Journal Home > Volume 16 , Issue 1

In the past decade, nanozymes - a unique class of nanomaterials with inherent enzyme-mimetic properties - have fascinated researchers, revealing unexpected enzyme-like activity of nanomaterials previously considered biologically inert. In particular, as metal-free catalyst for biological processes, carbon-based nanozymes have grown in popularity due to their exceptional physical and chemical characteristics. So far, a variety of carbon-based nanozymes with various structures such as fullerene, graphene oxide, carbon dot, carbon nanotube, and carbon nanosphere have been reported possessing a wide range of enzyme-like properties. However, the structure-activity relationship of the carbon-based nanozymes have not yet been comprehensively discussed. In this review, we thoroughly examine the recent findings on the structure-activity connection of carbon nanozymes, in an effort to comprehend the underlying mechanism of carbon nanozymes and throw light on the future direction of the systematic design and construction of functionally specific carbon nanozymes. We also will address the broad range of applications of carbon nanozymes from in vitro detection to replacing specific enzymes in living systems.


menu
Abstract
Full text
Outline
About this article

Carbon-based Nanozymes: How Structure Affects Performance

Show Author's information Jiuyang He1( )Yinyin Hou2,4Zixia Zhang2,4Junying Zhang1Xiyun Yan2,3,4Kelong Fan2,3,4( )Minmin Liang1( )
Experimental Center of Advanced Materials, School of Materials Science & Engineering, Beijing Institute of Technology, Beijing 100081, China
CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
Nanozyme Medical Center, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450052, China
University of Chinese Academy of Sciences, Beijing 101408, China

Abstract

In the past decade, nanozymes - a unique class of nanomaterials with inherent enzyme-mimetic properties - have fascinated researchers, revealing unexpected enzyme-like activity of nanomaterials previously considered biologically inert. In particular, as metal-free catalyst for biological processes, carbon-based nanozymes have grown in popularity due to their exceptional physical and chemical characteristics. So far, a variety of carbon-based nanozymes with various structures such as fullerene, graphene oxide, carbon dot, carbon nanotube, and carbon nanosphere have been reported possessing a wide range of enzyme-like properties. However, the structure-activity relationship of the carbon-based nanozymes have not yet been comprehensively discussed. In this review, we thoroughly examine the recent findings on the structure-activity connection of carbon nanozymes, in an effort to comprehend the underlying mechanism of carbon nanozymes and throw light on the future direction of the systematic design and construction of functionally specific carbon nanozymes. We also will address the broad range of applications of carbon nanozymes from in vitro detection to replacing specific enzymes in living systems.

Keywords: biomedical applications, structure-activity relationship, enzyme-like activity, rational design, carbon nanozyme

References(121)

[1]

H. Wei, E.K. Wang. Nanomaterials with enzyme-like characteristics (nanozymes): Next-generation artificial enzymes. Chemical Society Reviews, 2013, 42: 6060−6093. https://doi.org/10.1039/C3CS35486E

[2]
X.Y. Yan, L.Z. Gao. Nanozymology: In: Nanozymology. Nanostructure Science and Technology. Springer: Singapore, 2020.
DOI
[3]

M.M. Liang, X.Y. Yan. Nanozymes: From new concepts, mechanisms, and standards to applications. Accounts of Chemical Research, 2019, 52(8): 2190−2200. https://doi.org/10.1021/acs.accounts.9b00140

[4]

L.Z. Gao, J. Zhuang, L. Nie, et al. Intrinsic peroxidase-like activity of ferromagnetic nanoparticles. Nature Nanotechnology, 2007, 2(9): 577−583. https://doi.org/10.1038/nnano.2007.260

[5]

Y.J. Chen, P.X. Wang, H.G. Hao, et al. Thermal atomization of platinum nanoparticles into single atoms: An effective strategy for engineering high-performance nanozymes. Journal of the American Chemical Society, 2021, 143(44): 18643−18651. https://doi.org/10.1021/jacs.1c08581

[6]

S.F. Ji, B. Jiang, H.G. Hao, et al. Matching the kinetics of natural enzymes with a single-atom iron nanozyme. Nature Catalysis, 2021, 4(5): 407−417. https://doi.org/10.1038/s41929-021-00609-x

[7]

Y.J. Chen, B. Jiang, H.G. Hao, et al. Atomic-level regulation of cobalt single-atom nanozymes: Engineering high-efficiency catalase mimics. Angewandte Chemie International Edition, 2023, 62(19): e202301879. https://doi.org/10.1002/anie.202301879

[8]

