Yifan
Lai†
a,
Jingyu
Wang†
a,
Ning
Yue
a,
Qiaochu
Zhang
a,
Jiangjiexing
Wu
*bc,
Wei
Qi
a and
Rongxin
Su
*abc
aState Key Laboratory of Chemical Engineering, Tianjin Key Laboratory of Membrane Science and Desalination Technology, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P.R. China. E-mail: surx@tju.edu.cn
bZhejiang Institute of Tianjin University, Ningbo, Zhejiang 315201, P.R. China. E-mail: wujiangjiexing2007@126.com; wjjx1987@tju.edu.cn
cSchool of Marine Science and Technology, Tianjin University, Tianjin 300072, P.R. China
First published on 30th January 2023
The field of nanozymes is developing rapidly. In particular, glutathione peroxidase (GPx)-like nanozymes, which catalytically reduce H2O2/organic hydroperoxides to H2O/alcohols, have attracted considerable attention. GPx-like nanozymes are powerful antioxidant enzymes known to combat oxidative stress. They have broad applications, including cytoprotection, anti-inflammation, neuroprotection, tumor therapy, and anti-aging. Although much progress has been made, GPx-like nanozymes have not been well discussed or fully reviewed as other nanozymes. This review aims to summarize recent advances on GPx-like nanozymes from the vantage point of mechanism, classification, and bioapplication. Future prospects for advancing their design and application are also discussed.
Jingyu Wang is currently pursuing her Ph.D. degree at the School of Chemical Engineering and Technology, Tianjin University under the supervision of Prof. Rongxin Su. She received her B.S. degree from Tianjin University in 2021. Her research interests are the synthesis and applications of functional nanomaterials. |
GPx, an antioxidant enzyme, is responsible for maintaining the cellular redox balance. In 1957, GPx was identified as a protective factor for hemoglobin within erythrocytes.17 Then, selenium was found to be essential to GPx.18,19 The identified crystal structure of GPx further revealed a catalytically active site containing selenium in the form of selenocysteine.20 Studies on GPx showed that it could catalyze the reduction of H2O2/organic hydroperoxides to H2O/alcohols under reduced GSH.21 Consequently, it can be effective in inhibiting and fighting oxidative stress. It suffers, however, from various intrinsic defects as most natural enzymes, such as being too expensive, unstable, and difficult to mass produce.22 Therefore, it is vital to explore GPx mimics extensively to overcome the limitations mentioned above and facilitate practical applications.23
Due to the discovery that GPx is a selenoenzyme, researchers initially looked at organoselenium compounds.24–29 Other compounds, such as organotellurium compounds, have also been identified as potential GPx mimics.30 Following the discovery in 2007 of magnetic Fe3O4 nanoparticles to possess peroxidase (POD)-like activity, nanozymes, nanomaterials with enzyme-like properties, have gained widespread attention.31–35 Since nanozymes are low-cost, highly stable, multifunctional, and easy to mass produce, they are considered the next generation of artificial enzymes.36–38 The discovery that V2O5 nanowires resemble GPx further catalyzed the breakthrough and opened up new possibilities.39 As of today, GPx-like nanozymes have been explored and developed for applications including cytoprotection, anti-inflammation, neuroprotection, and tumor therapy (Scheme 1). In order to highlight the recent progress, an overview of the development of GPx-like nanozymes is provided. This review will cover GPx-like nanozymes’ mechanism, classification, and bioapplications. A discussion on the challenges and future developing prospects of GPx-like nanozymes ends the paper.
Fig. 1 (A) Ping-pong mechanism of natural GPx. Reprinted with permission from ref. 42. Copyright (2013) Elsevier. (B) A catalytic mechanism for V2O5 nanowires to exhibit GPx-like activity. Reprinted with permission from ref. 39. Copyright (2014) Springer Nature. |
In some cases, however, peroxide intermediates would not form at all. This is because the active site would first interact with GSH rather than H2O2, changing its redox state. Typical examples are the GPx-like catalytic reactions of CuV2O6 with GSH and H2O2, which differ greatly from the previously reported V2O5 nanowires.51 As shown in Fig. 2A and B, the absence of peaks at 1150 cm−1 and 930–940 cm−1 in Raman and infrared spectra indicated that no V-peroxide species formed. Additional X-ray photoelectron spectroscopic analysis of CuV2O6 interacting with GSH and extra H2O2 allowed further investigation of the intermediates (Fig. 2C and D). Following interaction with GSH, the valence state of V was clearly exhibited as +IV, and after the addition of excess H2O2, both +V and +IV products were formed. Fig. 2E shows the two equations corresponding to the reactions involved. According to the above results, the GPx-like activity of CuV2O6 was caused by a redox change in the vanadium center. The redox valence changes of active centers have also been observed in other types of nanozymes, such as Mn-,52 Cu-,53,54 Ru-based,55etc. In some bimetallic nanozymes, both active sites would produce valence redox changes.56–58 As an example, Sn2+/Sn4+ and Fe2+/Fe3+ redox couples played an important role in catalyzing the GPx-like reaction of SnFe2O4.57
Fig. 2 (A and B) Changes in the Raman and infrared spectra of CuV2O6 reacting with H2O2 with time. (C and D) X-ray photoelectron spectroscopic analysis of V 2p after CuV2O6 has been treated with 10 mM GSH and 0.1 M H2O2, respectively. (E) Catalytic equations for the GPx-like activity of CuV2O6. Adapted with permission from ref. 51. Copyright (2022) Royal Society of Chemistry. |
