resulting nanozymes uncontrolled. Experimental data on COFs have collectively shown that COFs as a framework or carrier may cause complete or partial pore blockage on their surface, which significantly impacts the exposure of the active sites of the nanozymes. The inherent instability and structural recalcitrance are the primary obstacles to HOF development [48]. Previous research has conclusively demonstrated that the incorporation of functional groups typically shapes into new hydrogen bonds and alters the topology, which may have an effect on the function of SAzymes.

Optimization of active center

The experimental findings reveal an unbreakable link between particular atomic metal centers and catalytic activity. Ou et al. [49] suspected that material properties are affected by the spatial location of monatomic atoms. To examine their photocatalytic antibacterial activity, they created Cu single-atom-site nanozymes both within and outside of polyheptazinimide nano-(PHI) platforms. The results show that the interlayer-localized copper material (CuL/PHI) has a broader antimicrobial effect on a wide range of bacterial strains compared to other spatially arranged materials, and can achieve the same antimicrobial effect as antibiotics. During the catalytic process, Rh(V)N4 and the preferentially formed Rh(V)-O-N4 structure can act as active centers, enabling the SAzyme to exhibit excellent POD-like activity. This structure improves catalyst utilization and significantly reduces reaction energy through the “two-sided oxygen chain” catalytic reaction pathway [50]. According to Wang et al. [51], metal-H2O2 interactions may have an impact on the catalytic activity of CAT. It was demonstrated that Ir-N4 had the strongest interaction with H2O2, the lowest H2O2 decomposition barrier, and the highest CAT-like activity out of the five active centers based on platinum group metals (M-N4, M = Ir, Ru, Rh, Pt, and Pd). In a manner similar to this, Cheng et al. [5] synthesized various active sites of metal-N to produce artificial metalloenzymes and a library of metalloenzyme imitators. Among the artificial metal nanozymes, they found that the Fe-like metal-centered nanozymes had the highest OXD-like activity, metal-centered nanozymes with a copper-like center demonstrated the highest level of POD-like activity.

Precious metals have strong catalytic activity and noble metal-precious metal bimetallic nanocatalysts have been reported. Fan et al. [53] wrapped a thin layer of palladium around gold nanorods (AuNRs) as the core to form Au@PdNRs, which showed notable oxidase-like activities under dark environment and plasma resonance excitation, and the enhanced catalytic activity was mainly due to the full exposure of the Pd surface used for catalysis. Both non-precious and precious characteristics of metals are displayed by nanozymes made of both types of metals. An ultrasmall single-atom Pt/CeO2 with continuous catalytic activity was created by Yan et al. [54]. The highly ordered arrangement of metal atoms in the resultant nanocage allowed the SAzymes to have POD- and GSH peroxidase like (GSH-Px-like) catalytic activity and efficient ROS production under ultrasonic irradiation. The single-atom Pt generated a selective distribution on the crystalline surface of CeO2. Similarly, Zhong et al. [55] demonstrated that PtCu3 nanocages can also be used as copper-based nanomaterials as novel acoustic sensitizers to generate ROS under irradiation.

It can be inferred from several experiments that the synergistic action of metals in multimetallic catalysts may result in greater selectivity and catalytic efficiency than in monometallic catalysts. A Cu/Zn bimetallic atom nanoconjugate enzyme (Cu/PMCS) was created by Liu et al. [56]. The findings indicated that doping Cu atoms increased the catalytic activity and GSH depletion of this nanoconjugate enzyme, which improved its anti-tumor capacity. Additionally, according to Lv et al. [57], the peroxidase catalytic activity of nanorods containing Au and Pt bimetallic atoms was significantly higher than that of nanozymes containing single metal atoms. Therefore, it is clear that adding more metal sites will increase the catalytic activity of nanozymes. The catalytic activity of single-atom nano-enzymes is connected with the ratio of metal atoms in addition to the influence of the quantity of metal atoms. Cai et al. [58] discovered that the shape, structure, and content of the products are influenced by the Au–Pd synthesis atomic ratio. In model oxidation reactions, 0D/2D Au–Pd nanocomposites demonstrated significantly increased peroxidase-mimetic catalysis. This was theorized to be directly related to changes in the electronic structure of Au–Pd and an increase in its specific surface area ratio.

Overall, a number of experiments have been carried out recently with the aim of enhancing the enzyme-like catalytic activity of single-atom nanozymes. These experiments range from the different types of single atoms and their spatial positions corresponding to different kinds of enzyme-like activities to the interactions between polyatoms and the changes in the ratio of polymetallic atoms significantly affecting the catalytic activity, which have produced encouraging results, and we anticipate the emergence of more effective and straightforward optimization based on the results.

Synthesis strategy of SAzymes

Synthesis process

SAzymes have been synthesized in various ways, such as by constructing defect sites on metal hydroxide/oxide substrates, by creating spatially restricted domain effects on the substrate material, etc., which basically improve the interaction of metal atoms with the coordinating heteroatoms. For SAzymes, the most classical synthesis strategy is wet chemistry, including coprecipitation, impregnation, and ion exchange [59]. Using zeolite Y as a carrier, Cheng et al. [60] developed a straightforward wet impregnation technique to produce a stable nanocomposite (CeO2/Y). Tang et al. [61] developed a novel one-step wet chemical process after optimizing a number of investigations, and they used BSA to guide the formation of two-dimensional nanosheets (Fig. 6A). To further improve the stability, biocompatibility, and surface functioning of the resulting BSA-NOTA-modified MnO2 nanosheets (M-NS), they also presented an acoustic chemical synthesis method.

Fig. 6
figure 6

Copyright 2019. Wiley–VCH. B Schematic of the synthetic process for the Ni SAs@S/N-FCS. Reproduced with permission [65]. Copyright 2022. Wiley–VCH. C Schematic diagram of the synthesis process of ZnSA-HCNT. Reproduced with permission [66]. Copyright 2022. Wiley–VCH. D Illustration of the preparation process of PtTS-SAzyme. Reproduced with permission [67]. Copyright 2021. American Chemical Society. E Schematic of the Preparation Strategy for Fe–NO/NC. Reproduced with permission [68]. Copyright 2020. American Chemical Society. F Schematic showing the preparation of the SAFe-SWCNT film. Reproduced with permission [69]. Copyright 2021. Elsevier

Strategies for the synthesis of single-atom nanozymes. A Schematic illustration of two-step synthesis of M-NS. First, a novel “wet-chemical method” was applied to form 2D nanosheets under the direction of BSA. Second, a unique “sono-chemical method” was introduced to further enhance the stability, biocompatibility, and surface functionality of the M-NS. Reproduced with permission [61].

SAzymes can be prepared by direct pyrolysis for those precursor metal blocks or metal materials composed of thermodynamically unstable metals. Most types of SAzymes in the current study, especially those based on nitrogen-doped carbon materials, are mainly prepared by pyrolysis [62]. In the direct pyrolysis pathway, it is of utmost importance to break the metal–metal bonds of the precursor carriers and to minimize the escape of metal atoms at high temperatures [63]. Huang et al. [64] prepared single Fe atom nano-enzymes with Fe-N5 active sites by pyrolyzing MOFs at 900 °C under N2 atmosphere. Zhao et al. [65] pyrolyzed at 950 °C to generate Ni single-atom catalysts (Fig. 6B). Similar to this, Li et al. [66] created zinc monoatomic nanozymes through the pyrolysis and high-temperature adsorption reactions of zinc with imidazole (Fig. 6C). Coincidentally, Chen et al. [67] reversed the thermal sintering process and directly atomized platinum nanoparticles (Pt NPs) into single atoms to produce high-performance nanozymes (Fig. 6D). However, the direct pyrolysis of MOFs has its drawbacks: during the pyrolysis process, the structure of MOFs tends to collapse due to high temperatures, leading to volume contraction, which is unfavorable for the high dispersion of monatomic metals.

In order to encourage the in-situ formation of volatile metal species in metal nanoparticles and induce their anchoring to the carrier, the gas migration technique entails the introduction of corrosive or reducing gases, such as ammonia, phosphine, and hydrogen chloride. Using a combination of NC, Fe NPs, and NH4Cl as precursors for pyrolysis, Li et al. [68] found that the resulting FeCl2 could be readily vapor-diffused into the carriers at 500 °C, forming Fe monoatoms (Fig. 6E). In a similar manner, Yu et al. [69] produced SAFe-SWCNT thin films by loading Fe NPs into single-walled carbon nanotubes (SWCNT) and then annealing the SWCNT during the process of XeF2 introduction (Fig. 6F).

Methods for dispersing metal atoms

Single-atom metalloids can be obtained directly from precursors, while achieving a uniform dispersion of individual atoms and stopping them from migrating and aggregating to form NPs are the main goals of the synthesis of SAzymes [70]. UP to date, approaches based on enhanced metal-carrier interactions to capture isolated metal atoms on carriers mainly include spatial confinement, defect anchoring strategy and coordination stabilization. In addition, the research frequently uses chemical etching, gas migration, direct pyrolysis, and the electrochemical deposition approach.

Spatial confinement

The essence of spatial confinement lies in the encapsulation and separation of the metal precursor to immobilize the active center of the metal atom. Individual Pt atoms were carefully encapsulated in the six-membered ring of a Sodalite (SOD) cage within a Y molecular sieve by Chen et al. [71] using a template-guided approach. (Fig. 7A). In order to create single-atom Mo SAzymes with varying coordination numbers, Wang et al. [72] enclosed MoO2(acac)2 molecules in ZIF-8 pores. Following pyrolysis, the Mn2+ absorbed by the hollow structure was transformed into single-atom Mn sites in the SAzymes. The molybdenum single-atom nanosize enzyme (MoSA-Nx-C) was established and verified for its peroxidase-like specificity (Fig. 7B). By coordinating monoatomic manganese and nitrogen atoms in a hollow zeolite imidazolite skeleton, Yang et al. [73] created a polyethylene glycolated manganese-based SAE (Mn/PSAE). Mn/PSAE demonstrated notable therapeutic effects by producing a range of ROS and photothermal activities through stimulation of the tumor microenvironment (Fig. 7C).