C.Y. Hong, X.G. Meng, J.Y. He, et al. Nanozyme: A promising tool from clinical diagnosis and environmental monitoring to wastewater treatment. Particuology, 2022, 71: 90−107. https://doi.org/10.1016/j.partic.2022.02.001

[9]
P.X. Wang, S.L. Liu, M.X. Hu, et al. Peroxidase-like nanozymes induce a novel form of cell death and inhibit tumor growth in vivo. Advanced Functional Materials, 2020, 30(21): 2000647.
DOI
[10]

H.W. Kroto, J.R. Heath, S.C. O’Brien, et al. C60: Buckminsterfullerene. Nature, 1985, 318(6042): 162−163. https://doi.org/10.1038/318162a0

[11]

S. Iijima. Helical microtubules of graphitic carbon. Nature, 1991, 354(6348): 56−58. https://doi.org/10.1038/354056a0

[12]

K.S. Novoselov, A.K. Geim, S.V. Morozov, et al. Electric field effect in atomically thin carbon films. Science, 2004, 306(5696): 666−669. https://doi.org/10.1126/science.1102896

[13]

A. Ruggiero, C.H. Villa, E. Bander, et al. Paradoxical glomerular filtration of carbon nanotubes. Proceedings of the National Academy of Sciences of the United States of America, 2010, 107(27): 12369−12374. https://doi.org/10.1073/pnas.0913667107

[14]

H. Tokuyama, S. Yamago, E. Nakamura, et al. Photoinduced biochemical activity of fullerene carboxylic acid. Journal of the American Chemical Society, 1993, 115(17): 7918−7919. https://doi.org/10.1021/ja00070a064

[15]

X.L. Zuo, C. Peng, Q. Huang, et al. Design of a carbon nanotube/magnetic nanoparticle-based peroxidase-like nano complex and its application for highly efficient catalytic oxidation of phenols. Nano Research, 2009, 2(8): 617−623. https://doi.org/10.1007/s12274-009-9062-3

[16]

Yuqing, Miao,. Layer-by-layer assembled hybrid film of carbon nanotubes/iron oxide nanocrystals for reagentless electrochemical detection of H2O2. Sensors and Actuators B:Chemical, 2009, 138(1): 182−188. https://doi.org/10.1016/j.snb.2008.12.045

[17]

Y.J. Song, K.G. Qu, C. Xu, et al. Visual and quantitative detection of copper ions using magnetic silica nanoparticles clicked on multiwalled carbon nanotubes. Chemical Communications, 2010, 46(35): 6572−6574. https://doi.org/10.1039/c0cc01593h

[18]

Y.J. Song, K.G. Qu, C. Zhao, et al. Graphene oxide: Intrinsic peroxidase catalytic activity and its application to glucose detection. Advanced Materials, 2010, 22(19): 2206−2210. https://doi.org/10.1002/adma.200903783

[19]

H.J. Sun, Y. Zhou, J.S. Ren, et al. Carbon nanozymes: Enzymatic properties, catalytic mechanism, and applications. Angewandte Chemie International Edition, 2018, 57(30): 9224−9237. https://doi.org/10.1002/anie.201712469

[20]

H. Ding, B. Hu, B. Zhang, et al. Carbon-based nanozymes for biomedical applications. Nano Research, 2021, 14(3): 570−583. https://doi.org/10.1007/s12274-020-3053-9

[21]

C. Cha, S.R. Shin, N. Annabi, et al. Carbon-based nanomaterials: Multifunctional materials for biomedical engineering. ACS Nano, 2013, 7(4): 2891−2897. https://doi.org/10.1021/nn401196a

[22]

P.J. Krusic, E. Wasserman, P.N. Keizer, et al. Radical reactions of C60. Science, 1991, 254(5035): 1183−1185. https://doi.org/10.1126/science.254.5035.1183

[23]

L.L. Dugan, J.K. Gabrielsen, S.P. Yu, et al. Buckminsterfullerenol free radical scavengers reduce excitotoxic and apoptotic death of cultured cortical neurons. Neurobiology of Disease, 1996, 3(2): 129−135. https://doi.org/10.1006/nbdi.1996.0013

[24]

L.L. Dugan, D.M. Turetsky, C. Du, et al. Carboxyfullerenes as neuroprotective agents. Proceedings of the National Academy of Sciences of the United States of America, 1997, 94(17): 9434−9439. https://doi.org/10.1073/pnas.94.17.9434

[25]

L.L. Dugan, E.G. Lovett, K.L. Quick, et al. Fullerene-based antioxidants and neurodegenerative disorders. Parkinsonism &Related Disorders, 2001, 7(3): 243−246. https://doi.org/10.1016/S1353-8020(00)00064-X