Fig. 3 (A) The flow chart of the ordered mechanism. Adapted with permission from ref. 41. Copyright (2004) John Wiley and Sons. (B) The construction of selenopeptide-modified Au NPs. (C and D) The double-reciprocal plots of GPx-like activity for selenopeptides. (E and F) The double-reciprocal plots of GPx-like activity for selenopeptide-modified Au NPs. Adapted with permission from ref. 59. Copyright (2020) Royal Society of Chemistry. |
Fig. 4 A brief timeline for the development of GPx-like nanozymes (natural and artificial enzymes are listed for better understanding). Note: for each active atom, only representative nanomaterials are listed, and more detailed information can be found in Table 1. |
Active atom | Nanozyme | Types of enzyme-like activities | Application | Function of GPx-like activity | Ref. |
---|---|---|---|---|---|
(A) The “Active atom” and “Nanozyme” columns are listed in alphabetical order. (B) Organoselenium compounds, organotellurium compounds, and nanozymes obtained by combining them with supramolecular-assembled scaffolds are not listed. More information can be found in ref. 65. | |||||
Au | Au24Ag1 clusterzyme | GPx/CAT | Relieving acute neuroinflammation and treating trauma brain injury | Scavenging H2O2 and lipid peroxides | 104 |
Au25 clusterzyme | GPx (highest)/CAT/SOD | Relieving acute neuroinflammation and treating trauma brain injury | Scavenging H2O2 and lipid peroxides | 103 and 104 | |
Au24Cd1 clusterzyme | GPx/CAT/SOD (highest) | Relieving neuroinflammation by inhibiting interleukin (IL)-1β and IL-6 | Scavenging H2O2 and lipid peroxides | 103 | |
Au24Cu1 clusterzyme | GPx/CAT (highest)/SOD | Relieving neuroinflammation by inhibiting tumor necrosis factor (TNF)-α | Scavenging H2O2 and lipid peroxides | 103 | |
Ce | CeO2 | GPx/CAT/POD/SOD | Relieving neuroinflammation and treating trauma brain injury noninvasively | Scavenging H2O2 and synergizing with SOD- and CAT-like activities to eliminate oxidative stress | 105 and 106 |
Cr/CeO2 | GPx/CAT/POD/SOD | Relieving neuroinflammation and treating trauma brain injury noninvasively | Scavenging H2O2 and synergizing with SOD- and CAT-like activities to eliminate oxidative stress | 106 | |
Pt/CeO2 | GPx/CAT/POD/SOD | Relieving neuroinflammation and treating trauma brain injury noninvasively | Scavenging H2O2 and synergizing with SOD- and CAT-like activities to eliminate oxidative stress | 105 | |
PEG/Ce-Bi@DMSN | GPx/CAT/POD | Relieving tumor hypoxia and depleting overexpressed GSH for tumor therapy | Depleting overexpressed GSH to decrease the scavenging of ROS and improve ˙OH-mediated tumor nanocatalytic therapy | 119 | |
Co | Co/PMCS | GPx/CAT/SOD | Alleviating systematic inflammation and treating sepsis | Scavenging H2O2 and synergizing with SOD- and CAT-like activities to eliminate oxidative stress | 107 |
Cu | COF-618-Cu | GPx/CAT | Relieving tumor hypoxia and depleting overexpressed GSH for tumor therapy | Depleting overexpressed GSH to decrease the scavenging of ROS and improve the photodynamic and photothermal therapy efficacy | 97 |
COF-909-Cu | GPx/POD/SOD | Elevating H2O2 levels and inducing pyroptosis for tumor therapy | Depleting overexpressed GSH | 98 | |
Cu nanoclusters (Cu NCs) | GPx/CAT/POD/SOD | Defending cells from oxidative stress by scavenging excess ROS | Scavenging H2O2 and fine-tuning the concentration of H2O2 for cell signalling | 96 | |
Cu-TCPP MOF nanodots (CTMDs) | GPx/SOD | Alleviating acute kidney injury during endotoxemia | Scavenging H2O2 at the inflammatory sites and avoiding the toxicity of SOD-like activity | 95 | |
Cu-doped polypyrrole (CuPP) | GPx/CAT/POD | Relieving tumor hypoxia and reprogramming macrophages for tumor therapy | Depleting overexpressed GSH to decrease the scavenging of ROS and improve ˙OH-mediated tumor immunotherapy | 54 | |
CuO NP-polyoxometalate (POM) | GPx/CAT/OXD/POD | Constructing multi-function sensors and sensing ascorbic acid and Fe2+ | Constructing a fluorometric sensor for Fe2+ | 94 | |
CuO, Cu2O | GPx | — | — | 51 | |
CuxO nanoparticle clusters | GPx/CAT/POD/SOD | Inhibiting neurotoxicity and ameliorating Parkinson's disease | Scavenging H2O2 and synergizing with SOD- and CAT-like activities to protect cells from ROS cytotoxicity | 90 | |
CuxO@EM-K | GPx/CAT/SOD | Clearing peripheral amyloid-β associated with Alzheimer's disease | Scavenging H2O2 and synergizing with CAT- and SOD-like activities to mitigate Aβ-induced erythrocyte membrane oxidative damage | 91 | |
Cu5.4O nanoparticles | GPx/CAT/SOD | Scavenging excessive ROS and alleviating inflammation-related diseases | Scavenging H2O2 and synergizing with SOD- and CAT-like activities to protect cells from ROS damage | 92 | |
Cu5.4O@Hep-PEG hydrogels | GPx/CAT/SOD | Inhibiting inflammation and promoting wound healing | Scavenging H2O2 and synergizing with CAT- and SOD-like activities to scavenge ROS from wound sites | 93 | |
PtCu3-PEG nanocages | GPx/POD | Depleting overexpressed GSH and enhancing sonodynamic tumor therapy | Depleting overexpressed GSH to decrease the scavenging of ROS in chemodynamic-enhanced sonodynamic cancer therapy | 53 | |
Fe | Fe-CDs@Ang | GPx/CAT/OXD/POD/SOD/TPx | Activating autophagy-lysosome pathway for drug-resistant glioblastoma therapy | Depleting overexpressed GSH to make tumor cells more sensitive to the ROS changes | 110 |
Fe-N/C SACs | GPx/CAT/OXD/POD | Protection of cells from oxidative stress by scavenging ROS | Scavenging H2O2 and regulating the concentration of H2O2 to deliver cellular signals | 108 | |