Fig. 7
figure 7

Copyright 2022. Wiley–VCH. B Schematic illustration for the fabrication strategy of MoSA-Nx-C catalysts. Reproduced with permission [72]. Copyright 2021. Elsevier. C Coordination of monoatomic manganese and nitrogen atoms in a hollow zeolitic imidazolite skeleton to construct Mn/PSAE. Reproduced with permission [73]. Copyright 2021. Wiley–VCH. D Schematic diagram for the synthesis of Ni5P4-Ru. Reproduced with permission [74]. Copyright 2020. Wiley–VCH. E Schematic illustration of synthesis process for FeN5 SAzyme. Reproduced with permission [75]. Copyright 2022. Wiley–VCH

Methods for dispersing metal atoms: spatial confinement, defect anchoring strategy and coordination stabilization. A Illustration of the synthesis strategy to selectively encage single Pt atoms into the six-membered rings of SOD cages within Y zeolite. Reproduced with permission [71].

Defect anchoring strategy

Immobilizing metal atoms can also be accomplished effectively by carrier defects. By using crystal surface defects of CeO2 clusters to trap Pt atoms, Yan et al. [54] successfully synthesized single-atom Pt/CeO2 clusters for the treatment of traumatic brain damage. He et al. [74] constructed Ni(OH)2-rich nickel vacancies with RuCl3 and obtained the target Ni5P4-Ru SAC by subsequent phosphorylation treatment, where strong interactions between the nickel vacancy defects and Ru cations enhanced the Ru doping rate (Fig. 7D). Defects on the carbon carriers that served as anchor sites for the insertion of Fe atoms were created by Xu and associates [75]. The Fe atoms inserted into the flaws were immobilized by the N species in the NC substrate, and the FeN5 SAzymes exhibited superior peroxidase-like activity in the tumor microenvironment. ZIF-8 precursors covered with SiO2 that were pyrolyzed to cause Zn volatilization (Fig. 7E).

Coordination stabilization

With regard to the ligand stabilization doctrine, it has been shown that metal-nonmetal bonds can significantly enhance metal-carrier interactions [70]. In order to improve the metal-carrier interactions and the charge redistribution of the surrounding atoms, Au atoms were added to NiFe layered double hydroxides by Zhang et al. [76], where Au atoms on O atoms produced the hydroxides. As a result, catalytic performance was enhanced. This led to improved catalytic performance. Ag atoms were uniformly distributed on TiO2 carriers by Wang et al. [77], who then created Ag SAzymes with potent antiviral properties by means of strong Ag–O metal-carrier interactions. In order to facilitate the effective oxidative breakdown of propranolol in water, single Fe atoms were trapped in two-dimensional MoS nanosheets (Fe MoS) by Huang et al. [78] as extremely reactive catalysts for the non-homogeneous activation of sulfites.

The spatial confinement approach encapsulates fleeing metal atoms primarily through metal-carrier interactions. The stability of SAzymes is mostly caused by the interaction between the metal and the carrier [79]. However, the matrix framework in the wet-chemical synthesis method hides some of the metal active centers, resulting in a reduced usage of monoatomic metals. Similar to how the gas migration strategy chooses certain gases with corrosive or reducing properties to make the metal nanoparticles more volatile and increase their anchoring efficiency, the direct pyrolysis strategy uses high temperature to forcibly break strong metal–metal (M-M) bonds in the metal nanoparticles in the metal nanozymes.

Bioenzymatic-like catalytic activity of SAzymes

Artificial enzymes of different types and structures have different catalytic mechanisms, which lead to different final catalytic results (Fig. 8). This section will discuss several oxidoreductases mimicked by SAzymes that have been identified in recent studies, including POD, OXD, CAT, SOD, and GSH-Px (Table 1).

Fig. 8
figure 8

Schematic diagrams of the main enzyme-like catalytic activities of various nanozymes. POD-like activity of the nanozymes catalyzes peroxidation reaction using H2O2 as substrate. OXD-like activity nanozymes catalyze the generation of H2O2 in the presence of O2. POD-like activity and OXD-like activity nanozymes usually promote the production of ROS in organisms, resulting in a bactericidal effect. CAT-like activity helps to catalyze the decomposition of H2O2 into O2 and H2O. SOD-like activity mainly catalyzes the disproportionation of superoxide anion radicals (O2·-) into H2O2 and O2.POD-like activity and OXD-like activity nanozymes mainly scavenge excess ROS in the body and mitigate oxidative stress damage. OXD oxidase, POD peroxidase, CAT catalase, SOD superoxide dismutase, ROS reactive oxygen species

Table 1 Summary of various bioenzymatic-like catalytic activity of SAzymes

POD-mimetic activity

With H2O2 serving as the electron acceptor, the POD-like activity of SAzymes catalyzes the synthesis of -OH, which severely damages cancer cells through oxidative stress by producing copious amounts of reactive oxygen species. POD-like SAzymes are widely distributed, and their nanomaterials contain a wide variety of transition metals, primarily Fe, Cu, Au, Pt, Co, and Ce [80]. Research has demonstrated that Fe-N-C single-atom nano-enzymes (Fe-N-C SAzymes), which are prepared by high-temperature calcination, have peroxidase-like activity [81]. More precisely, their enzymatic activity is less than that of peroxidase-like activity and is more akin to that of natural metalloproteinases. Fe-N-C SAzymes can detect H2O2 colorimetrically and have a better catalytic efficiency, which makes them more useful in biology. For example, Fe-N-C single atom nanozymes have remarkable peroxidase mimic activity. Butyrylcholinesterase (BChE) activity can be highly sensibly biosensitized with these SAzymes. POD activity is also shown by other SAzymes that include Fe single atoms as their active centers [82]. New SAzymes based on carbon nanotubes loaded with Fe single atoms (CNT/FeNC) were used to detect H2O2, ascorbic acid, and glucose. These enzymes also showed good peroxidase-like activity [83]. According to the previously cited Wu et al. [84], Cu-N-C was produced on carbon nanosheets with a high concentration of Cu sites by replacing Fe in SAzymes with Cu, and it was shown that the peroxidase activity remained present. Furthermore, due of the dense dispersion of active copper atoms (≈5.1 wt%), the Cu-N-C SAzymes showed remarkable activity in mimicking natural peroxidase. Excellent POD activity was also shown by a series of single-atom nano-enzymatic liquids with different metal active centers. Furthermore, research on molybdenum single-atom nanozymes (MoSA-Nx-C) showed that the quantity of ligands at a particular molybdenum site controlled the specificity of the enzyme similar to peroxidase, offering a useful approach for the logical construction of the targeted nanozymes [72]. Carbon nanomaterials derived from zinc-based zeolite imidazolate frameworks (ZIF-8), which have zinc atoms dispersed atomically, can be used to make efficient single-atom peroxidase mimics [85].

OXD-mimetic activity

Using O2 as the reaction substrate, OXD-like activities are a class of redox reactions that can imitate amino acid oxidase (AAO), uric acid oxidase (UOX), and glucose oxidase (GOX), depending on the kind of hydrogen donor [80]. Whereas SAzymes with limited reducing activity convert O2 to H2O, those with significant reducing characteristics convert O2 straight to H2O2. To examine the structure–activity link, Xu et al. [86] looked at Fe-N-C single-atom catalysts with two distinct coordination structures (NG-Heme and G-Heme). In particular, the contact between the active site and the intermediate can eventually boost its intrinsic oxidase activity. It was discovered that NG-Heme exhibits stronger oxidase activity than G-Heme. It has a high sensitivity for colorimetric carcinoembryonic antigen detection in clinical settings. Other research had looked into the performance of Fe-N-C SAzymes as oxidase-like SAzymes. These enzymes have atomically dispersed metal active sites that can be activated to produce reactive oxygen species [87]. Ru single atom loading based on carbon dots has produced SAzymes with strong stability, outstanding biocompatibility, and great activity [88]. Ru SAzymes can function as surrogates for glutathione oxidases, peroxidases, and other oxidative enzymes. ROS formation and glutathione depletion can be catalyzed simultaneously by them, which can exacerbate ROS-induced cell damage and ultimately lead to cancer cell death—a crucial stage in tumor treatment. Zhu et al. [73] reported that a manganese-based PEGylated SAN (Mn/PSAE) could efficiently produce large amounts of harmful ROS and mimic the different catalytic capabilities of OXD, POD, and CAT enzymes in addition to having better photothermal properties. FeN5, when contained within a carbon nanoframework (FeN5SA/CNF), possesses a well-defined active part and important synergistic effects among its single-atom nano-enzymatic active centers, which confer outstanding oxidase-like activity and multifunctional antimicrobial activities [64]. This implies that the axial N coordination in FeN5SA/CNF has an oxidase-like driving effect, increasing its catalytic activity relative to other SAzymes.

CAT-mimetic activity

An important antioxidant enzyme in the metabolism of ROS and H2O2 is CAT. Cancer cells suffer significant oxidative damage when there is an abundance of ROS. By breaking down endogenous H2O2 into O2, the CAT-like activity of SAzymes helps to improve the hypoxic environment around tumors in cancer cells. CAT-like nanozymes mimic natural CAT by generating H2O and O2 from H2O2 as substrates, providing electrons from the catalytic activity center to align and disproportionate H2O2. With atomically dispersed active centers, Cao et al. [89] constructed Co/PMCS as an enzyme-mimicking single-atom catalyst. It atomically dispersed active centers, which can eliminate H2O2 by mimicking superoxide dismutase, catalase, and glutathione peroxidase, and its elimination efficiency is significantly higher than that of other nanozymes. With single-atom ruthenium serving as an active catalytic site anchored in the metal–organic framework Mn3[Co(CN)6]2, the self-assembled photodynamic therapy nanoreagent OxgeMCC-r SAzymes produces OxgeMCC-r with a high loading capacity and CAT-like activity. Endogenous H2O2 reacts with single-atom Ru in OxgeMCC-r to generate O2 in situ, reducing hypoxic conditions in the tumor microenvironment [28]. The dual catalytic activity of single-atom iron-dispersed N-doped mesoporous carbon nanospheres (SAFe-NMCNs) nanozymes imitate both oxidase and peroxidase [90]. Experiments conducted in vitro and in vivo shown that the SAFe-NMCNs nanozymes could effectively limit the proliferation of tumor cells and had a synergistic therapeutic effect when combined with photothermal-enhanced nanocatalytic therapy. It is possible to mimic the actions of catalase (CAT) and SOD by anchoring atomically dispersed Fe-N4 sites on N-doped porous carbon materials (Fe-SAs/NC) [91]. Because of this, Fe-SAs/NC can work as a dual-purpose single-atom-based enzyme (SAzyme) that scavenges reactive oxygen species (ROS) and gets rid of extra ROS that are created when cells are under oxidative stress.