[26]

S.S. Ali, J.I. Hardt, K.L. Quick, et al. A biologically effective fullerene (C60) derivative with superoxide dismutase mimetic properties. Free Radical Biology and Medicine, 2004, 37(8): 1191−1202. https://doi.org/10.1016/j.freeradbiomed.2004.07.002

[27]

R.M. Li, M.M. Zhen, M.R. Guan, et al. A novel glucose colorimetric sensor based on intrinsic peroxidase-like activity of C60-carboxyfullerenes. Biosensors and Bioelectronics, 2013, 47: 502−507. https://doi.org/10.1016/j.bios.2013.03.057

[28]

A.S. Boutorine, M. Takasugi, C. Hélène, et al. Fullerene–oligonucleotide conjugates: Photoinduced sequence-specific DNA cleavage. Angewandte Chemie International Edition in English, 1995, 33(23-24): 2462−2465. https://doi.org/10.1002/anie.199424621

[29]
A.K. Geim, Graphene: status and prospects. Science, 2009, 324(5934): 1530–1534.
DOI
[30]

C.L. Su, M. Acik, K. Takai, et al. Probing the catalytic activity of porous graphene oxide and the origin of this behaviour. Nature Communications, 2012, 3(1): 1298. https://doi.org/10.1038/ncomms2315

[31]
N. Song, Y. Yu, Y. Zhang, et al. Bioinspired hierarchical self‐assembled nanozyme for efficient antibacterial treatment. Advanced Materials, 2023: 2210455.
DOI
[32]

Y.H. Hu, Y.Y. Zhu, F. Muhanmmad, et al. Nitrogen-Doped Carbon Nanomaterials as Highly Active and Specific Peroxidase Mimics. Chemistry of Materials, 2018, 30(18): 6431−6439. https://doi.org/10.1021/acs.chemmater.8b02726

[33]

S.Y. Luo, M. Sha, F. Tian, et al. Nitrogen and boron co-doped graphene nanoribbons as peroxidase-mimicking nanozymes for enhanced biosensing. Chinese Chemical Letters, 2022, 33(1): 344−348. https://doi.org/10.1016/j.cclet.2021.06.018

[34]

Y.Y. Huang, C.Q. Liu, F. Pu, et al. A GO-Se nanocomposite as an antioxidant nanozyme for cytoprotection. Chemical Communications, 2017, 53: 3082−3085. https://doi.org/10.1039/C7CC00045F

[35]

H.T. Li, Z.K. Kang, Y. Liu, et al. Carbon nanodots: synthesis, properties and applications. Journal of Materials Chemistry, 2012, 22(46): 24230. https://doi.org/10.1039/C2JM34690G

[36]

X.Y. Xu, R. Ray, Y.L. Gu, et al. Electrophoretic analysis and purification of fluorescent single-walled carbon nanotube fragments. Journal of the American Chemical Society, 2004, 126(40): 12736−12737. https://doi.org/10.1021/ja040082h

[37]

H. Ding, Y.J. Cai, L.Z. Gao, et al. Exosome-like nanozyme vesicles for H2O2-responsive catalytic photoacoustic imaging of xenograft nasopharyngeal carcinoma. Nano Letters, 2019, 19(1): 203−209. https://doi.org/10.1021/acs.nanolett.8b03709

[38]

S.Y. Lim, W. Shen, Z.Q. Gao. Carbon quantum dots and their applications. Chemical Society Reviews, 2015, 44(1): 362−381. https://doi.org/10.1039/c4cs00269e

[39]

F.L. Yuan, S.H. Li, Z.T. Fan, et al. Shining carbon dots: synthesis and biomedical and optoelectronic applications. Nano Today, 2016, 11(5): 565−586. https://doi.org/10.1016/j.nantod.2016.08.006

[40]

C.Q. Ding, A.W. Zhu, Y. Tian. Functional surface engineering of C-dots for fluorescent biosensing and in vivo bioimaging. Account of Chemical Research, 2014, 47(1): 20−30. https://doi.org/10.1021/ar400023s

[41]

J.C.G. Esteves da Silva, H.M.R. Goncalves. Analytical and bioanalytical applications of carbon dots. Trends in Analytical Chemistry, 2011, 30(8): 1327−1336. https://doi.org/10.1016/j.trac.2011.04.009

[42]

H. Ding, S.B. Yu, J.S. Wei, et al. Full-color light-emitting carbon dots with a surface-state-controlled luminescence mechanism. ACS Nano, 2016, 10(1): 484−491. https://doi.org/10.1021/acsnano.5b05406