KCa(H2O)2[FeIII(CN)6]·H2O (CaPB) | GPx/CAT/POD/SOD | Regulating inflammation and inhibiting ferroptosis in acute kidney injury therapy | Scavenging H2O2 and synergizing with CAT- and SOD-like activities to effectively scavenge diverse RONS | 111 | |
Mn | C-Mn3O4 nanoparticles | GPx | Passing the blood–brain barrier and treating Huntington's disease | Scavenging H2O2 and reducing the associated oxidative damage, thereby preventing the mitochondrial dysfunctions and the concomitant redox imbalance | 49 |
HSA-Mn3O4 | GPx/CAT/SOD | Alleviating reperfusion-induced nervous system damage in ischemic stroke | Scavenging H2O2 and synergizing with CAT- and SOD-like activities to effectively scavenge ROS | 86 | |
LiMn2O4 | GPx/CAT/SOD | Treating inflammatory bowel disease with a low dosage | Scavenging H2O2 and synergizing with CAT- and SOD-like activities to achieve self-cascading antioxidant activity and effectively scavenge ROS | 89 | |
MnO2-BSA nanoparticles | GPx/CAT/OXD/POD/SOD | Protection of cells from oxidative stress by scavenging ROS | Scavenging H2O2 | 88 | |
Mn3O4 nanoflowers | GPx/CAT/SOD | Providing cytoprotection in Parkinson's disease | Scavenging H2O2 and fine-tuning the concentration of H2O2 for cell signaling | 84 | |
Keeping NO bioavailability in human endothelial cells | Scavenging H2O2 and fine-tuning the concentration of H2O2 for cell signaling | 52 | |||
Mn3O4 in nanoform | GPx/CAT/SOD | Defending cells from oxidative stress by scavenging excess ROS | Scavenging H2O2 and fine-tuning the concentration of H2O2 for cell signaling | 85 | |
Mn3O4 nanorods | GPx/CAT/POD/SOD | Scavenging excessive ROS and ameliorating Parkinson's disease | Scavenging H2O2 and synergizing with CAT- and SOD-like activities to effectively scavenge ROS | 87 | |
Mo | MoS2 | GPx/CAT/POD/SOD | Relieving hepatic fibrosis and suppressing inflammation | Scavenging H2O2 and synergizing with CAT- and SOD-like activities and constituting a self-cascade platform to inhibit ROS production | 112 |
MoO3 nanowires | GPx | — | — | 50 | |
Pt | Au@Pt nanoparticles | GPx/polyphenol oxidase /lipoxygenase | — | — | 113 |
Janus BPQDs/Pt/AFSNs | GPx/CAT/OXD/POD/SOD | Scavenging excessive ROS and treating ROS-induced ear inflammation | Scavenging H2O2 | 114 | |
Ru | Pt(IV)–Ru hybrid prodrug | GPx/CAT/POD | Alleviating tumor hypoxia, enhancing oxidative stress, and depleting GSH | Depleting overexpressed GSH to overcome cisplatin resistance | 55 |
RuO2 nanoparticles | GPx/CAT/POD/SOD | Alleviating acute kidney injury | Scavenging H2O2 and synergizing with CAT- and SOD-like activities and exhibiting the cytoprotective effect on oxidative stress-induced nephrotoxicity | 115 | |
Se | 2D MoSe2@PVP nanosheets | GPx/CAT/POD/SOD | Scavenging RONS with heat-resistance and alleviating acute pancreatitis | Scavenging H2O2 and synergizing with CAT- and SOD-like activities to effectively scavenge ROS | 76 |
Fe2NC@Se nanoparticles | GPx/CAT/OXD/SOD | Inhibiting neural apoptosis after cerebral ischemia-reperfusion injury | Scavenging H2O2 and synergizing with CAT- and SOD-like activities to effectively scavenge ROS | 73 | |
GO-Se nanocomposite | GPx | Protection of cells from oxidative stress by scavenging ROS | Scavenging H2O2 and protecting cells from ROS damage | 43 | |
GO (mPEGSe)2 | GPx | Modulating the balance of ROS | Scavenging H2O2 and protecting cells from a high ROS level | 71 | |
HEP-SeNPs | GPx/CAT/SOD | Defending cells from oxidative stress by scavenging excess ROS | Scavenging H2O2 and synergizing with CAT- and SOD-like activities to effectively scavenge ROS | 79 | |
Lf-Au-Bi2Se3 nanodots | GPx/CAT/POD/SOD | Passing the blood–brain barrier and treating Parkinson's disease | Scavenging H2O2 and synergizing with CAT- and SOD-like activities to alleviate ROS levels and ameliorate the mitochondrial state | 74 | |
MoSe2-PVP nanoparticles | GPx/CAT/POD/SOD | Scavenging RONS with heat-resistance and alleviating acute pancreatitis | Scavenging H2O2 | 75 | |
PEG-modified Mo3Se4 nanoflakes | GPx/CAT/POD/SOD | Inhibiting colitis by preventing oxidative damage and intestine barrier breakdown | Scavenging H2O2 and synergizing with CAT- and SOD-like activities to scavenge ROS and reconstitute the Nrf2-mediated cellular anti-oxidative system | 77 | |
MSe-HA nanoparticles | GPx | Treating local inflammation and sepsis injury simultaneously | Scavenging H2O2 and inhibiting the overexpression of pro-inflammatory cytokines | 45 | |
PDASeCys | GPx/CAT/SOD | Treating Parkinson's disease | Scavenging H2O2 and synergizing with CAT- and SOD-like activities to effectively scavenge ROS | 78 | |
Se-DG | GPx | Serving as a green catalyst in alkene epoxidation with H2O2 | Supporting the ability to activate H2O2 | 120 | |
Se@Me@MnO2 | GPx/CAT/SOD | Resisting ROS-mediated damage and alleviating ear inflammation | Scavenging H2O2 and synergizing with CAT- and SOD-like activities to scavenge ROS | 121 | |
Se@pDA | GPx | Alleviating lung inflammation and protecting cells against oxidative damage | Scavenging H2O2 effectively | 44 | |
(Sec-Arg-Gly-Asp-Cys)-modified Au nanoparticles | GPx | — | — | 59 | |
UiO-66-Se | GPx | — | — | 72 | |
V | CuV2O6, MnV2O6, ZnV2O6 | GPx | Increasing the NO bioavailability and protecting cells from excessive ROS | Scavenging H2O2 and mediating the release of NO from S-nitrosothiols at physiological pH | 51 |
MIL-47(V)-X (X = F, Br, NH2, CH3, OH, and H) | GPx | Alleviating a broad-spectrum inflammation including ear injury and colitis | Scavenging H2O2 and inducing the M1 to M2 phenotypic polarization of macrophages | 48 | |