SOD-mimetic activity

The transition metal elements Cu, Zn, Mn, and Fe are the key components of antioxidant metalloenzymes, which are found in organisms and regulate ROS levels in cells. These enzymes have SOD-like activity. Consequently, Cu, Mn, and Au are also the primary components of SOD-like activity in nanozymes including transition metal elements [80]. Because of their special qualities, carbon-based SOD mimetic nanozymes have lately been employed as promising antioxidant nanotherapeutics. The majority of reactive oxygen species (ROS) in the body are produced by superoxide anions, which can be scavenged by SAzymes having SOD-like activity. Yan et al. [54] developed a single-atom Pt/CeO with long-lasting catalytic activity, and the single-atom Pt greatly increased the endogenous CAT activity, resulting in greatly increased scavenging activity and enzyme-like activity of RONS of the final SAzymes compared to other materials. Co/PMCS was found to have atomically dispersed ligand-unsaturated active centers and hence also possesses SOD-like, CAT- and glutathione peroxidase activities [89]. In the end, this lowers pro-inflammatory cytokine levels and shields organs from harm. Due to its wealth of electronic energy levels and surplus of transition metal electronic states, Au offers a strong foundation for the development of atomic-level enzymes. It was demonstrated that the activities of Au25, Au24Cu1, and Au24Cd1 are similar to those of GSH-Px, CAT, and SOD, respectively [92]. Au24Cu1 reduces peroxides in the damaging brain, whereas Au24Cd1 prioritizes the use of superoxides and significantly reduces inflammatory factors. In order to enhance the suppression of tumor growth, Lu et al. [93] created an N-doped carbon sphere doped with monoatomic copper, called Cu-HNCS, whose SAzymes may directly catalyze the breakdown of oxygen and hydrogen peroxide into ROS. And more research revealed that monoatomic copper was the primary source of the strong catalytic activity of Cu-HNCS.

GSH-Px-mimetic activity

The primary mechanism of catalysis in single-atom nanozymes exhibiting glutathione peroxidase activity involves the depletion of intracellular glutathione superoxide (GSH), resulting in the elimination of microbes and cancerous cells. Cu single-atom sites/N-doped porous carbon (Cu SASs/NPC) can function as GSH-Px-like nanozymes, which can deplete GSH in bacteria and hence greatly enhance the bactericidal effect. Cu SASs/NPC have a greater glutathione (GSH) depletion capacity than non-Cu-doped NPC [94]. In addition, copper SAzymes based on a bionic single-atom nano-enzyme system have been prepared in other studies and validated to have excellent POD-like activity [95]. It demonstrated effective tumor targeting both in vivo and in vitro, preventing the ability of cells to synthesize GSH from the source. An adaptive iron mutation platform developed by Cao et al. [96] builds upon single-atom nanozymes (SAzymes). By enabling SAzymes to deplete GSH in tumor cells on demand, the platform speeds up safe and selective iron apoptosis. Studies on breast and colon cancers have shown evidence of this adaptive anti-tumor response.

Biological applications of SAzymes in gastrointestinal (GI) diseases

While nanozymes have catalytic activity similar to that of genuine enzymes, they suffer from a number of serious problems, such as insufficient substrate selectivity, confusing architectures, and imprecise catalytic processes [97]. SAzymes have the advantages of facile separation, homogenous active sites, customizable coordination environments, and high atom utilization because they allow for the rational design of the coordination environment and the selection of metal atoms and their valence states. In particular, nitrogen-doped carbon-loaded SAzymes have structurally similar metal-Nx(M-Nx) sites to those of natural enzymes, and thus are considered to have a wide range of prospects for mimicking natural enzyme activities [85]. This section summarizes the biological application areas of nanozymes, primarily based on recent basic research, as illustrated in the image. These fields include bioassays, antibacterial, anticancer therapy, and anti-inflammatory effects (Fig. 9).

Fig. 9
figure 9

Various biological applications of nanozymes in gastrointestinal diseases

Antibacterial application

ROS and reactive oxygen species (RNS) are the most common free radicals, and excessive accumulation of free radicals can help nanoparticles to fight against microorganisms or to reduce microbial resistance [98]. SAzymes can undergo a Fenton-like reaction, which is utilized to kill bacteria by the ROS produced by the reaction, and have demonstrated excellent bactericidal effects in GI diseases (Table 2).

Table 2 Biological applications of SAzymes in Gastrointestinal (GI) Diseases

Due to their broad-spectrum and drug-free antibacterial capabilities, single-atom nanozymes with antimicrobial activity have been increasingly significant in antimicrobial therapy in recent years [99]. Escherichia coli, Staphylococcus aureus, and some multidrug-resistant bacteria (MDR) are also common pathogens in GI disorders. The zinc-based ZIF-8 with atomically dispersed zinc atoms was reported by Xu et al. [100] and its derived carbon nanomaterials can be used as effective single-atom peroxidase mimics. Lastly, it was confirmed that SAzyme with unsaturated Zn-N4 sites is a powerful antimicrobial agent for wound treatment. It has strong antibacterial action against Pseudomonas aeruginosa as well as outstanding peroxidase-like activity. They conducted an analysis based on the particular mechanism by which Ag single atoms with high electrical conductivity could stimulate the MnO2 oxygen vacancies, hence facilitating the entry of reactive compounds that are photocatalytic. In addition, the photothermal conversion efficiency is enhanced by the catalysis of single-atom Ag, leading to the enhancement of the redox properties of the crystalline materials. Additionally, it has been investigated that Cu single-atom site/N-doped porous carbon (Cu SASs/NPC) was discovered to exhibit POD-like activity after being effectively synthesized using a sequence of pyrolysis-adsorption procedures [56]. The POD activity was greatly increased by doping the material with a single atom of Cu. Additionally, photo-thermal characteristics of the materials were specifically and simultaneously optimized to speed up the consumption of GSH. This synergistic effect enabled Cu SASs/NPC to exhibit excellent antimicrobial properties against Escherichia coli. and methicillin-resistant S. aureus (MRSA). The effective in vitro antibacterial and in vivo anti-infective qualities of single iron atom nanocatalysts have also been emphasized in a number of research conducted in recent years. Huo et al. [101] created nitrogen-doped amorphous carbon (SAF NCs) nanocatalysts attached with single iron atoms to cause peroxidase-like activity in the presence of H2O2, which efficiently eliminated Staphylococcus aureus and Escherichia coli. In order to eradicate intracellular MRSA, Liu et al. [102] created the highly sought-after missile-like nanotherapeutic medication FeSAs@Sa.M. and produced extremely toxic ROS through enzymatic activity at the center of FeSAs. A multifunctional Cu single-atom nanozyme (l-Arg@Cu-SAzymes) loaded with l-arginine was created by Qiu et al. [103]. This strategy was thought to be a promising way to treat MDR infection because it synergized with ROS and RNS to give the therapeutic system a strong antimicrobial efficacy and an enhanced tissue remodeling ability. In addition, antimicrobial activities against Escherichia coli [64]. and MRSA [89] have been demonstrated in ongoing experiments.

Despite the fact that ROS typically cause great harm to bacterial cells by damaging their cell membranes, as demonstrated by the aforementioned research, single-atom nanozymes are crucial for antibacterial applications primarily because of their biomimetic properties (particularly their POD-like activity).

Antitumor therapy

One of the primary mechanisms for inducing apoptosis in tumor cells is the generation of ROS. In the biological arena, particularly in the area of anti-tumor therapy, they have achieved remarkable strides due to the multiple mimetic enzyme activities of SAzymes and the simultaneous creation of multiple ROS [104].

Wang et al. [105] designed a protein-supported single-atom copper nanozyme (BSA-Cu SAzymes), which possesses ROS-generating and GSH-depleting effects, to effectively restore the elevated autophagy level of F. nucleatum and the ROS resistance of the tumor cells in situ to synergistically killing colorectal cancer (CRC) cells (Fig. 10). Furthermore, BSA-Cu SAzymes have a good biosafety profile, making them a promising new treatment for colorectal cancer as they can be processed by the kidneys. Also relevant to the tumor microenvironment, probiotics may enable cancer biotherapy by secreting antitumor or immunomodulatory drugs in the tumor microenvironment. However, the efficacy and accuracy of probiotics in cancer treatment is limited, and the addition of nanozymes can ameliorate this shortcoming. Furthermore, studies on colon cancer both in vivo and in vitro have shown that the composite nanozyme system can efficiently suppress tumor growth via photothermally enhanced nano-catalytic synergistic therapies, offering a novel way to boost the effectiveness of chemodynamic therapy (CDT) [106, 107].

Fig. 10
figure 10

Formation and characterization of BSA-Cu SAN. a Schematic illustration of the synthesis of BSA-Cu SAN and its function of destroying pathogen-tumor symbionts for antitumor therapy, b Negative-stain electron microscopy image of BSA-Cu SAN. White bots represent BSA-Cu SAN, some of which are marked by yellow arrows. c Atomic HAADF image of BSA-Cu SAN with SAED pattern inset in the bottom left corner and partial single Cu atoms highlighted in yellow dash circles. d The intensity spectrum image along the distance in the yellow dash tangle in (c). e Pseudo-color image of corresponding intensity of (c). f The enlarged image of (e). g Elemental mapping of BSA-Cu SAN. h Cu 2p XPS spectra of BSA-Cu SAN. i Binding site of Cu+ on BSA based on the highest docking score. j Protein-ligand interaction diagram for Cu+. Reproduced with permission [105]. Copyright 2023, Springer Nature

Liu et al.’s study [108] produced novel and effective Ir-N5 single-atom nanozymes for the treatment of liver tumors. These SAzymes mimic the enzyme cascade and disrupt the redox and metabolic homeostasis of the tumor region, producing an anticancer effect at the tumor site and amplifying oxidative stress by increasing ROS, which kills tumor cells with less impact on normal cells, resulting in an effective cancer therapy. One of the worst tumors is pancreatic cancer (PDAC), and using nanomaterials offers a fresh strategy to treat drug-resistant pancreatic. Currently, more experiments in the field of pancreatic cancer are carried out through the synergistic action of drugs or organic molecules with nanozymes to target delivery to the tumor site and reduce the drug resistance of cancer cells. Zhao et al. [109] synergized a metal-based SAzymes coupling (Fe@Fe3O4) with a naturally occurring biologically active organic molecule (ginseng saponin RG3) to construct a novel nanomedicine, which releases Fe3+ in the tumor TME and Fe2+ in the tumor TME and efficiently generates ROS thereby promoting cancer cell apoptosis. This also provides a new strategy for metal–organic nanocomposites to play a role in anticancer therapy.