[43]

G.S. Hong, S. Diao, A.L. Antaris, et al. Carbon nanomaterials for biological imaging and nanomedicinal therapy. Chemical Reviews, 2015, 115(19): 10816−10906. https://doi.org/10.1021/acs.chemrev.5b00008

[44]
M. Bacon, S.J. Bradley, T. Nann, Graphene quantum dots. Particle & Particle Systems Characterization, 2014, 31(4): 415–428.
DOI
[45]

B.K. Walther, C.Z. Dinu, D.M. Guldi, et al. Nanobiosensing with graphene and carbon quantum dots: Recent advances. Materials Today, 2020, 39: 23−46. https://doi.org/10.1039/C2CC00110A

[46]

J.H. Shen, Y.H. Zhu, X.L. Yang, et al. Graphene quantum dots: Emergent nanolights for bioimaging, sensors, catalysis and photovoltaic devices. Chemical Communications, 2012, 48(31): 3686−3699. https://doi.org/10.1039/c2cc00110a

[47]

K. Hola, Y. Zhang, Y. Wang, et al. Carbon dots—Emerging light emitters for bioimaging, cancer therapy and optoelectronics. Nano Today, 2014, 9(5): 590−603. https://doi.org/10.1016/j.nantod.2014.09.004

[48]

H.J. Sun, L. Wu, W.L. Wei, et al. Recent advances in graphene quantum dots for sensing. Materials Today, 2013, 16(11): 433−442. https://doi.org/10.1016/j.mattod.2013.10.020

[49]

J. Peng, W. Gao, B.K. Gupta, et al. Graphene quantum dots derived from carbon fibers. Nano Letters, 2012, 12(2): 844−849. https://doi.org/10.1021/nl2038979

[50]

Q. Liu, B.D. Guo, Z.Y. Rao, et al. Strong two-photon-induced fluorescence from photostable, biocompatible nitrogen-doped graphene quantum dots for cellular and deep-tissue imaging. Nano Letters, 2013, 13(6): 2436−2441. https://doi.org/10.1021/nl400368v

[51]

H.J. Sun, N. Gao, K. Dong, et al. Graphene quantum dots-band-aids used for wound disinfection. Acs Nano, 2014, 8(6): 6202−6210. https://doi.org/10.1021/nn501640q

[52]

J.C. Ge, M.H. Lan, B.J. Zhou, et al. A graphene quantum dot photodynamic therapy agent with high singlet oxygen generation. Nature Communications, 2014, 5: 4596. https://doi.org/10.1038/ncomms5596

[53]

Y. Zhang, C.Y. Wu, X.J. Zhou, et al. Graphene quantum dots/gold electrode and its application in living cell H2O2 detection. Nanoscale, 2013, 5(5): 1816−1819. https://doi.org/10.1039/C3NR33954H

[54]

J.Y. Zhang, X.M. Lu, D.D. Tang, et al. Phosphorescent carbon dots for highly efficient oxygen photosensitization and as photo-oxidative nanozymes. ACS Applied Materials &Interfaces, 2018, 10(47): 40808−40814. https://doi.org/10.1021/acsami.8b15318

[55]

J.Y. Zhang, Y. Lin, S.H. Wu, et al. Self-photo-oxidation for extending visible light absorption of carbon dots and oxidase-like activity. Carbon, 2021, 182: 537−544. https://doi.org/10.1016/j.carbon.2021.06.053

[56]

D.D. Zhang, Q.Y. Han, W.D. Liu, et al. Au-decorated N-rich carbon dots as peroxidase mimics for the detection of acetylcholinesterase activity. ACS Applied Nano Materials, 2022, 5(2): 1958−1965. https://doi.org/10.1021/acsanm.1c03613

[57]

D.Z. Yang, Q.L. Li, Q. Zhang, et al. A multifunctional nanozyme-based enhanced system for tert-butyl hydroquinone assay by surface-enhanced Raman scattering. Microchimica Acta, 2022, 189(1): 1−9. https://doi.org/10.1007/s00604-021-05135-y

[58]

Y.J. Song, X.H. Wang, C. Zhao, et al. Label-free colorimetric detection of single nucleotide polymorphism by using single-walled carbon nanotube intrinsic peroxidase-like activity. Chemistry – A European Journal, 2010, 16(12): 3551−3865. https://doi.org/10.1002/chem.200902643

[59]