GdVO4/Eu3+ nanoparticles | GPx | Exerting a direct antioxidant effect and activating GSH signalling for anti-aging | Scavenging H2O2 and activating GSH signalling for anti-aging | 81 and 82 | |
VN4 single-atom | GPx/CAT/POD | Accelerating the scalp healing from brain trauma | Scavenging H2O2 and lipid peroxides accumulated in the scalp following injuries | 83 | |
V2O5 nanosheets | GPx | Counteracting redox stress and blocking reactivation and replication of HIV-1 | Scavenging H2O2, counteracting redox stress and blocking the reactivation and replication of HIV-1 | 122 | |
V2O5 nanowires | GPx | Scavenging ROS and protecting cells from oxidative stress | Scavenging H2O2 and supplying the antioxidant system of the cell | 39 and 46 | |
V2O5@pDA@MnO2 | GPx/CAT/SOD | Alleviating ear inflammation and protecting cells against oxidative damage | Scavenging H2O2 and synergizing with CAT- and SOD-like activities to scavenge ROS | 123 | |
2D V2C MXenzyme | GPx/CAT/POD/SOD/TPx/hydroperoxide lyase | Treating ROS-mediated inflammatory and neurodegenerative diseases | Scavenging H2O2 and maintaining appropriate H2O2 levels in cells | 47 | |
Protecting nerves against ischemic stroke and serving for MR imaging | 80 | ||||
W | WS2-PVP nanoflowers | GPx/CAT/SOD | Reducing inflammation caused by acute liver injury | Scavenging H2O2 and catalyzing GSH to reduce peroxide | 118 |
Zn | ZnO | GPx/glycosidase | Catalysing the generation of NO from endogenous and exogenous prodrugs | Catalytically decomposing endogenous GSNO to generate NO | 124 |
Multiple active centers | BN-GDY | GPx/POD | Inhibiting colorectal cancer cell proliferation through attenuating tumor hypoxia | Depleting overexpressed GSH and hindering the GSH-induced ROS scavenging capacity | 125 |
CuFeP | GPx/POD | Depleting overexpressed GSH and enhancing oxidative stress for tumor therapy | Depleting overexpressed GSH to decrease the scavenging of ROS and improve ˙OH-mediated tumor therapy | 58 | |
CuFe2O4@PEG | GPx/CAT/POD | Enhancing oxidative stress, reducing GSH, and alleviating tumor hypoxia | Depleting overexpressed GSH to decrease the scavenging of ROS and improve ˙OH-mediated tumor therapy | 117 | |
IMOP | GPx/CAT | Relieving tumor hypoxia and depleting overexpressed GSH for tumor therapy | Depleting overexpressed GSH to decrease the scavenging of ROS | 116 | |
MnFe2O4@MOF | GPx/CAT | Relieving tumor hypoxia and depleting overexpressed GSH for tumor therapy | Depleting overexpressed GSH to decrease the scavenging of ROS | 56 | |
SnFe2O4 | GPx/CAT | Relieving tumor hypoxia and depleting overexpressed GSH for tumor therapy | Depleting overexpressed GSH to decrease the scavenging of ROS and improve ˙OH- and O2˙−-mediated synergistic tumor therapy | 57 | |
ZIF-67/Cu0.76Co2.24O4 nanospheres | GPx/POD/SOD/laccase | Monitoring of 3,4-dihydroxyphenylacetic acid based on laccase-like activity | — | 126 | |
O-NZ | GPx/SOD | Scavenging RONS and alleviating acute brain injury | Scavenging H2O2 | 127 |
Active atom | Nanozyme | Substrate | K m/mM | V max/μM min−1 | K cat/s−1 | Experiential conditions | Ref. | |
---|---|---|---|---|---|---|---|---|
Au | Au25 clusterzyme | H2O2 | 470 | C(GSH) = 2 mM, C(NADPH) = 200 mM, C(GR) = 1.7 U mL−1, C(nanozyme) = 10 ng μL−1 | 103 | |||
Au24Cd1 clusterzyme | H2O2 | 100 | ||||||
Au24Cu1 clusterzyme | H2O2 | 340 | ||||||
Co | Co/PMCS | H2O2 | 0.26 | 17.44 | 25 °C, pH = 7.4, C(GSH) = 2 mM, C(NADPH) = 0.4 mM, C(GR) = 1.7 U, C(nanozyme) = 2.5 μM | 107 | ||
GSH | 2.81 | 12.97 | 25 °C, pH = 7.4, C(H2O2) = 240 μM, C(NADPH) = 0.4 mM, C(GR) = 1.7 U, C(nanozyme) = 2.5 μM | |||||
Cu | Cu nanoclusters (Cu NCs) | H2O2 | 0.097 | 6.781 | 37 °C, pH = 7.2, C(GSH) = 1 mM, C(DTNB) = 1 mM, C(nanozyme) = 20 μg mL−1 | 96 | ||
GSH | 0.913 | 4.766 | 37 °C, pH = 7.2, C(H2O2) = 1 mM, C(DTNB) = 1 mM, C(nanozyme) = 20 μg mL−1 | |||||
Cu-TCPP MOF nanodots (CTMDs) | H2O2 | 0.199 ± 0.12 | 25 °C, pH = 7.4, C(GSH) = 2 mM, C(NADPH) = 0.4 mM, C(GR) = 1.7 U, C(nanozyme) = 3.5 μg mL−1 (with the Cu content) | 95 | ||||
GSH | 1.25 ± 0.48 | 25 °C, pH = 7.4, C(H2O2) = 240 μM, C(NADPH) = 0.4 mM, C(GR) = 1.7 U, C(nanozyme) = 3.5 μg mL−1 (with the Cu content) | ||||||
CuO NP-POM | H2O2 | 0.15 | 7.806 | pH = 7.4, C(GSH) = 1 mM, C(NADPH) = 0.1 mM, C(GR) = 2 μM, C(nanozyme) = 100 μg mL−1 | 94 | |||
GSH | 0.11 | 4.71 | pH = 7.4, C(H2O2) = 1 mM, C(NADPH) = 0.1 mM, C(GR) = 2 μM, C(nanozyme) = 100 μg mL−1 | |||||
CuxO nanoparticle clusters | H2O2 | 0.163 | 295 | 25 °C, pH = 7.4, C(GSH) = 2 mM, C(NADPH) = 200 μM, C(GR) = 1.7 U, C(nanozyme) = 20 μg mL−1 | 90 | |||
GSH | 1.89 | 188 | 25 °C, pH = 7.4, C(H2O2) = 500 μM, C(NADPH) = 200 μM, C(GR) = 1.7 U, C(nanozyme) = 20 μg mL−1 | |||||
Fe | Fe-CDs@Ang | H2O2 | 4.52 | 81.96 | C (GSH) = 8.4 mM, C(NADPH) = 300 μM, C(nanozyme) = 100 μg mL−1 | 110 | ||
Mn | C-Mn3O4 nanoparticles | H2O2 | 1.09 ± 0.06 | 95 ± 11 | 1.152 ± 0.009 | 25 °C, pH = 7.4, C(GSH) = 2 mM, C(NADPH) = 400 μM, C(GR) = 1.7 U, C(nanozyme) = 1.3 μM | 49 | |
GSH | 1.36 ± 0.09 | 64 ± 8 | 0.779 ± 0.005 | 25 °C, pH = 7.4, C(H2O2) = 240 μM, C(NADPH) = 400 μM, C(GR) = 1.7 U, C(nanozyme) = 1.3 μM | ||||
MnO2-BSA nanoparticles | H2O2 | 0.15 | 4 | C(GSH) = 2 mM, C(nanozyme) = 10 μg mL−1 | 88 | |||
GSH | 4.03 | 16 | C(H2O2) = 2.5 mM, C(nanozyme) = 10 μg mL−1 | |||||
Mn3O4 nanoflowers | H2O2 | 0.196 | 56 | 25 °C, pH = 7.4, C(GSH) = 2 mM, C(NADPH) = 200 μM, C(GR) = 1.7 U, C(nanozyme) = 10 ng μL−1 | 84 | |||
GSH | 1.16 | 78 | 25 °C, pH = 7.4, C(H2O2) = 1 mM, C(NADPH) = 200 μM, C(GR) = 1.7 U, C(nanozyme) = 10 ng μL−1 | |||||
Mn3O4 cubes | H2O2 | 0.989 | 18.2 | 25 °C, pH = 7.4, C(GSH) = 2 mM, C(NADPH) = 200 μM, C(GR) = 1.7 U, C(nanozyme) = 10 ng μL−1 | 52 | |||