Based on the cytochrome P450 structure, Sun et al. [110] created sandwich-like Sazymes and discovered that IrN4-S-TMN4 (TM = Co, Rh, Pd) showed selective CO2 reduction to CO with a special artificial CO dehydrogenase (CODH). They hypothesize that the unique enzyme-like activity and selectivity of SANs result from the precise control of sulfur atoms over the electron density, and that this control is contingent upon the nature and characteristics of the transition metals present in the surface layer. Feng et al. [111] immobilized ultramicro gold nanozymes in a metal–organic skeleton to establish a gold-iron SAzyme, which showed promising anti-esophageal cancer effects in vivo via the ferroptosis pathway. The same application of the ferroptosis pathway was also validated among the palladium nanozymes constructed by Zhang et al. [112]. Furthermore, an adaptive SAzymes-based iron death platform was constructed. This platform not only specifically enhances ROS generation activity, but also empowers SAzymes to consume GSH on demand in tumor cells, thereby accelerating selective and safe ferroptosis. The above anti-tumor responses have been demonstrated in colon cancers [96].

The limitations of traditional tumor treatment techniques have spawned the development of novel nanotechnology for the treatment of tumors. Currently, the application of nanozymes in gastrointestinal tumors is mainly limited to colorectal, liver and pancreatic cancers, and more studies are needed to explore their biological applications in other solid tumors (Table 2).

Antioxidative therapy

Through chemical redox processes, antioxidant nanostructures can neutralize ROS. Numerous studies have suggested that by scavenging ROS and lowering the release of inflammatory factors, nanozymes can display superior anti-inflammatory and antioxidant benefits in the treatment of ulcerative colitis and inflammatory bowel disease (IBD) alone [9, 113]. Nevertheless, the contribution of biological SAzymes structures, which resemble natural enzymes in their activity, to the elimination of ROS remains incompletely understood.

ROS are scavenged by atomically dispersed Fe-N4 sites anchored on N-doped porous carbon materials (Fe-SAs/NC), which act as a bifunctional SAzyme for the removal of excess ROS produced during cellular oxidative stress. These sites mimic two antioxidant enzymes, CAT-like and SOD-like [91]. Comparable Fe-N/C monoatomic catalysts also exhibit glutathione-like, catalase-like, oxidase-like, and peroxidase-like activity. Intracellular H2O2 levels were successfully regulated by Fe-N/C SAzymes [114]. The findings of the two studies mentioned above demonstrated that the synthesized Fe-N/C SACs and Fe-SAs/NC were both more effective at scavenging the excess ROS that the oxidatively stressed cells produced. This suggests that the SAzymes could effectively shield the cells from the harm caused by cellular oxidative stress by scavenging the intracellular ROS, opening the door to the treatment of diseases linked to ROS. Not coincidentally, single-atom Pt/CeO2 treats brain injury by scavenging RONS by a mechanism that exhibits POD-like, CAT-like, and OXD-like multienzymatic catalytic activities [54]. Similar phenomena were found in an in vitro E. coli mouse model, which demonstrated the antioxidant activity of Co-SAzymes [89]. The CAT-like, SOD-like, and GSH-Px-like activities of Co-SAzymes reduced ROS and RNS in sepsis by 60–80%, and pro-inflammatory cytokine levels were reduced to 10% after 2 weeks of treatment. Because of its unique activity and kinetic features that are naturally similar to those of APX, graphitized carbon nitride (Cu SAs/CN) anchored with isolated single copper atoms can be employed to successfully protect H2O2-treated cells from oxidative damage in vitro [115]. Liu et al. [116] synthesized Pt@PCN222-Mn on the basis of manganese (III) porphyrin, which was doped with Mn-MOF to mimic superoxide dismutase and convert oxygen radicals into hydrogen peroxide. The experiments also mimicked the catalytic activity of catalase by doping platinum nanoparticles. Both in vitro and in vivo experimental measurements demonstrated the synergistic ROS scavenging ability of this integrated cascade of nanozymes.

Recent research has demonstrated that single-atom nanozymes, with their huge specific surface area, unusual electron packing, and superior electrical conductivity, possess intrinsic antioxidant characteristics. The application of nanozymes is a viable strategy to remove the excess intracellular ROS and to maintain the homeostasis of the cellular redox system because, despite the presence of several antioxidant natural enzymes such as CAT, SOD, and GSH-Px in the cellular system, the overexpression of ROS induced by pathological conditions inhibits the activity of the natural enzymes [117, 118] (Table 2).

Biological detection

Excess GSH is a cancer biomarker that is crucial for preserving intracellular redox balance in tumor cells. Hemoglobin (Hb) as a Fe source embedded in zeolite imidazolate framework (ZIF-8) was utilized by Chen et al. [119] to create porous single-atom Fe nanozymes (pFe SAzymes) in a convenient and large-scale method, and then developed a pFe SAzymes-GSH assay, which can accurately detect GSH levels in tumor cells transplanted from liver in situ. The pFe SAzymes-GSH assay can detect GSH levels in liver in situ transplanted tumor cells with millimolar accuracy and avoids the need for complex manipulation, making it a simple, fast and accurate visualization method for identifying tumor boundaries. Catalytic nanomaterials called iron SAzymes (Fe-N-C SAzymes) were also made, and it was discovered that adenine and thymine exhibited a greater adsorption affinity on Fe-N-C SAzymes. Sun et al. [120] designed Apt/Fe-N-C SAzymes for the colorimetric assay of cancer cells based on the observation that one DNA sequence (adenine) in duplex DNA binds to Fe-N-C SAzymes and the other DNA sequence (i.e., aptamer) binds to cancer cells. This discovery offers a novel application of SAzymes in biomedicine. Nanozymes have also been used to distinguish between normal and cancerous cells for early identification of cancer. SAzymes has been utilized in investigations for the detection of various blood indicators in addition to tumor-related diagnoses (Table 2). In order to enable practical uric acid (UA) monitoring in serum samples, Hu et al. [121] investigated an A-Co-NG single-atom catalyst for electrochemical UA detection for the first time. They did this by attaching high-density and isolated cobalt atoms on an N-doped graphene substrate. Fe-SAzyme, which was produced by Zhou et al. [122], has a built-in colorimetric assay for galactose measurement and can be utilized as a substitute approach for diagnosing galactosemia. A flow-injection chemiluminescence immunoassay was created for the quick and accurate detection of serum 5-fluorouracil (5-Fu) in serum [123] based on the validation of the Fenton-like activity of Co-SAzymes.

Discussion and perspectives

Among the deadliest cancers, gastrointestinal cancers cause around one-third of cancer-related deaths globally [124]. A number of gastrointestinal and liver conditions, such as inflammatory bowel disease (IBD) [125], colorectal cancer (CRC) [126], and alcohol-associated liver disease [127], have been related to alterations in the human gut microbiota. When inflammation reaches advanced stages, it can result in multiple organ dysfunction syndrome (MODS), infectious necrosis, and systemic inflammatory response syndrome (SIRS). One such condition is pancreatitis, which has a high morbidity and mortality rate and can be fatal [128]. Even though the digestive system is now developing quickly, new technologies are still required to solve current issues due to the diversity and complexity of disorders of the digestive tract, which places more demands on early diagnosis and accurate treatment.

Environmental protection, antibacterial, anticancer, and sensing are just a few of the many applications for SAzymes. Rethinking the connection between the structure of SAzymes and their active roles has proven difficult in light of the substantial changes that have occurred recently in the content, structure, and morphology of these molecules. Enzymes possessing atomically distributed metal active sites, as well as the ability to bind any ligand on appropriate carriers, exhibit significant promise for medicinal uses. This work establishes the groundwork for a later discussion of the function of SAzymes in gastrointestinal disorders by reviewing the most recent design principles and the preparation procedure of SAzymes, as well as by classifying and elaborating the biomimetic activities.

In addition, the complex internal environment and immune microenvironment in the body may affect the efficacy of SAzymes. The future research of SAzymes strategy is suggested by the significant number of vacancies in their study of inflammatory digestive system disorders such pancreatitis and cholecystitis. Currently, clinical investigations of SAzymes are not met, mostly due to the uncertainty over their biosafety. The ability of single-atom nanozymes to withstand degradation is one of the more significant variables. Despite numerous animal investigations have demonstrated that many SAzymes do not have harmful effects on other organs, the risks associated with a continuous presence of these enzymes in the bloodstream remain unclear, particularly with regard to critical organs like the heart and brain [129]. Furthermore, since different SAzymes have different active centers and framework structures, toxicity occurs when their structural composition, elemental loading composition, internal environment, and mode of administration are changed. Future research should focus on improving the in vivo biostability of single-atom nanozymes and clarifying the scope of their uses in microbial bioassay, targeted drug delivery, immune regulation and anti-inflammatory therapy [130, 131]. A thorough safety evaluation that takes into account of pharmacokinetics, body organ distribution, and in vivo metabolism is still necessary for the clinical use of SAzymes.

Availability of data and materials

Not applicable.

References

  1. Meyers WC. A study of gastric mucosa in various diseases affecting the upper part of the gastrointestinal tract. Gastroenterology. 1948;10:923–38.

    CAS  PubMed  Google Scholar 

  2. Hanauer SB. The burdens of digestive diseases. Nat Rev Gastroenterol Hepatol. 2009;6:377.

    Article  PubMed  Google Scholar 

  3. Islami F, Miller KD, Siegel RL, Fedewa SA, Ward EM, Jemal A. Disparities in liver cancer occurrence in the United States by race/ethnicity and state. CA Cancer J Clin. 2017;67:273–89.

    Article  PubMed  Google Scholar 

  4. Ap K. Pancreatic cancer epidemiology: understanding the role of lifestyle and inherited risk factors. Nat Rev Gastroenterol Hepatol. 2021. https://doi.org/10.1038/s41575-021-00457-x.

    Article  Google Scholar 

  5. Dekker E, Tanis P, Vleugels J, Kasi P, Wallace M. Colorectal cancer. Lancet. 2019. https://doi.org/10.1016/S0140-6736(19)32319-0.