H. Wang, P.H. Li, D.Q. Zhang, et al. Unraveling the enzymatic activity of oxygenated carbon nanotubes and their application in the treatment of bacterial infections. Nano Letters, 2018, 18(6): 3344−3351. https://doi.org/10.1021/acs.nanolett.7b05095

[60]

X.M. Dang, H.M. Zhao. Bimetallic Fe/Mn metal-organic-frameworks and Au nanoparticles anchored carbon nanotubes as a peroxidase-like detection platform with increased active sites and enhanced electron transfer. Talanta, 2020, 210: 120678. https://doi.org/10.1016/j.talanta.2019.120678

[61]

H. Wang, H. Jiang, S. Wang, et al. Fe3O4–MWCNT magnetic nanocomposites as efficient peroxidase mimic catalysts in a Fenton-like reaction for water purification without pH limitation. Royal Socirty of Chemistey Advances, 2014, 4: 45809−45815. https://doi.org/10.1039/C4RA07327D

[62]
P. Gayathri, P.A.S. Kumar, An iron impurity in multiwalled carbon nanotube complexes with chitosan that biomimics the heme-peroxidase function. Chemistry – A European Journal, 2013. 19(50): 17103–17112.
DOI
[63]

M.H. Yeh, L.Y. Lin, C.L. Sun, et al. Multiwalled carbon nanotube@reduced graphene oxide nanoribbon as the counter electrode for dye-sensitized solar cells. The Journal of Physical Chemistry C, 2014, 118(30): 16626−16634. https://doi.org/10.1021/jp412542d

[64]
K.L. Fan, J.Q. Xi, L.Fan, et al., In vivo guiding nitrogen-doped carbon nanozyme for tumor catalytic therapy. Nature Communications, 2018, 9: 1440.
DOI
[65]

Q. Liang, J.Q. Xi, X.J.J. Gao, et al. A metal-free nanozyme-activated prodrug strategy for targeted tumor catalytic therapy. Nano Today, 2020, 35: 100935. https://doi.org/10.1016/j.nantod.2020.100935

[66]

Y.J. Sang, Y.Y. Huang, W. Li, et al. Bioinspired design of Fe3+-doped mesoporous carbon nanospheres for enhanced nanozyme activity. Chemistry A European Journal, 2018, 24(28): 7259−7263. https://doi.org/10.1002/chem.201801010

[67]

J.L. Zhu, G. Luo, X.X. Xi, et al. Cu2+-modified hollow carbon nanospheres: an unusual nanozyme with enhanced peroxidase-like activity. Microchimica Acta, 2021, 188(1): 8. https://doi.org/10.1007/s00604-020-04690-0

[68]

Y. Wang, A. Cho, G.R. Jia, et al. Tuning local coordination environments of manganese single-atom nanozymes with multi-enzyme properties for selective colorimetric biosensing. Angewandte Chemie, 2023, 135(15): e202300119. https://doi.org/10.1002/anie.202300119

[69]

H. Wang, C.Q. Liu, Z. Liu, et al. Specific oxygenated groups enriched graphene quantum dots as highly efficient enzyme mimics. Small, 2018, 14(13): 1703710. https://doi.org/10.1002/smll.201703710

[70]

X.L. Ren, J. Liu, J. Jing, et al. One-pot synthesis of active copper-containing carbon dots with laccase-like activities. Nanoscale, 2015, 7(46): 19641−19646. https://doi.org/10.1039/C5NR04685H

[71]

L.P. Lin, Y.L. Xiao, Y.H. Wang, et al. Hydrothermal synthesis of nitrogen and copper co-doped carbon dots with intrinsic peroxidase-like activity for colorimetric discrimination of phenylenediamine isomers. Microchimica Acta, 2019, 186: 288. https://doi.org/10.1007/s00604-019-3404-y

[72]

L. Su, Y.X. Cai, L. Wang, et al. Hemin@carbon dot hybrid nanozymes with peroxidase mimicking properties for dual (colorimetric and fluorometric) sensing of hydrogen peroxide, glucose and xanthine. Microchimica Acta, 2020, 187(2): 132. https://doi.org/10.1007/s00604-019-4103-4

[73]

A. Lee, W. Kang, J.S. Choi. Highly Enhanced Enzymatic Activity of Mn-Induced Carbon Dots and Their Application as Colorimetric Sensor Probes. Nanomaterials, 2021, 11(11): 3046. https://doi.org/10.3390/nano11113046

[74]

W.Y. Wang, Y.Zhou, X.R. Zhu, et al. Biocompatible ruthenium single-atom catalyst for cascade enzyme-mimicking therapy. ACS Applied Materials &Interfaces, 2021, 13(38): 45269−45278. https://doi.org/10.1021/acsami.1c12706