Mn3O4 polyhedra | H2O2 | 2.845 | 16.9 | |||||
Mn3O4 flakes | H2O2 | 0.431 | 39.1 | |||||
Mn3O4 nanoflowers | H2O2 | 0.19 | 54.5 | |||||
Mo | MoO3 nanowires | H2O2 | 5.61 ± 0.18 | 840.34 ± 26.48 | 25 °C, pH = 7.4, C(GSH) = 2 mM, C(NADPH) = 0.2 mM, C(GR) = 1.7 U, C(nanozyme) = 20 μg mL−1 | 50 | ||
GSH | 3.06 ± 0.03 | 323.62 ± 3.09 | 25 °C, pH = 7.4, C(H2O2) = 200 μM, C(NADPH) = 0.2 mM, C(GR) = 1.7 U, C(nanozyme) = 20 μg mL−1 | |||||
MoS2 | H2O2 | 0.04 | 39.6 | 1.05 × 10−3 | C(GSH) = 2 mM, C(NADPH) = 0.4 mM, C(GR) = 1.7 U, C(nanozyme) = 100 μg mL−1 | 112 | ||
GSH | 1.68 | 56.4 | 1.50 × 10−3 | C(H2O2) = 240 μM, C(NADPH) = 0.4 mM, C(GR) = 1.7 U, C(nanozyme) = 100 μg mL−1 | ||||
Pt | Janus BPQDs/Pt/AFSNs | H2O2 | 0.076 | 990 | pH = 7.4, C(GSH) = 2 mM, C(NADPH) = 0.4 mM, C(GR) = 1.7 U, C(nanozyme) = 10 μg mL−1 | 114 | ||
Janus Pt/AFSNs | H2O2 | 0.11 | 920 | |||||
Ru | RuO2 nanoparticles | H2O2 | 0.0172 | 47.0 | C(GSH) = 2.0 mM, C(NADPH) = 200 μM, C(GR) = 2.0 U, C(nanozyme) = 20 μg mL−1 | 115 | ||
GSH | 0.870 | 65.1 | C(H2O2) = 0.5 mM, C(NADPH) = 200 μM, C(GR) = 2.0 U, C(nanozyme) = 20 μg mL−1 | |||||
Se | Fe2NC@Se nanoparticles | GSH | 0.072 | 14.025 | 37 °C, pH = 7.4, C(H2O2) = 0.5 mM, C(NADPH) = 0.25 mM, C(GR) = 1.0 U mL−1 | 73 | ||
GO-Se nanocomposite | H2O2 | 0.04 | 30 | 25 °C, pH = 7.4, C(GSH) = 2 mM, C(NADPH) = 0.4 mM, C(GR) = 1.7 U, C(nanozyme) = 0.01 mg mL−1 | 43 | |||
GSH | 0.72 | 49.2 | 25 °C, pH = 7.4, C(H2O2) = 240 μM, C(NADPH) = 0.4 mM, C(GR) = 1.7 U, C(nanozyme) = 0.01 mg mL−1 | |||||
Lf-Au-Bi2Se3 nanodots | H2O2 | 0.61 | 1280 | 25 °C, pH = 7.4, C(GSH) = 2 mM, C(NADPH) = 200 μM, C(GR) = 1.7 U, C(nanozyme) = 10 μg mL−1 | 74 | |||
MSe nanoparticles | H2O2 | 0.15772 | 27.36 | 25 °C, pH = 7.4, C(GSH) = 2 mM, C(NADPH) = 0.4 mM, C(GR) = 1.7 U, C(nanozyme) = 20 μg mL−1 | 45 | |||
MSe-HA nanoparticles | H2O2 | 0.1598 | 26.81 | |||||
Se NPs | H2O2 | 0.20059 | 13.51 | |||||
PDASeCys | H2O2 | 1.37 | 87 | 25 °C, C(GSH) = 1.5 mM, C(NADPH) = 1.25 mM, C(GR) = 1 U, C(nanozyme) = 0.5 mg mL−1 | 78 | |||
GSH | 2.56 | 219.6 | 25 °C, C(H2O2) = 3 mM, C(NADPH) = 1.25 mM, C(GR) = 1 U, C(nanozyme) = 0.5 mg mL−1 | |||||
Se-DG | Se-DG-1 | H2O2 | 0.2398 ± 0.061 | 604.8 ± 85.32 | 24 °C, pH = 7, C(GSH) = 4000 μM, C(NADPH) = 100 μM, C(GR) = 1.02 U | 120 | ||
Se-DG-2 | H2O2 | 0.1448 ± 0.0334 | 499.2 ± 50.64 | |||||
Se-DG-6 | H2O2 | 0.0826 ± 0.0174 | 446.6 ± 32.82 | |||||
Se-DG-1 | GSH | 0.3482 ± 0.0216 | 241.8 ± 2.52 | 24 °C, pH = 7, C(H2O2) = 100 μM, C(NADPH) = 100 μM, C(GR) = 1.02 U | ||||
Se-DG-2 | GSH | 0.3654 ± 0.0263 | 249.6 ± 3.12 | |||||
Se-DG-6 | GSH | 0.7753 ± 0.028 | 332.4 ± 8.58 | |||||
(Sec-Arg-Gly-Asp -Cys)-modified Au nanoparticles | URGDC | H2O2 | 0.28 | 0.014 | 37 °C, pH = 7.0, C(GSH) = 1 mM, C(NADPH) = 0.25 mM, C(GR) = 1 U | 59 | ||
MPC 1 | H2O2 | 0.22 | 0.247 | |||||
MPC 2 | H2O2 | 0.18 | 0.261 | |||||
URGDC | GSH | 0.72 | 0.014 | 37 °C, pH = 7.0, C(H2O2) = 0.5 mM, C(NADPH) = 0.25 mM, C(GR) = 1 U | ||||
MPC 1 | GSH | 0.64 | 0.247 | |||||
MPC 2 | GSH | 0.32 | 0.261 | |||||
V | CuV2O6 | H2O2 | 0.02383 | 156.25 | 25 °C, pH = 7.4, C(GSH) = 2 mM, C(NADPH) = 0.2 mM, C(GR) = 1.7 U, C(nanozyme) = 20 μg mL−1 | 51 | ||
MIL-47(V)-Br | H2O2 | 0.022 | 96 | pH = 7.4, C(GSH) = 2 mM, C(NADPH) = 0.4 mM, C(GR) = 1.7 U | 48 | |||
MIL-47(V)-CH3 | H2O2 | 0.010 | 84 | |||||
MIL-47(V)- F | H2O2 | 0.007 | 108 | |||||
MIL-47(V)-H | H2O2 | 0.018 | 42 | |||||
MIL-47(V)-NH2 | H2O2 | 0.003 | 114 | |||||
MIL-47(V)-OH | H2O2 | 0.027 | 66 | |||||
MIL-47(V)-Br | GSH | 3.99 | 186 | pH = 7.4, C(H2O2) = 0.4 mM, C(NADPH) = 0.4 mM, C(GR) = 1.7 U | ||||
MIL-47(V)-CH3 | GSH | 1.71 | 150 | |||||
MIL-47(V)- F | GSH | 3.70 | 216 | |||||
MIL-47(V)-H | GSH | 2.67 | 84 | |||||
MIL-47(V)-NH2 | GSH | 2.85 | 210 | |||||
MIL-47(V)-OH | GSH | 3.18 | 96 | |||||
VN4 single-atom | H2O2 | 0.050 | 7.90 | 0.0535 | pH = 7.4, C(GSH) = 2 mM, C(NADPH) = 200 μM, C(GR) = 1.7 U·mL−1, C(nanozyme) = 3.8 μM | 83 | ||
GSH | 1.31 | 8.55 | 0.1667 | pH = 7.4, C(H2O2) = 0.2 mM, C(NADPH) = 200 μM, C(GR) = 1.7 U·mL−1, C(nanozyme) = 3.8 μM | ||||
V2O5 nanowires | H2O2 | 0.11 | 430 | 0.065 | 25 °C, pH = 7.4, C(GSH) = 2 mM, C(NADPH) = 0.4 mM, C(GR) = 1.7 U, C(nanozyme) = 0.020 mg mL−1 | 39 | ||
GSH | 2.22 | 830 | 25 °C, pH = 7.4, C(H2O2) = 240 μM, C(NADPH) = 0.4 mM, C(GR) = 1.7 U, C(nanozyme) = 0.020 mg mL−1 | |||||
V2O5 nanowires | H2O2 | 0.04 ± 0.00 | 192.31 ± 6.58 | 25 °C, pH = 7.4, C(GSH) = 2 mM, C(NADPH) = 0.2 mM, C(GR) = 1.7 U, C(nanozyme) = 20 μg mL−1 | 50 | |||
GSH | 1.28 ± 0.06 | 279.3 ± 18.2 | 25 °C, pH = 7.4, C(H2O2) = 200 μM, C(NADPH) = 0.2 mM, C(GR) = 1.7 U, C(nanozyme) = 20 μg mL−1 | |||||
V2O5 nanowires | H2O2 | 0.0444 ± 0.0017 | 192.3 ± 6.6 | 25 °C, pH = 7.4, C(GSH) = 2 mM, C(NADPH) = 0.2 mM, C(GR) = 1.7 U, C(nanozyme) = 20 ng μL−1 | 46 | |||
V2O5 nanosheets | H2O2 | 0.0573 ± 0.0038 | 233.1 ± 16.3 | |||||
V2O5 nanoflowers | H2O2 | 0.0925 ± 0.0034 | 340.1 ± 21.3 | |||||
V2O5 nanospheres | H2O2 | 0.1437 ± 0.0023 | 458.7 ± 19.6 | |||||
V2O5 nanowires | GSH | 1.28 ± 0.0612 | 279.3 ± 18.2 | 25 °C, pH = 7.4, C(H2O2) = 200 μM, C(NADPH) = 0.2 mM, C(GR) = 1.7 U, C(nanozyme) = 20 ng μL−1 | ||||
V2O5 nanosheets | GSH | 3.425 ± 0.1217 | 467.3 ± 55.8 | |||||
V2O5 nanoflowers | GSH | 1.671 ± 0.041 | 432.9 ± 18.1 | |||||
V2O5 nanospheres | GSH | 1.958 ± 0.0633 | 602.4 ± 40.3 | |||||
V2O5 ultrathin nanosheets | H2O2 | 0.1122 ± 0.0038 | 476.2 ± 34.1 | 25 °C, pH = 7.4, C(GSH) = 2 mM, C(NADPH) = 0.2 mM, C(GR) = 1.7 U, C(nanozyme) = 20 ng μL−1 | 122 | |||
V2O5@pDA@MnO2 | H2O2 | 0.16 | 88 | 25 °C, pH = 7.4, C(GSH) = 2 mM, C(NADPH) = 0.4 mM, C(GR) = 1.7 U, C(nanozyme) = 0.0125 mg mL−1 | 123 | |||