    Article  PubMed  Google Scholar 

  6. Hindson J. Digestive disease week 2023. Nat Rev Gastroenterol Hepatol. 2023;20:483.

    Article  PubMed  Google Scholar 

  7. Wadhwa V, Patel P, Grover D, Ali F, Thosani N. Interventional gastroenterology in oncology. CA: A Cancer J Clin. 2023. https://doi.org/10.3322/caac.21766.

    Article  Google Scholar 

  8. Zhong J, Yang X, Gao S, Luo J, Xiang J, Li G, Liang Y, Tang L, Qian C, Zhou J, Zheng L, Zhang K, Zhao J. Geometric and electronic structure-matched superoxide dismutase-like and catalase-like sequential single-atom nanozymes for osteoarthritis recession. Adv Funct Mater. 2023;33:2209399.

    Article  CAS  Google Scholar 

  9. Cao Y, Cheng K, Yang M, Deng Z, Ma Y, Yan X, Zhang Y, Jia Z, Wang J, Tu K, Liang J, Zhang M. Orally administration of cerium oxide nanozyme for computed tomography imaging and anti-inflammatory/anti-fibrotic therapy of inflammatory bowel disease. J Nanobiotechnol. 2023;21:21.

    Article  CAS  Google Scholar 

  10. Geng H, Chen J, Tu K, Tuo H, Wu Q, Guo J, Zhu Q, Zhang Z, Zhang Y, Huang D, Zhang M, Xu Q. Carbon dot nanozymes as free radicals scavengers for the management of hepatic ischemia-reperfusion injury by regulating the liver inflammatory network and inhibiting apoptosis. J Nanobiotechnol. 2023;21:500.

    Article  CAS  Google Scholar 

  11. Zhe Y, Wang J, Zhao Z, Ren G, Du J, Li K, Lin Y. Ascorbate oxidase-like nanozyme with high specificity for inhibition of cancer cell proliferation and online electrochemical DOPAC monitoring. Biosens Bioelectron. 2023;220:114893.

    Article  CAS  PubMed  Google Scholar 

  12. Li M, Chen J, Wu W, Fang Y, Dong S. Oxidase-like MOF-818 nanozyme with high specificity for catalysis of catechol oxidation. J Am Chem Soc. 2020;142:15569–74.

    Article  CAS  PubMed  Google Scholar 

  13. Cao C, Zhang T, Yang N, Niu X, Zhou Z, Wang J, Yang D, Chen P, Zhong L, Dong X, Zhao Y. POD nanozyme optimized by charge separation engineering for light/pH activated bacteria catalytic/photodynamic therapy. Signal Transduct Target Ther. 2022;7:86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Wang R, Qiu M, Zhang L, Sui M, Xiao L, Yu Q, Ye C, Chen S, Zhou X. Augmenting immunotherapy via bioinspired MOF-based ROS homeostasis disruptor with nanozyme-cascade reaction. Adv Mater. 2023;35:e2306748.

    Article  PubMed  Google Scholar 

  15. Jing H, Ren Y, Zhou Y, Xu M, Krizkova S, Heger Z, Lu Q, Wang S, Liang X, Adam V, Li N. Remodeling of the liver fibrosis microenvironment based on nilotinib-loaded multicatalytic nanozymes with boosted antifibrogenic activity. Acta Pharm Sin B. 2023;13:5030–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Liu L, Zhang Y, Li X, Deng J. Microenvironment of pancreatic inflammation: calling for nanotechnology for diagnosis and treatment. J Nanobiotechnology. 2023;21:443.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Min DK, Kim YE, Kim MK, Choi SW, Park N, Kim J. Orally administrated inflamed colon-targeted nanotherapeutics for inflammatory bowel disease treatment by oxidative stress level modulation in colitis. ACS Nano. 2023;17:24404–16.

    Article  CAS  PubMed  Google Scholar 

  18. Wang C, Lv Z, Yang W, Feng X, Wang B. A rational design of functional porous frameworks for electrocatalytic CO2 reduction reaction. Chem Soc Rev. 2023;52:1382–427.

    Article  CAS  PubMed  Google Scholar 

  19. Ding G, Zhao J, Zhou K, Zheng Q, Han S-T, Peng X, Zhou Y. Porous crystalline materials for memories and neuromorphic computing systems. Chem Soc Rev. 2023;52:7071–136.

    Article  CAS  PubMed  Google Scholar 

  20. Xu K, Zhang S, Zhuang X, Zhang G, Tang Y, Pang H. Recent progress of MOF-functionalized nanocomposites: from structure to properties. Adv Colloid Interface Sci. 2023;323:103050.

    Article  PubMed  Google Scholar 

  21. Ma K, Cheung YH, Kirlikovali KO, Xie H, Idrees KB, Wang X, Islamoglu T, Xin JH, Farha OK. Fibrous Zr-MOF nanozyme aerogels with macro-nanoporous structure for enhanced catalytic hydrolysis of organophosphate toxins. Adv Mater. 2023. https://doi.org/10.1002/adma.202300951.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Xiang K, Wu H, Liu Y, Wang S, Li X, Yang B, Zhang Y, Ma L, Lu G, He L, Ni Q, Zhang L. MOF-derived bimetallic nanozyme to catalyze ROS scavenging for protection of myocardial injury. Theranostics. 2023;13:2721–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Liang R-R, Ru-Han A, Shun-Qi X, Qi Q-Y, Zhao X. Fabricating organic nanotubes through selective disassembly of two-dimensional covalent organic frameworks. J Am Chem Soc. 2020;142:70–4.

    Article  CAS  PubMed  Google Scholar 

  24. Sun Q, Fu CW, Aguila B, Perman J, Wang S, Huang H-Y, Xiao F-S, Ma S. Pore environment control and enhanced performance of enzymes infiltrated in covalent organic frameworks. J Am Chem Soc. 2018;140:984–92.

    Article  CAS  PubMed  Google Scholar 

  25. An S, Xu Q, Ni Z, Hu J, Peng C, Zhai L, Guo Y, Liu H. Construction of covalent organic frameworks with crown ether struts. Angew Chem Int Ed Engl. 2021;60:9959–63.

    Article  CAS  PubMed  Google Scholar 

  26. Yao S, Zhao X, Wang X, Huang T, Ding Y, Zhang J, Zhang Z, Wang ZL, Li L. Bioinspired electron polarization of nanozymes with a human self-generated electric field for cancer catalytic therapy. Adv Mater. 2022;34:e2109568.

    Article  PubMed  Google Scholar 

  27. Zhang Z, Ye Y, Xiang S, Chen B. Exploring multifunctional hydrogen-bonded organic framework materials. Acc Chem Res. 2022;55:3752–66.

    Article  CAS  PubMed  Google Scholar 

  28. Wang D, Wu H, Phua SZF, Yang G, Qi Lim W, Gu L, Qian C, Wang H, Guo Z, Chen H, Zhao Y. Self-assembled single-atom nanozyme for enhanced photodynamic therapy treatment of tumor. Nat Commun. 2020;11:357.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Liang J, Johannessen B, Wu Z, Webster RF, Yong J, Zulkifli MYB, Harbort JS, Cheok YR, Wen H, Ao Z, Kong B, Chang SLY, Scott J, Liang K. Regulating the coordination environment of mesopore-confined single atoms from metalloprotein-MOFs for highly efficient biocatalysis. Adv Mater. 2022;34:e2205674.

    Article  PubMed  Google Scholar 

  30. Li J, Gao M, Xia X, Cen Y, Wei F, Yang J, Wang L, Hu Q, Xu G. Spherical hydrogel sensor based on PB@Fe-COF@Au nanoparticles with triplet peroxidase-like activity and multiple capture sites for effective detection of organophosphorus pesticides. ACS Appl Mater Interfaces. 2023;15:6473–85.

    Article  CAS  PubMed  Google Scholar 

  31. Gao P, Wang K, Wei R, Shen X, Pan W, Li N, Tang B. A covalent organic framework-derived M1 macrophage mimic nanozyme for precise tumor-targeted imaging and NIR-II photothermal catalytic chemotherapy. Biomater Sci. 2023;11:7616–22.

    Article  CAS  PubMed  Google Scholar 

  32. Gao P, Wei R, Chen Y, Li X, Pan W, Li N, Tang B. Pt nanozyme-bridged covalent organic framework-aptamer nanoplatform for tumor targeted self-strengthening photocatalytic therapy. Biomaterials. 2023;297:122109.

    Article  CAS  PubMed  Google Scholar 

  33. Zhang L, Zhang W, Nie Y, Wang Y, Zhang P. Covalent organic framework-supported ultrasmall Rh nanoparticles as peroxidase mimics for colorimetric sensing of cysteine. J Colloid Interface Sci. 2023;636:568–76.

    Article  CAS  PubMed  Google Scholar 

  34. Zhang L, Liu Z, Deng Q, Sang Y, Dong K, Ren J, Qu X. Nature-inspired construction of MOF@COF nanozyme with active sites in tailored microenvironment and pseudopodia-like surface for enhanced bacterial inhibition. Angew Chem Int Ed Engl. 2021;60:3469–74.

    Article  CAS  PubMed  Google Scholar 

  35. Liu B, Pan X, Nie D, Hu X, Liu E, Liu T. Ionic hydrogen-bonded organic frameworks for ion-responsive antimicrobial membranes. Adv Mater. 2020;32:e2005912.

    Article  PubMed  Google Scholar 

  36. Tong L, Lin Y, Kou X, Shen Y, Shen Y, Huang S, Zhu F, Chen G, Ouyang G. Pore-environment-dependent photoresponsive oxidase-like activity in hydrogen-bonded organic frameworks. Angew Chem Int Ed Engl. 2023;62:e202218661.

    Article  CAS  PubMed  Google Scholar 

  37. Yin N, Wang Y, Liu Y, Niu R, Zhang S, Cao Y, Lv Z, Song S, Liu X, Zhang H. A cholesterol metabolic regulated hydrogen-bonded organic framework (HOF)-based biotuner for antibody non-dependent immunotherapy tailored for glioblastoma. Adv Mater. 2023;35:e2303567.

    Article  PubMed  Google Scholar 

  38. Liu B, Pan X, Zhang D, Wang R, Chen J, Fang H, Liu T. Construction of function-oriented core-shell nanostructures in hydrogen-bonded organic frameworks for near-infrared-responsive bacterial inhibition. Angew Chem Int Ed. 2021;60:25701–7.