[75]

Z. Lin, Q. Zeng, Q. Deng, et al. Citric acid-derived carbon dots as excellent cysteine oxidase mimics for cysteine sensing. Sensors and Actuators B:Chemical, 2022, 359(15): 131563. https://doi.org/10.1016/j.snb.2022.131563

[76]

A. Prakobkij, P. Jarujamrus, S. Chunta, et al. Nitrogen-doped carbon dots/Ni-MnFe-layered double hydroxides (N-CDs/Ni-MnFe-LDHs) hybrid nanomaterials as immunoassay label for low-density lipoprotein detection. Microchimica Acta, 2022, 189: 72. https://doi.org/10.1007/s00604-022-05173-0

[77]

L. Su, S.N. Qin, Y.X. Cai, et al. Co, N-doped carbon dot nanozymes with acid pH-independence and substrate selectivity for biosensing and bioimaging. Sensors and Actuators B-Chemical, 2022, 353(15): 131150. https://doi.org/10.1016/j.snb.2021.131150

[78]

Y.H. Wang, J.C. Yao, Z.L. Cao, et al. Peroxidase-mimetic copper-doped carbon-dots for oxidative stress-mediated broad-spectrum and efficient antibacterial activity. Chemistry-A European Journal, 2022, 28: e202104174. https://doi.org/10.1002/chem.202104174

[79]

Y.C. Yang, T. Li, Y. Qin, et al. Construct of carbon nanotube-supported Fe2O3 hybrid nanozyme by atomic layer deposition for highly efficient dopamine sensing. Frontiers in Chemistry, 2020, 8: 564968. https://doi.org/10.3389/fchem.2020.564968

[80]

S.C. Yao, X.Y. Zhao, X.Y. Wang, et al. Bioinspired electron polarization of nanozymes with a human self-generated electric field for cancer catalytic therapy. Advanced Materials, 2022, 34: 21095. https://doi.org/10.1002/adma.202109568

[81]

A.K. Shukla, C. Sharma, A. Acharya. Bioinspired metal-free fluorescent carbon nanozyme with dual catalytic activity to confront cellular oxidative damage. ACS Applied Materials &Interfaces, 2021, 13(13): 15040−15052. https://doi.org/10.1021/acsami.1c03130

[82]

H.L. Yang, B.L. Xu, S.S. Li, et al. A photoresponsive nanozyme for synergistic catalytic therapy and dual phototherapy. Small, 2021, 17(10): e2007090. https://doi.org/10.1002/smll.202007090

[83]

X.B. Zhou, M.K. Wang, X.G. Su. Sensitive glutathione S-transferase assay based on Fe-doped hollow carbon nanospheres with oxidase-like activity. Sensors and Actuators B-Chemical, 2021, 338: 129777. https://doi.org/10.1016/j.snb.2021.129777

[84]

Y.T. Su, F. Wu, Q.X. Song, et al. 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. https://doi.org/10.1016/j.biomaterials.2021.121325

[85]

J.Q. Xi, Y.Q. Wang, X.J. Gao, et al. Reverse intratumor bacteria-induced gemcitabine resistance with carbon nanozymes for enhanced tumor catalytic-chemo therapy. Nano Today, 2022, 43: 101395. https://doi.org/10.1016/j.nantod.2022.101395

[86]

J.Q. Xi, G. Wei, Q.W. Wu, et al. Light-enhanced sponge-like carbon nanozyme used for synergetic antibacterial therapy. Biomaterials Science, 2019, 7(10): 4131−4141. https://doi.org/10.1039/C9BM00705A

[87]

Z.L. Xu, P.Z. Sun, J.J. Zhang, et al. High-efficiency platinum-carbon nanozyme for photodynamic and catalytic synergistic tumor therapy. Chemical Engineering Journal, 2020, 399: 125797. https://doi.org/10.1016/j.cej.2020.125797

[88]

L. Fan, X.D. Xu, C.H. Zhu, et al. Tumor catalytic–photothermal therapy with yolk–shell Gold@Carbon nanozymes. ACS Applied Materials &Interfaces, 2018, 10(5): 4502−4511. https://doi.org/10.1021/acsami.7b17916

[89]

Z.Q. Liu, W. Li, W.B. Sheng, et al. Tunable hierarchically structured meso-macroporous carbon spheres from a solvent-mediated polymerization-induced self-assembly. Journal of the American Chemical Society, 2023, 145(9): 5310−5319. https://doi.org/10.1021/jacs.2c12977