GSH | 7.2 | 190 | 25 °C, pH = 7.4, C(H2O2) = 240 μM, C(NADPH) = 0.4 mM, C(GR) = 1.7 U, C(nanozyme) = 0.0125 mg mL−1 | |||||
2D V2C MXenzyme | H2O2 | 10.86 | 9600 | C(GSH) = 8.4 mM, C(NADPH) = 300 μM | 47 | |||
Multiple active centres | ZIF-67/Cu0.76Co2.24O4 nanospheres | H2O2 | 0.549 | 0.045 | pH = 7.4, C(GSH) = 2 mM, C(NADPH) = 0.2 mM, C(GR) = 1.7 U, C(nanozyme) = 100 μg mL−1 | 126 | ||
GSH | 0.484 | 0.036 | pH = 7.4, C(H2O2) = 1 mM, C(NADPH) = 0.2 mM, C(GR) = 1.7 U, C(nanozyme) = 100 μg mL−1 | |||||
O-NZ | H2O2 | 0.8204 | 72.97 | 25 °C, pH = 7.4, C(GSH) = 2 mM, C(NADPH) = 200 μM, C(GR) = 1.7 U, C(nanozyme) = 2.5 μM | 127 | |||
GSH | 0.8688 | 18.49 | 25 °C, pH = 7.4, C(H2O2) = 200 μM, C(NADPH) = 200 μM, C(GR) = 1.7 U, C(nanozyme) = 2.5 μM |
Fig. 5 (A) An illustration of how UiO-66-Se is synthesized. Reprinted with permission from ref. 72. Copyright (2018) Springer Nature. (B) Schematic illustration of the synthesis of Fe2NC@Se. Adapted with permission from ref. 73. Copyright (2022) John Wiley and Sons. (C and D) Diagram illustrating GO-Se nanocomposite synthesis and GPx-like activity. Adapted with permission from ref. 43. Copyright (2017) Royal Society of Chemistry. |
Also, selenium nanoparticles (Se NPs) and metal selenides (e.g., Bi2Se3, MoSe2, and Mo3Se4) were used to mimic GPx.43,74–77 By adding ascorbic acid to a GO/selenium dioxide solution, Qu, Ren, and co-workers fabricated GO-Se nanocomposites in which Se NPs were well modified on the surface of GO. As demonstrated by the typical glutathione reductase (GR) coupled assay, Se NPs had GPx-like activity, and GO's large surface area and rapidly transferring electrons could enhance the activity (Fig. 5C and D).43 As well as GO, some surface coatings and modifications were also used to improve the dispersion and stability, such as polyvinylpyrrolidone (PVP),75,76 polydopamine (pDA),44,78 hyaluronic acid (HA),45 and polysaccharides.79 The coating or modification would even serve as a synergistic antioxidant. The synergetic antioxidant machinery in organisms, for instance, inspired Qu, Ren, and co-workers to develop pDA coated Se NPs, which acted as non-enzymatic antioxidants and antioxidant enzymes, respectively. By exploiting the O2˙− and ˙OH scavenging properties of pDA, Se@pDA nanocomposites demonstrated more potent antioxidant capacity. An application of this synergistic antioxidant system will be described in the Application section.44
Fig. 6 (A and B) GPx-like activities of different V2O5. Adapted with permission from ref. 46. Copyright (2018) John Wiley and Sons. (C and D) A schematic of the synthesis of MIL-47(V)-X MOFs and their GPx-like activities. Adapted with permission from ref. 48. Copyright (2021) John Wiley and Sons. |
Following these pioneering studies, other V-based nanomaterials, such as V2C MXenzyme,47,80 orthovanadate NP,81,82 MIL-47(V) MOF,48 VN4 single-atom,83 MnV2O6,51 ZnV2O6,51 and CuV2O6,51 also exhibited excellent GPx-like activities. As shown in Fig. 6C, a MIL-47(V)-H MOF nanozyme mimicking GPx was synthesized by coordinating the 1,4-benzenedicarboxylicacid ligand with V3+ and modulated its GPx-like activity by ligand engineering.48 In particular, F, Br, NH2, CH3, and OH were substituted for H in the benzenedicarboxylicacid ligand to create MIL-47(V)-X MOFs (X = F, Br, NH2, CH3, OH, and H). Their GPx-like activities were then investigated and are shown in Fig. 6D. These isostructural MOFs all exhibited GPx-like activity, and the substitutions enhanced the catalytic activity, with MIL-47(V)-NH2 being the most active. In addition, a control experiment using only ligands was conducted under identical conditions to clarify why MIL-47(V)-X MOFs differ in their catalytic activity. It is unlikely that ligands alone would have GPx-like activities, ruling out the possibility that their intrinsic catalytic activity could influence the results. As a result of the X-ray photoelectron spectroscopy results and subsequent analysis, the electronic impact of substitution on the metal nodes was found to be responsible for the difference in catalytic activity between the MIL-47(V)-X MOFs.
Fig. 7 (A and B) An illustration of the valence-engineering strategy for modulating the GPx-like activity of Li-doped samples. Adapted with permission from ref. 89. Copyright (2022) John Wiley and Sons. (C) TEM images of Mn3O4 with different morphologies and their GPx-like activities. Adapted with permission from ref. 52. Copyright (2018) John Wiley and Sons. |
Besides valences, other factors may play an important role in modulating the GPx-like activities of manganese oxide nanozymes. For example, Mn3O4 with different morphologies, including cubes, polyhedra, hexagonal plates, flakes, and flowers, exhibited different GPx-like activities, following the order: flowers > flakes > hexagonal plates ≈ polyhedra ≈ cubes (Fig. 7C).52 The higher activity of flowers and flakes was due to the more active sites and higher electrical conductivity than the other three morphologies. Another study conducted by Sun and co-workers demonstrated that the GPx-like activity of Mn3O4 nanoparticles was related to the crystal facet.87 They synthesized three morphologies, including nanospheres, nanocubes, and nanorods, that were encircled by the {101}, {200}, and {103} crystal facets. The results demonstrated that the GPx-like activity of {103}-faceted nanorods was superior to that of {101}-faceted nanospheres and {200}-faceted nanocubes. This is attributed to the increased reaction energy of the Mn3O4 nanorod surface, which had superior ROS scavenging capability.