    Article  CAS  Google Scholar 

  39. Cai Z, Xia Y, Ito Y, Ohtani M, Sakamoto H, Ito A, Bai Y, Wang Z, Yamauchi Y, Fujita T. General synthesis of MOF nanotubes via hydrogen-bonded organic frameworks toward efficient hydrogen evolution electrocatalysts. ACS Nano. 2022;16:20851–64.

    Article  CAS  PubMed  Google Scholar 

  40. Wang C, Pan C, Wei X, Yang F, Wu W, Mao L. Emissive carbon dots derived from natural liquid fuels and its biological sensing for copper ions. Talanta. 2020;208:120375.

    Article  CAS  PubMed  Google Scholar 

  41. Kang L, Hu Y, Liu L, Wu J, Zhang S, Zhao Q, Ding F, Li Q, Zhang J. Growth of close-packed semiconducting single-walled carbon nanotube arrays using oxygen-deficient TiO2 nanoparticles as catalysts. Nano Lett. 2015;15:403–9.

    Article  CAS  PubMed  Google Scholar 

  42. Molaei MJ. Carbon quantum dots and their biomedical and therapeutic applications: a review. RSC Adv. 2019;9:6460–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Li X, Ding S, Lyu Z, Tieu P, Wang M, Feng Z, Pan X, Zhou Y, Niu X, Du D, Zhu W, Lin Y. Single-atomic iron doped carbon dots with both photoluminescence and oxidase-like activity. Small. 2022;18:e2203001.

    Article  PubMed  Google Scholar 

  44. Zhang L, Dong Q, Hao Y, Wang Z, Dong W, Liu Y, Dong Y, Wu H, Shuang S, Dong C, Chen Z, Gong X. Drug-primed self-assembly of platinum-single-atom nanozyme to regulate cellular redox homeostasis against cancer. Adv Sci. 2023;10:e2302703.

    Article  Google Scholar 

  45. Han Y, Ge K, Zhao Y, Bottini M, Fan D, Wu W, Li L, Liu F, Gao S, Liang X, Zhang J. Modulating the coordination environment of carbon-dot-supported Fe single-atom nanozymes for enhanced tumor therapy. Small. 2023. https://doi.org/10.1002/smll.202306656.

    Article  PubMed  Google Scholar 

  46. Yu H, Tang K, Cai Z, Lin X, Huang Y, Yu T, Zhang Q, Wang Q, Wu L, Yang L, Shan H, Luo H. Carbon dots-based nanozyme for drug-resistant lung cancer therapy by encapsulated doxorubicin/siRNA cocktail. Int J Nanomed. 2023;18:933–48.

    Article  CAS  Google Scholar 

  47. Ma Y, Zhang M, Wu J, Zhao Y, Du X, Huang H, Zhou Y, Liu Y, Kang Z. The key effect of carboxyl group and CuN2 O2 coordinate structure for Cu, N Co-doped carbon dots with peroxidase-like property. Small. 2023;19:2300883.

    Article  CAS  Google Scholar 

  48. Yu D, Zhang H, Ren J, Qu X. Hydrogen-bonded organic frameworks: new horizons in biomedical applications. Chem Soc Rev. 2023;52:7504–23.

    Article  CAS  PubMed  Google Scholar 

  49. Ou H, Qian Y, Yuan L, Li H, Zhang L, Chen S, Zhou M, Yang G, Wang D, Wang Y. Spatial position regulation of cu single atom site realizes efficient nanozyme photocatalytic bactericidal activity. Adv Mater. 2023;35:2305077.

    Article  CAS  Google Scholar 

  50. Ji S, Jiang B, Hao H, Chen Y, Dong J, Mao Y, Zhang Z, Gao R, Chen W, Zhang R, Liang Q, Li H, Liu S, Wang Y, Zhang Q, Gu L, Duan D, Liang M, Wang D, Yan X, Li Y. Matching the kinetics of natural enzymes with a single-atom iron nanozyme. Nat Catal. 2021;4:407–17.

    Article  CAS  Google Scholar 

  51. Wang Z, Wang W, Wang J, Wang D, Liu M, Wu Q, Hu H. Single-atom catalysts with ultrahigh catalase-like activity through electron filling and orbital energy regulation. Adv Funct Mater. 2023;33:2209560.

    Article  CAS  Google Scholar 

  52. Cao S, Zhao Z, Zheng Y, Wu Z, Ma T, Zhu B, Yang C, Xiang X, Ma L, Han X, Wang Y, Guo Q, Qiu L, Cheng C. A library of ROS-catalytic metalloenzyme mimics with atomic metal centers. Adv Mater. 2022;34:e2200255.

    Article  PubMed  Google Scholar 

  53. Fan H, Li Y, Liu J, Cai R, Gao X, Zhang H, Ji Y, Nie G, Wu X. Plasmon-enhanced oxidase-like activity and cellular effect of Pd-coated gold nanorods. ACS Appl Mater Interfaces. 2019;11:45416–26.

    Article  CAS  PubMed  Google Scholar 

  54. Yan R, Sun S, Yang J, Long W, Wang J, Mu X, Li Q, Hao W, Zhang S, Liu H, Gao Y, Ouyang L, Chen J, Liu S, Zhang X-D, Ming D. Nanozyme-based bandage with single-atom catalysis for brain trauma. ACS Nano. 2019;13:11552–60.

    Article  CAS  PubMed  Google Scholar 

  55. Zhong X, Wang X, Cheng L, Tang Y, Zhan G, Gong F, Zhang R, Hu J, Liu Z, Yang X. GSH-depleted PtCu3 nanocages for chemodynamic- enhanced sonodynamic cancer therapy. Adv Funct Mater. 2020;30:1907954.

    Article  CAS  Google Scholar 

  56. Liu L, Zhang H, Xing S, Zhang Y, Shangguan L, Wei C, Peng F, Liu X. Copper-zinc bimetallic single-atom catalysts with localized surface plasmon resonance-enhanced photothermal effect and catalytic activity for melanoma treatment and wound-healing. Adv Sci. 2023;10:2207342.

    Article  CAS  Google Scholar 

  57. Lv F, Gong Y, Cao Y, Deng Y, Liang S, Tian X, Gu H, Yin J-J. A convenient detection system consisting of efficient Au@PtRu nanozymes and alcohol oxidase for highly sensitive alcohol biosensing. Nanoscale Adv. 2020;2:1583–9.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Cai S, Fu Z, Xiao W, Xiong Y, Wang C, Yang R. Zero-dimensional/two-dimensional AuxPd100- x nanocomposites with enhanced nanozyme catalysis for sensitive glucose detection. ACS Appl Mater Interfaces. 2020;12:11616–24.

    Article  CAS  PubMed  Google Scholar 

  59. Fan Y, Liu S, Yi Y, Rong H, Zhang J. Catalytic nanomaterials toward atomic levels for biomedical applications: from metal clusters to single-atom catalysts. ACS Nano. 2021;15:2005–37.

    Article  CAS  PubMed  Google Scholar 

  60. Cheng X, Huang L, Yang X, Elzatahry AA, Alghamdi A, Deng Y. Rational design of a stable peroxidase mimic for colorimetric detection of H2O2 and glucose: a synergistic CeO2/Zeolite Y nanocomposite. J Colloid Interface Sci. 2019;535:425–35.

    Article  CAS  PubMed  Google Scholar 

  61. Tang W, Fan W, Zhang W, Yang Z, Li L, Wang Z, Chiang Y-L, Liu Y, Deng L, He L, Shen Z, Jacobson O, Aronova MA, Jin A, Xie J, Chen X. Wet/sono-chemical synthesis of enzymatic two-dimensional MnO2 nanosheets for synergistic catalysis-enhanced phototheranostics. Adv Mater. 2019;31:1900401.

    Article  Google Scholar 

  62. Sun T, Xu L, Wang D, Li Y. Metal organic frameworks derived single atom catalysts for electrocatalytic energy conversion. Nano Res. 2019;12:2067–80.

    Article  CAS  Google Scholar 

  63. Umer M, Umer S, Anand R, Mun J, Zafari M, Lee G, Kim KS. Transition metal single atom embedded GaN monolayer surface for efficient and selective CO2 electroreduction. J Mater Chem A. 2022;10:24280–9.

    Article  CAS  Google Scholar 

  64. Huang L, Chen J, Gan L, Wang J, Dong S. Single-atom nanozymes. Sci Adv. 2019;5:5490.

    Article  Google Scholar 

  65. Zhao Y, Lu XF, Fan G, Luan D, Gu X, Lou XWD. Surface-exposed single-Ni atoms with potential-driven dynamic behaviors for highly efficient electrocatalytic oxygen evolution. Angew Chem Int Ed Engl. 2022;61:e202212542.

    Article  CAS  PubMed  Google Scholar 

  66. Li X, Ye W, Xu P, Huang H, Fan J, Yuan R, Zheng M-S, Wang M-S, Dong Q. An encapsulation-based sodium storage via Zn-single-atom implanted carbon nanotubes. Adv Mater. 2022;34:e2202898.

    Article  PubMed  Google Scholar 

  67. Chen Y, Wang P, Hao H, Hong J, Li H, Ji S, Li A, Gao R, Dong J, Han X, Liang M, Wang D, Li Y. Thermal atomization of platinum nanoparticles into single atoms: an effective strategy for engineering high-performance nanozymes. J Am Chem Soc. 2021;143:18643–51.

    Article  CAS  PubMed  Google Scholar 

  68. Li Y, Wang S, Wang X-S, He Y, Wang Q, Li Y, Li M, Yang G, Yi J, Lin H, Huang D, Li L, Chen H, Ye J. Facile top-down strategy for direct metal atomization and coordination achieving a high turnover number in CO2 photoreduction. J Am Chem Soc. 2020;142:19259–67.

    Article  CAS  PubMed  Google Scholar 

  69. Meng Y, Li J, Zhao S, Shi C, Li X, Zhang L, Huo P, Liu C, Cheng H. Fluorination-assisted preparation of self-supporting single-atom Fe-N-doped single-wall carbon nanotube film as bifunctional oxygen electrode for rechargeable Zn-Air batteries. Appl Catal B. 2021;294:120239.

    Article  CAS  Google Scholar 

  70. Peng C, Pang R, Li J, Wang E. Current advances on the single-atom nanozyme and its bioapplications. Adv Mater. 2023. https://doi.org/10.1002/adma.202211724.