[90]

J.X. Chen, X.L. Zheng, J.X Zhang, et al. Bubble-templated synthesis of nanocatalyst Co/C as NADH oxidase mimic. National Science Review, 2022, 9(3): nwab186. https://doi.org/10.1093/nsr/nwab186

[91]

Z.R. Wang, R.F. Zhang, X.Y. Yan, et al. Structure and activity of nanozymes: Inspirations for de novo design of nanozymes. Materials Today, 2020, 41: 81−119. https://doi.org/10.1016/j.mattod.2020.08.020

[92]

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

[93]

N.S. Heo, H.P. Song, A.M. Lee, et al. Rosette-shaped graphitic carbon nitride acts as a peroxidase mimic in a wide pH range for fluorescence-based determination of glucose with glucose oxidase. Microchimica Acta, 2020, 187(5): 286. https://doi.org/10.1007/s00604-020-04249-z

[94]

F. Li, S. Li, X.C. Guo, et al. Chiral carbon dots mimicking topoisomerase I to mediate the topological rearrangement of supercoiled DNA enantioselectively. Angewandte Chemie International Edition, 2020, 59(27): 11087−11092. https://doi.org/10.1002/anie.202002904

[95]
I.A. Parinov, S.H. Chang, V.K. Gupta. Advanced Materials: Proceedings of the International Conference on “Physics and Mechanics of New Materials and Their Applications” PHENMA 2017. Cham: Springer International Publishing, 2018.
DOI
[96]

W.H. Gao, J.Y. He, L. Chen, et al. Deciphering the catalytic mechanism of superoxide dismutase activity of carbon dot nanozyme. Nature Communications, 2023, 14: 160. https://doi.org/10.1038/s41467-023-35828-2

[97]

Q. Wang, T. Ina, W.T. Chen, et al. Evolution of Zn(II) single atom catalyst sites during the pyrolysis-induced transformation of ZIF-8 to N-doped carbons. Science Bulletin, 2020, 65(20): 1743−1751. https://doi.org/10.1016/j.scib.2020.06.020

[98]
S.J. Zhuo, J. Feng, M. Li, et al., Manganese (II)-doped carbon dots as effective oxidase mimics for sensitive colorimetric determination of ascorbic acid. Microchimica Acta, 2019, 186: 745.
DOI
[99]

Y.F. Fan, M.Y. Cui, Y.M. Liu, et al. Selection and characterization of DNA aptamers for constructing colorimetric biosensor for detection of PBP2a. Spectrochimica Acta Part A:Molecular and Biomolecular Spectroscopy, 2020, 228: 117735. https://doi.org/10.1016/j.saa.2019.117735

[100]

Q. Luo, Y.L. Li, X.B. Huo, et al. Atomic chromium coordinated graphitic carbon nitride for bioinspired antibiofouling in seawater. Advanced Science, 2022, 9(8): 2105346. https://doi.org/10.1002/advs.202105346

[101]

B. Jiang, Z.J. Guo, M.M. Liang. Recent progress in single-atom nanozymes research. Nano Research, 2023, 16(2): 1878−1889. https://doi.org/10.1007/s12274-022-4856-7

[102]

K.W. Wan, B. Jiang, T. Tan, et al. Surface-mediated production of complexed •OH radicals and Fe=O species as a mechanism for iron oxide peroxidase-like nanozymes. Small, 2022, 18(50): 2204372. https://doi.org/10.1002/smll.202204372

[103]

G.H. Tang, J.Y. He, J.W. Liu, et al. Nanozyme for tumor therapy: surface modification matters. Exploration, 2021, 1(1): 75−89. https://doi.org/10.1002/EXP.20210005

[104]
Y.J. Gao, G. Hu, W. Zhang, et al. π–π Interaction intercalation of layered carbon materials with metallocene. Dalton Transactions, 2011, 40(17): 4542–4547.
DOI
[105]

B.F. Shi, Y.B. Su, L.L. Zhang, et al. Facilely prepared Fe3O4/nitrogen-doped graphene quantum dot hybrids as a robust nonenzymatic catalyst for visual discrimination of phenylenediamine isomers. Nanoscale, 2016, 8(20): 10814−10822. https://doi.org/10.1039/C6NR02725C

[106]

Q.B. Wang, J.P. Lei, S.Y. Demg, et al. Graphene-supported ferric porphyrin as a peroxidase mimic for electrochemical DNA biosensing. Chemical Communications, 2013, 49(9): 916−918. https://doi.org/10.1039/C2CC37664D

[107]