Fig. 8 (A and B) An illustration of the synthesis of CuxO-Phe and its morphology. (C and D) GPx-like activities of various CuxOs and other metal oxides. Adapted with permission from ref. 90. Copyright (2019) American Chemical Society. (E and F) Schematic illustration of different metal-containing COFs and their GPx-like activities. Adapted with permission from ref. 98. Copyright (2022) John Wiley and Sons. |
Furthermore, other Cu-containing nanomaterials, such as MOF nanodots,95 PtCu3 nanocages,53 Cu nanoclusters,96 Cu-doped polypyrroles,54 and covalent organic frameworks (COFs),97,98 exhibited excellent GPx-like properties. In a variety of fields, COFs have attracted widespread attention due to their inherent porosity, high crystallinity, wide functionality, and strong biocompatibility. It is also possible to make GPx mimics by replacing N-doped carbon with COF, i.e. by decorating Cu2+ into COF scaffolds. As shown in Fig. 8E, COF-909-Cu was recently synthesized by Sun, Deng, and co-workers through decorating Cu2+ into COF-909 via a post-modification method.98 In comparison to COF-909, COF-909-Ni, and COF-909-Fe, COF-909-Cu exhibited the highest GPx-like activity (Fig. 8F).
Fig. 9 (A and B) Successful synthesis of CeO2 and Pt/CeO2, as well as their GPx-like activities. Adapted with permission from ref. 105. Copyright (2019) American Chemical Society. (C) GPx-like activities of CeO2 and different metal-doped CeO2. (D) GPx-like activities of Cr/CeO2 nanozymes with varying doping concentrations. Adapted with permission from ref. 106. Copyright (2021) Ivyspring International Publisher. |
Fig. 10 (A) The SEM image of HEK293T cells with V2O5-untreated (UT), as well as V2O5-treated for 0 and 2 h. (B and C) The H2O2-scavaging ability of V2O5 under different treatments. Adapted with permission from ref. 39. Copyright (2014) Springer Nature. (D and E) MnO2 NPs with multienzyme antioxidant activities and the cell protection effect and mechanism, as well as western blot expression of apoptosis-related proteins. Adapted with permission from ref. 88. Copyright (2022) American Chemical Society. |
In addition to scavenging ROS, producing nitric oxide (NO), which may be depleted by ROS, is also essential for maintaining endothelial cell function. In a recent study, ZnO with GPx-like activity was found to decompose S-nitrosoglutathione catalytically to produce NO. NO-sensitive electrodes were used to characterize the production of NO, and ZnO was found to increase NO production sixfold and maintain this activity for half a year. Meanwhile, due to its glycosidase-like activity, ZnO could promote NO production from exogenous β-gal-NONOate, potentially contributing to sustained NO supply. Taking advantage of the NO-generating function, ZnO showed great promise in treating endothelial dysfunction and its associated cardiovascular disease.124
Fig. 11 (A) An illustration of V2O5@pDA@MnO2 scavenging ROS. Adapted with permission from ref. 123. Copyright (2016) John Wiley and Sons. (B) Schematic illustration of CaPB nanozymes in the treatment of AKI. Adapted with permission from ref. 111. Copyright (2021) John Wiley and Sons. |
Aside from ROS, excessive reactive nitrogen species (RNS) like ˙NO and peroxynitrite (ONOO−) were also shown to contribute to inflammation. Sepsis, for example, is a condition that results in high morbidity and mortality due to the immoderate production of reactive oxygen and nitrogen species (RONS). At the beginning, the immune system was activated by pathogens and generated excessive H2O2 and HOCl in plasma. Due to its unique capability, H2O2 could enter cells easily and then not only participated in secondary reactions to produce ˙OH but also activated nuclear transcription factor-κB, which stimulated the production of ˙NO, O2˙−, and ONOO−. It has been discovered that Co single-atom catalysts (Co/PMCS) are capable of multiple enzyme-like activities that can be used to scavenge RONS in order to achieve the desirable therapeutic efficacy. Through pyrolysis, Co/PMCS was produced, which mimicked SOD, GPx, and CAT to eliminate ROS, and also removed ˙NO through the unoccupied Co-porphyrin center. According to in vivo experiments, Co/PMCS could significantly reduce RONS and pro-inflammatory cytokines in tissues, thereby mitigating inflammation. Treatment with Co/PMCS resulted in an increase in the 14-day survival rate of sepsis mice from 10% to 60%. Since H2O2 was critical in the redox mechanisms of sepsis, the GPx-like activity of Co/PMCS was possible to play a vital role in achieving the desirable therapy efficacy.107 In addition, AKI and acute pancreatitis were associated with the over-production of RONS. As shown in Fig. 11B, ultrasmall CaPB synthesized with excellent RONS scavenging ability could cross the renal filtration threshold and prevent renal damage caused by AKI. The treatment of CaPB reduced the expression of pro-inflammatory cytokines TNF-α and IL-1β while enhancing the expression of anti-inflammatory cytokines IL-10. Furthermore, CaPB could inhibit ferroptosis through elevated expression of GPx4, showing potential for clinical application.111
H2O2 was considered to be among the most harmful ROS due to its longer life than O2˙−, its ease of diffusion across lipid membranes, as well as its precursor role in creating the highly harmful ˙OH. Therefore, several studies have focused on using GPx-like activity alone as an anti-inflammatory agent. Se NPs with GPx-like activity, for example, have been shown to effectively reduce inflammation. Moreover, their antioxidant coatings, such as pDA and hyaluronic acid, enhanced Se NPs’ ROS scavenging ability.44,45 In contrast to Se NPs, Wei and co-workers synthesized MIL-47(V)-X MOF nanozymes with modulated GPx-like activity using a ligand engineering strategy (seeing section 2.2). The most active MIL-47(V)-NH2 was chosen for further anti-inflammation study, and the least active MIL-47(V)-H was also studied for comparison. As shown in Fig. 12A, PMA-induced ear inflammation was successfully developed with the ear swollen, and MIL-47(V) treatment alleviated this phenomenon. An additional dextran sulfate sodium (DSS)-induced colitis model was also treated with MIL-47(V) to demonstrate broad-spectrum anti-inflammation (Fig. 12B). With both recovered colon length and decreased inflammatory cytokines TNF-α and IL-1β (Fig. 12C and D), MIL-47(V) showed satisfactory therapeutic efficacy, even better than commercial drugs 5-aminosalicylic acid (5-ASA). In both models, MIL-47(V)-NH2 with higher GPx-like activity demonstrated superior performance over MIL-47(V)-H, suggesting that the principles of structure–activity relationships might also guide the design of nanozyme-based therapeutics.48
Fig. 12 (A) A comparison of hematoxylin and eosin-stained images of ear tissue with various treatments. (B) Colon images after various treatments. (C) TNF-α and (D) IL-1β levels in colon homogenates after different treatments. Adapted with permission from ref. 48. Copyright (2021) John Wiley and Sons. |
Fig. 13 (A) Principal component analysis considering all motor phenotypes. (B) Anxiety-like behavior evaluated by the open field test and elevated plus maze tests. (C) Depression-like behavior evaluated by immobility time, climbing time, swimming time, and latency to first immobility. (D) Object recognition behavior evaluated by the novel object recognition test, including the experimental setup, the movement pattern of the mice, and the novel object exploration time compared to the familiar one. Adapted with permission from ref. 49. Copyright (2021) John Wiley and Sons. |