    Article  PubMed  Google Scholar 

  71. Chen Q, Peng P, Yang G, Li Y, Han M, Tan Y, Zhang C, Chen J, Jiang K, Liu L, Ye C, Xing E. Template-guided regioselective encaging of platinum single atoms into Y zeolite: enhanced selectivity in semihydrogenation and resistance to poisoning. Angew Chem Int Ed Engl. 2022;61:e202205978.

    Article  CAS  PubMed  Google Scholar 

  72. Wang Y, Jia G, Cui X, Zhao X, Zhang Q, Gu L, Zheng L, Li LH, Wu Q, Singh DJ, Matsumura D, Tsuji T, Cui Y, Zhao J, Zheng W. Coordination number regulation of molybdenum single-atom nanozyme peroxidase-like specificity. Chem. 2021;7:436–49.

    Article  CAS  Google Scholar 

  73. Zhu Y, Wang W, Cheng J, Qu Y, Dai Y, Liu M, Yu J, Wang C, Wang H, Wang S, Zhao C, Wu Y, Liu Y. Stimuli-responsive manganese single-atom nanozyme for tumor therapy via integrated cascade reactions. Angew Chem Int Ed Engl. 2021;60:9480–8.

    Article  CAS  PubMed  Google Scholar 

  74. He Q, Tian D, Jiang H, Cao D, Wei S, Liu D, Song P, Lin Y, Song L. Achieving efficient alkaline hydrogen evolution reaction over a Ni5 P4 catalyst incorporating single-atomic Ru sites. Adv Mater. 2020;32:e1906972.

    Article  PubMed  Google Scholar 

  75. Xu B, Li S, Zheng L, Liu Y, Han A, Zhang J, Huang Z, Xie H, Fan K, Gao L, Liu H. A bioinspired five-coordinated single-atom iron nanozyme for tumor catalytic therapy. Adv Mater. 2022;34:e2107088.

    Article  PubMed  Google Scholar 

  76. Zhang J, Liu J, Xi L, Yu Y, Chen N, Sun S, Wang W, Lange KM, Zhang B. Single-atom Au/NiFe layered double hydroxide electrocatalyst: probing the origin of activity for oxygen evolution reaction. J Am Chem Soc. 2018;140:3876–9.

    Article  CAS  PubMed  Google Scholar 

  77. Wang D, Zhang B, Ding H, Liu D, Xiang J, Gao XJ, Chen X, Li Z, Yang L, Duan H, Zheng J, Liu Z, Jiang B, Liu Y, Xie N, Zhang H, Yan X, Fan K, Nie G. TiO2 supported single Ag atoms nanozyme for elimination of SARS-CoV2. Nano Today. 2021;40:101243.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Huang L, Wei X, Gao E, Zhang C, Hu X, Chen Y, Liu Z, Finck N, Lützenkirchen J, Dionysiou DD. Single Fe atoms confined in two-dimensional MoS2 for sulfite activation: a biomimetic approach towards efficient radical generation. Appl Catal B. 2020;268:118459.

    Article  CAS  Google Scholar 

  79. Akri M, Zhao S, Li X, Zang K, Lee AF, Isaacs MA, Xi W, Gangarajula Y, Luo J, Ren Y, Cui Y-T, Li L, Su Y, Pan X, Wen W, Pan Y, Wilson K, Li L, Qiao B, Ishii H, Liao Y-F, Wang A, Wang X, Zhang T. Atomically dispersed nickel as coke-resistant active sites for methane dry reforming. Nat Commun. 2019;10:5181.

    Article  PubMed  PubMed Central  Google Scholar 

  80. Chen Z, Yu Y, Gao Y, Zhu Z. Rational design strategies for nanozymes. ACS Nano. 2023;17:13062–80.

    Article  CAS  PubMed  Google Scholar 

  81. Jiao L, Xu W, Yan H, Wu Y, Liu C, Du D, Lin Y, Zhu C. Fe-N-C single-atom nanozymes for the intracellular hydrogen peroxide detection. Anal Chem. 2019;91:11994–9.

    Article  CAS  PubMed  Google Scholar 

  82. Niu X, Shi Q, Zhu W, Liu D, Tian H, Fu S, Cheng N, Li S, Smith JN, Du D, Lin Y. Unprecedented peroxidase-mimicking activity of single-atom nanozyme with atomically dispersed Fe–Nx moieties hosted by MOF derived porous carbon. Biosens Bioelectron. 2019;142:111495.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Cheng N, Li J, Liu D, Lin Y, Du D. Single-atom nanozyme based on nanoengineered Fe–N–C catalyst with superior peroxidase-like activity for ultrasensitive bioassays. Small. 2019;15:1901485.

    Article  CAS  Google Scholar 

  84. Wu Y, Wu J, Jiao L, Xu W, Wang H, Wei X, Gu W, Ren G, Zhang N, Zhang Q, Huang L, Gu L, Zhu C. Cascade reaction system integrating single-atom nanozymes with abundant Cu sites for enhanced biosensing. Anal Chem. 2020;92:3373–9.

    Article  CAS  PubMed  Google Scholar 

  85. Xu B, Wang H, Wang W, Gao L, Li S, Pan X, Wang H, Yang H, Meng X, Wu Q, Zheng L, Chen S, Shi X, Fan K, Yan X, Liu H. A single-atom nanozyme for wound disinfection applications. Angew Chem Int Ed Engl. 2019;58:4911–6.

    Article  CAS  PubMed  Google Scholar 

  86. Xu W, Song W, Kang Y, Jiao L, Wu Y, Chen Y, Cai X, Zheng L, Gu W, Zhu C. Axial ligand-engineered single-atom catalysts with boosted enzyme-like activity for sensitive immunoassay. Anal Chem. 2021;93:12758–66.

    Article  CAS  PubMed  Google Scholar 

  87. Wu Y, Jiao L, Luo X, Xu W, Wei X, Wang H, Yan H, Gu W, Xu BZ, Du D, Lin Y, Zhu C. Oxidase-like Fe-N-C single-atom nanozymes for the detection of acetylcholinesterase activity. Small. 2019;15:1903108.

    Article  CAS  Google Scholar 

  88. Wang W, Zhu Y, Zhu X, Zhao Y, Xue Z, Xiong C, Wang Z, Qu Y, Cheng J, Chen M, Liu M, Zhou F, Zhang H, Jiang Z, Hu Y, Zhou H, Wang H, Li Y, Liu Y, Wu Y. Biocompatible ruthenium single-atom catalyst for cascade enzyme-mimicking therapy. ACS Appl Mater Interfaces. 2021;13:45269–78.

    Article  CAS  PubMed  Google Scholar 

  89. Cao F, Zhang L, You Y, Zheng L, Ren J, Qu X. An enzyme-mimicking single-atom catalyst as an efficient multiple reactive oxygen and nitrogen species scavenger for sepsis management. Angew Chem Int Ed Engl. 2020;59:5108–15.

    Article  CAS  PubMed  Google Scholar 

  90. Su Y, Wu F, Song Q, Wu M, Mohammadniaei M, Zhang T, Liu B, Wu S, Zhang M, Li A, Shen J. Dual enzyme-mimic nanozyme based on single-atom construction strategy for photothermal-augmented nanocatalytic therapy in the second near-infrared biowindow. Biomaterials. 2022;281:121325.

    Article  CAS  PubMed  Google Scholar 

  91. Ma W, Mao J, Yang X, Pan C, Chen W, Wang M, Yu P, Mao L, Li Y. A single-atom Fe-N4 catalytic site mimicking bifunctional antioxidative enzymes for oxidative stress cytoprotection. Chem Commun. 2018;55:159–62.

    Article  Google Scholar 

  92. Liu H, Li Y, Sun S, Xin Q, Liu S, Mu X, Yuan X, Chen K, Wang H, Varga K, Mi W, Yang J, Zhang X-D. Catalytically potent and selective clusterzymes for modulation of neuroinflammation through single-atom substitutions. Nat Commun. 2021;12:114.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Lu X, Gao S, Lin H, Yu L, Han Y, Zhu P, Bao W, Yao H, Chen Y, Shi J. Bioinspired copper single-atom catalysts for tumor parallel catalytic therapy. Adv Mater. 2020;32:e2002246.

    Article  PubMed  Google Scholar 

  94. Wang X, Shi Q, Zha Z, Zhu D, Zheng L, Shi L, Wei X, Lian L, Wu K, Cheng L. Copper single-atom catalysts with photothermal performance and enhanced nanozyme activity for bacteria-infected wound therapy. Bioact Mater. 2021;6:4389–401.

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Zhu D, Ling R, Chen H, Lyu M, Qian H, Wu K, Li G, Wang X. Biomimetic copper single-atom nanozyme system for self-enhanced nanocatalytic tumor therapy. Nano Res. 2022;15:7320–8.

    Article  CAS  Google Scholar 

  96. Cao F, Sang Y, Liu C, Bai F, Zheng L, Ren J, Qu X. Self-adaptive single-atom catalyst boosting selective ferroptosis in tumor cells. ACS Nano. 2022;16:855–68.

    Article  CAS  PubMed  Google Scholar 

  97. 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;48:1004–76.

    Article  CAS  PubMed  Google Scholar 

  98. Zhang Q, Song L, Zhang K. Breakthroughs in nanozyme-inspired application diversity. Mat Chem Front. 2023;7:44–64.

    Article  CAS  Google Scholar 

  99. Yang D, Chen Z, Gao Z, Tammina SK, Yang Y. Nanozymes used for antimicrobials and their applications. Colloids Surf B Biointerfaces. 2020;195:111252.

    Article  CAS  PubMed  Google Scholar 

  100. Xia D, Liu H, Xu B, Wang Y, Liao Y, Huang Y, Ye L, He C, Wong PK, Qiu R. Single Ag atom engineered 3D-MnO2 porous hollow microspheres for rapid photothermocatalytic inactivation of E. coli under solar light. Appl Catal B-Environ. 2019;245:177–89.

    Article  CAS  Google Scholar 

  101. Huo M, Wang L, Zhang H, Zhang L, Chen Y, Shi J. Construction of single-iron-atom nanocatalysts for highly efficient catalytic antibiotics. Small. 2019;15:1901834.

    Article  Google Scholar 

  102. Liu H, Liu X, Wang H, Ren J, Qu X. A homing missile-like nanotherapeutic with single-atom catalytic sites for in situ elimination of intracellular bacterial pathogens. Small. 2023;19:2207510.