In contrast to HD, other neurodegenerative diseases lack direct links to GPx; therefore, GPx-like activity is usually combined with other antioxidant enzyme-like activities to achieve more comprehensive ROS scavenging. For example, CuxO nanozymes with SOD-, CAT-, and GPx-like activities were used in PD treatment to efficiently scavenge ROS, inhibit neurodegeneration, and improve memory.90 An evaluation of the effectiveness of adding GPx-like activity concluded that adding extra GPx-like activity in combination with SOD- and CAT-like activities would increase neuronal cell activity by 30%, thereby improving the therapeutic efficacy.78 If it comes to treating traumatic brain injury (TBI), the elimination of excessive O2˙− and ˙NO is more important. To treat TBI, an oligomeric nanozyme (O-NZ) based on N-doped graphite with active surface groups was synthesized. Carbogenic cores exhibited GPx- and SOD-like activities, and ultrafast electron transfer between the core and surface-active groups facilitated the high clearance of ˙OH, O2˙−, ˙NO, and ONOO−. As a result of O-NZ's excellent ability to eliminate RONS, the survival rate of mice with acute brain trauma improved from 50 to 90% after one month, and long-term cognition was improved significantly.127
Fig. 14 (A) Induction of autophagy-lysosome pathways in tumor therapy by the Fe-CDs@Ang enzymatic cascade. (B) Bioluminescence images of mice with gliomas. Adapted with permission from ref. 110. Copyright (2022) Elsevier. (C) An illustration of MnFe2O4@MOF for tumor therapy. Reprinted with permission from ref. 56. Copyright (2019) John Wiley and Sons. (D) An illustration of PtCu3 nanocages for tumor therapy. Adapted with permission from ref. 53. Copyright (2019) John Wiley and Sons. |
As well as enhancing the effectiveness of ROS produced by POD- and OXD-like nanozymes, GPx-like nanozymes can also improve the efficacy of other therapeutic strategies. For example, MnFe2O4@MOF nanozymes with CAT- and GPx-like activities were synthesized for enhanced photodynamic therapy (Fig. 14C). As a photo-sensitizer, MnFe2O4@MOF converted O2 into 1O2 under laser irradiation, thus resulting in obvious oxidative stress. The CAT-like activity could overcome hypoxia by continuously producing O2 from endogenous H2O2. The GPx-like activity could deplete overexpressed GSH and generate a better photodynamic effect. Further experiments showed that the consumption of GSH by MnFe2O4@MOF itself effectively decreases 1O2 depletion.56 A similar effect was observed when PtCu3 nanocages with GPx-like activity were used to consume GSH to enhance sonodynamic therapy, in which 1O2 and ˙OH were generated under ultrasound. An in vitro experiment confirmed that GSH could be continuously depleted via a circular reaction in the presence of H2O2, thus weakening the capacity of tumor cells scavenging ROS by GSH and enhancing sonotoxicity (Fig. 14D).53 Likewise, incorporating GPx- and CAT-like activities into chemotherapeutic drugs would provide a superior anticancer effect.55 The GPx-like activity could deplete GSH and minimize GSH-medicated cisplatin detoxification. With nanomaterials’ unique physicochemical properties, enzyme-like activities, such as POD, could be accelerated via photothermal effects. COF-909-Cu, for instance, possessed photothermal properties that enhanced the production of ˙OH and contributed to the chemodynamic therapy efficiency.98 In another example, a simple SnFe2O4 nanozyme was used to combine photothermal, photodynamic, and chemodynamic therapies, resulting in significant tumor growth inhibition.57 In both cases, GPx-like activity was responsible for reducing GSH levels.
Aside from therapeutic uses, GPx-like nanozymes could also be used for detection and epoxidation. A Fe2+ sensor based on GPx-like CuO-POM nanozymes was developed by Liu, Zhu, and co-workers. As o-phthalaldehyde reacted with GSH, a specific fluorescent isoindole derivative was formed, while the GPx-like activity consumed GSH as well, thus decreasing fluorescence. Since Fe2+ would recover fluorescence, a fluorometric sensor was constructed with a detection limit as low as 8.0 nM.94 In addition, the selenium-doped graphite carbocatalyst also demonstrated GPx-like activity in epoxidating multiple aromatic and aliphatic alkene compounds, with ∼90% conversion rate and ∼75% epoxide yield.120
(1) There is a lack of understanding of the structure–activity relationship and rational design methods of GPx-like nanozymes. GPx-like nanozyme research is currently largely based on screening and random synthesis. Meanwhile, the GPx-like activity is discovered along with other enzyme-like activities, hindering further understanding of the origin of GPx-like activity and its influencing factors. It is crucial for the success of future research on GPx-like nanozymes to have detailed findings regarding the structure–activity relationship. GPx-like nanozyme databases can be constructed using theoretical calculations, machine learning, and other methods along with experimental exploration to understand the relationship between enzyme activity and material properties. By doing so, it will be possible to design and develop highly active GPx-like nanozymes in a rational manner.
(2) There is not enough systematic analysis and in-depth research on GPx-like catalytic mechanisms and kinetics. To better understand catalytic reactions, it is essential to begin with understanding the internal mechanisms of GPx-like nanozymes. However, few studies have been conducted to fully understand the dynamics and mechanism of GPx-like nanozymes. The identification and characterization of active components, changes in the valence state of active site elements, and identification of intermediates in catalytic processes have not been thoroughly investigated. To explore more detailed catalytic mechanisms, practical theoretical computational methods as well as experimental techniques are needed, which is of great significance for future design and rational application.
(3) Research using GPx-like activity alone for biomedical treatment is relatively limited. A majority of bioapplications rely on multiple enzyme-like activities, with GPx-like activity only acting as an auxiliary component. It has been shown that, however, in some bioapplications, such as anti-inflammation, nanozymes can still exert the necessary effect even when there is merely GPx-like activity. It may be possible, therefore, to conduct more in-depth research on biological mechanisms and to understand the differences between multienzyme-like activities and only GPx-like activity in disease therapy, in addition to treating with only GPx-like nanozymes in the future. GPx-like nanozymes will benefit from these in-depth investigations in the future when developed for biomedical applications.
(4) Despite the success of GPx-like nanozymes in biomedical applications, their biosafety, potential toxicity, and pharmacokinetics remain the main obstacles to clinical application. Currently, nanozymes’ overall toxicity assessment, in vivo applicability, in vivo metabolic issues, and clinical adverse effects have not been sufficiently investigated. More attention should be paid to future research that encompasses everything from therapeutic modalities to toxicity assessments in pre-clinical trials. In order to improve the biosafety and increase the clinical translation potential of nanozymes, future research should correlate nanomaterial features and manufacturing procedures with their safety properties.
Footnote |
† These authors contributed to this work equally. |
This journal is © The Royal Society of Chemistry 2023 |