    Article  CAS  Google Scholar 

  103. Qiu X, Zhuang L, Yuan J, Wang H, Dong X, He S, Guan S, Chang Z, Bao P. Constructing multifunctional Cu single-atom nanozyme for synergistic nanocatalytic therapy-mediated multidrug-resistant bacteria infected wound healing. J Colloid Interface Sci. 2023;652:1712–25.

    Article  CAS  PubMed  Google Scholar 

  104. Chan M, Chen B-G, Huang W, Su T, Hsiao M, Liu R. Tunable single-atom nanozyme catalytic system for biological applications of therapy and diagnosis. Mater Today Adv. 2023;17:100342.

    Article  CAS  Google Scholar 

  105. Wang X, Chen Q, Zhu Y, Wang K, Chang Y, Wu X, Bao W, Cao T, Chen H, Zhang Y, Qin H. Destroying pathogen-tumor symbionts synergizing with catalytic therapy of colorectal cancer by biomimetic protein-supported single-atom nanozyme. Signal Transduct Target Ther. 2023;8:277.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Lv Q, Chi K, Shi X, Liu M, Li X, Zhou C, Shi L, Fan H, Liu H, Liu J, Zhang Y, Wang S, Wang L, Wang Z. Nanozyme-like single-atom catalyst combined with artesunate achieves photothermal-enhanced nanocatalytic therapy in the near-infrared biowindow. Acta Biomater. 2023;158:686–97.

    Article  CAS  PubMed  Google Scholar 

  107. Madhuvilakku R, Hong Y, Nila IS, Villagra Moran VM, Subramanian P, Khan ZA, Jeong S, You SG. Quantification of neuronal cell-released hydrogen peroxide using 3D mesoporous copper-enriched prussian blue microcubes nanozymes: a colorimetric approach in real time and anticancer effect. ACS Appl Mater Interfaces. 2023;15:55466–85.

    Article  CAS  PubMed  Google Scholar 

  108. Liu Y, Wang B, Zhu J, Xu X, Zhou B, Yang Y. Single-atom nanozyme with asymmetric electron distribution for tumor catalytic therapy by disrupting tumor redox and energy metabolism homeostasis. Adv Mater. 2023;35:2208512.

    Article  CAS  Google Scholar 

  109. Zhao X, Wu J, Zhang K, Guo D, Hong L, Chen X, Wang B, Song Y. The synthesis of a nanodrug using metal-based nanozymes conjugated with ginsenoside Rg3 for pancreatic cancer therapy. Nanoscale Adv. 2021;4:190–9.

    Article  PubMed  PubMed Central  Google Scholar 

  110. Sun H, Liu J. A feasible strategy for designing cytochrome P450-mimic sandwich-like single-atom nanozymes toward electrochemical CO2 conversion. J Colloid Interface Sci. 2024;661:482–92.

    Article  CAS  PubMed  Google Scholar 

  111. Feng N, Li Q, Bai Q, Xu S, Shi J, Liu B, Guo J. Development of an Au-anchored Fe single-atom nanozyme for biocatalysis and enhanced tumor photothermal therapy. J Colloid Interface Sci. 2022;618:68–77.

    Article  CAS  PubMed  Google Scholar 

  112. Chang M, Hou Z, Wang M, Yang C, Wang R, Li F, Liu D, Peng T, Li C, Lin J. Single-atom Pd nanozyme for ferroptosis-boosted mild-temperature photothermal therapy. Angew Chem Int Ed Engl. 2021;60:12971–9.

    Article  CAS  PubMed  Google Scholar 

  113. Deng Z, Ma W, Ding C, Wei C, Gao B, Zhu Y, Zhang Y, Wu F, Zhang M, Li R, Zhang M. Metal polyphenol network/cerium oxide artificial enzymes therapeutic nanoplatform for MRI/CT-aided intestinal inflammation management. Nano Today. 2023;53:102044.

    Article  CAS  Google Scholar 

  114. Lu M, Wang C, Ding Y, Peng M, Zhang W, Li K, Wei W, Lin Y. Fe-N/C single-atom catalysts exhibiting multienzyme activity and ROS scavenging ability in cells. Chem Commun. 2019;55:14534–7.

    Article  CAS  Google Scholar 

  115. Chen Y, Zou H, Yan B, Wu X, Cao W, Qian Y, Zheng L, Yang G. Atomically dispersed Cu nanozyme with intensive ascorbate peroxidase mimic activity capable of alleviating ROS-mediated oxidation damage. Adv Sci. 2022;9:2103977.

    Article  CAS  Google Scholar 

  116. Liu Y, Cheng Y, Zhang H, Zhou M, Yu Y, Lin S, Jiang B, Zhao X, Miao L, Wei C-W, Liu Q, Lin Y-W, Du Y, Butch CJ, Wei H. Integrated cascade nanozyme catalyzes in vivo ROS scavenging for anti-inflammatory therapy. Sci Adv. 2020;6:2695.

    Article  Google Scholar 

  117. Jo S-M, Zhang KAI, Wurm FR, Landfester K. Mimic of the cellular antioxidant defense system for a sustainable regeneration of nicotinamide adenine dinucleotide (NAD). ACS Appl Mater Interfaces. 2020;12:25625–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Morry J, Ngamcherdtrakul W, Yantasee W. Oxidative stress in cancer and fibrosis: opportunity for therapeutic intervention with antioxidant compounds, enzymes, and nanoparticles. Redox Biol. 2017. https://doi.org/10.1016/j.redox.2016.12.011.

    Article  PubMed  Google Scholar 

  119. Chen D, Xia Z, Guo Z, Gou W, Zhao J, Zhou X, Tan X, Li W, Zhao S, Tian Z, Qu Y. Bioinspired porous three-coordinated single-atom Fe nanozyme with oxidase-like activity for tumor visual identification via glutathione. Nat Commun. 2023;14:7127.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Sun L, Li C, Yan Y, Yu Y, Zhao H, Zhou Z, Wang F, Feng Y. Engineering DNA/Fe-N-C single-atom nanozymes interface for colorimetric biosensing of cancer cells. Anal Chim Acta. 2021;1180:338856.

    Article  CAS  PubMed  Google Scholar 

  121. Hu FX, Hu T, Chen S, Wang D, Rao Q, Liu Y, Dai F, Guo C, Yang HB, Li CM. Single-atom cobalt-based electrochemical biomimetic uric acid sensor with wide linear range and ultralow detection limit. Nanomicro Lett. 2020;13:7.

    PubMed  PubMed Central  Google Scholar 

  122. Zhou X, Wang M, Chen J, Xie X, Su X. Peroxidase-like activity of Fe-N-C single-atom nanozyme based colorimetric detection of galactose. Anal Chim Acta. 2020;1128:72–9.

    Article  CAS  PubMed  Google Scholar 

  123. Li J, Li Y, Wu K, Deng A, Li J. Ultra-sensitive detection of 5-fluorouracil by flow injection chemiluminescence immunoassay based on Fenton-like effect of single atom Co nanozyme. Talanta. 2023;265:124870.

    Article  CAS  PubMed  Google Scholar 

  124. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin. 2022;72:7–33.

    Article  PubMed  Google Scholar 

  125. Ni J, Wu G, Albenberg A, Tomov V. Gut microbiota and IBD: causation or correlation? Nat Rev Gastroenterol Hepatol. 2017;14:573–84.

    Article  PubMed  PubMed Central  Google Scholar 

  126. Wong SH, Jun Y. Gut microbiota in colorectal cancer: mechanisms of action and clinical applications. Nat Rev Gastroenterol Hepatol. 2019;16:690–704.

    Article  CAS  PubMed  Google Scholar 

  127. Lang S, Duan Y, Liu J, Torralba M, Kuelbs K, Ventura-Cots M, Abraldes J, Abraldes F, Verna E, Brown R, Brown V, Altamirano J, Caballería J, Shawcross D, Lucey M, Louvet A, Mathurin P, Garcia-Tsao G-T, Ho S, Tu X, Bataller R, Stärkel P, Fouts F, Schnabl B. Intestinal fungal dysbiosis and systemic immune response to fungi in patients with alcoholic hepatitis. Hepatology. 2020;71:522–38.

    Article  CAS  PubMed  Google Scholar 

  128. Tenner S, Baillie J, DeWitt J, Vege S. American college of gastroenterology guideline: management of acute pancreatitis. Am J Gastroenterol. 2013;108:1400–15.

    Article  CAS  PubMed  Google Scholar 

  129. Zhu Y, Liao Y, Zou J, Cheng J, Pan Y, Lin L, Chen X. Engineering single-atom nanozymes for catalytic biomedical applications. Small. 2023;19:2300750.

    Article  CAS  Google Scholar 

  130. Zhang Y, Wang T, Dong X, Zhu C, Peng Q, Liu C, Zhang Y, Chen F, Zhang K. Salivary amylase-responsive buccal tablets wipe out chemotherapy-rooted refractory oral mucositis. Adv Sci. 2024;11:2308439.

    Article  CAS  Google Scholar 

  131. Jiao R, Lin X, Zhang Q, Zhang Y, Qin W, Yang Q, Xu C, Chen F, Zhang K. Anti-tumor immune potentiation targets-engineered nanobiotechnologies: design principles and applications. Prog Mater Sci. 2024;142:101230.

    Article  CAS  Google Scholar 

Download references

Funding

Zhejiang Province's 2024 Key R&D Plan Project (Grant No. 2024C03048); Zhejiang Provincial Traditional Chinese Medicine Science and Technology Project (Grant No. GZY-ZJ-KJ-24093); Hangzhou Science and Technology Commission (202004A14); and the Construction Fund of Medical Key Disciplines of Hangzhou (OO20190001); Research Fund of Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital.

Author information

Authors and Affiliations

Authors

Contributions

# Hua S.J., Dong X.L. and Peng Q.X. contributed equally to this work. Conceptualization: Hua S.J.; Writing-original draft preparation: Hua S.J and Peng Q.X.; Writing-review and editing: Dong X.L.; Supervision: Zhang X.F., Zhang K.; Funding acquisition: Yang J.F.. All authors have read and agreed to the published version of the manuscript.

Corresponding authors

Correspondence to Kun Zhang, Xiaofeng Zhang or Jianfeng Yang.

Ethics declarations

Informed consent

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hua, S., Dong, X., Peng, Q. et al. Single-atom nanozymes shines diagnostics of gastrointestinal diseases. J Nanobiotechnol 22, 286 (2024). https://doi.org/10.1186/s12951-024-02569-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12951-024-02569-3

Keywords