Y. Tao, Y.H. Lin, Z.Z. Huang, et al. Incorporating graphene oxide and gold nanoclusters: a synergistic catalyst with surprisingly high peroxidase-like activity over a broad pH range and its application for cancer cell detection. Advanced Materials, 2013, 25(18): 2594−2599. https://doi.org/10.1002/adma.201204419

[108]

J.J. Liu, J.M. Gao, A.M. Zhang, et al. Carbon nanocage-based nanozyme as an endogenous H2O2-activated oxygenerator for real-time bimodal imaging and enhanced phototherapy of esophageal cancer. Nanoscale, 2020, 12(42): 21674−21686. https://doi.org/10.1039/d0nr05945e

[109]

E. Tacconelli, E. Carrara, A. Savoldi, et al. Discovery, research, and development of new antibiotics: the WHO priority list of antibiotic-resistant bacteria and tuberculosis. The Lancet Infectious Diseases, 2018, 18(3): 318−327. https://doi.org/10.1016/S1473-3099(17)30753-3

[110]
L.Z. Gao, Y. Liu, D.Y Kim, et al. Nanocatalysts promote Streptococcus mutans biofilm matrix degradation and enhance bacterial killing to suppress dental caries in vivo. Biomaterials, 2016, 101: 272284.
DOI
[111]

Y.H. Liu, B.L. Xu, M.Z. Lu, et al. Ultrasmall Fe-doped carbon dots nanozymes for photoenhanced antibacterial therapy and wound healing. Bioactive Materials, 2022, 12: 246−256. https://doi.org/10.1016/j.bioactmat.2021.10.023

[112]

J.Q. Xi, G. Wei, L.F. An, et al. Copper/carbon hybrid nanozyme: tuning catalytic activity by the copper state for antibacterial therapy. Nano Letters, 2019, 19(11): 7645−7654. https://doi.org/10.1021/acs.nanolett.9b02242

[113]

J. Yao, Y. Zheng, M. Zhou, et al. ROS scavenging Mn3O4 nanozymes for in vivo anti-inflammation. Chemical Science, 2018, 9(11): 2927−2933. https://doi.org/10.1039/C7SC05476A

[114]

N. Singh, M.A. Savanur, S. Srivastava, et al. A redox modulatory Mn3O4 nanozyme with multi-enzyme activity provides efficient cytoprotection to human cells in a parkinson's disease model. Angewandte Chemie International Edition, 2017, 56(45): 14267−14271. https://doi.org/10.1002/anie.201708573

[115]

Y.Y. Huang, Z. Liu, C.Q. Liu, et al. Self-assembly of multi-nanozymes to mimic an intracellular antioxidant defense system. Angewandte Chemie International Edition, 2016, 55(23): 6646−6650. https://doi.org/10.1002/anie.201600868

[116]

N. Gao, K. Dong, A.D. Zhao, et al. Polyoxometalate-based nanozyme: Design of a multifunctional enzyme for multi-faceted treatment of Alzheimer’s disease. Nano Research, 2016, 9(4): 1079−1090. https://doi.org/10.1007/s12274-016-1000-6

[117]

Z.H. Miao, S.S. Jiang, M.L. Ding, et al. Ultrasmall rhodium nanozyme with RONS scavenging and photothermal activities for anti-inflammation and antitumor theranostics of colon diseases. Nano Letters, 2020, 20(5): 3079−3089. https://doi.org/10.1021/acs.nanolett.9b05035

[118]

J. Zhao, H.H. Wang, H.Q. Geng, et al. Au/N-doped carbon dot nanozymes as light-controlled anti-and pro-oxidants. ACS Applied Nano Materials, 2021, 4(7): 7253−7263. https://doi.org/10.1021/acsanm.1c01215

[119]

D. Jana, Y.L. Zhao. Strategies for enhancing cancer chemodynamic therapy performance. Exploration, 2022, 2: 20210238. https://doi.org/10.1002/exp.20210238

[120]

G.X. Duan, Y.Z. Zhang, B.Q. Luan, et al. Graphene-induced pore formation on cell membranes. Scientific Reports, 2017, 7(1): 42767. https://doi.org/10.1038/srep42767

[121]

X.Y. Wang, R. Lei, H.D. Huang, et al. The permeability and transport mechanism of graphene quantum dots (GQDs) across the biological barrier. Nanoscale, 2015, 7(5): 2034−2041. https://doi.org/10.1039/C4NR04136D

Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Received: 04 September 2023
Revised: 03 November 2023
Accepted: 13 November 2023
Published: 26 December 2023
Issue date: March 2024

Copyright

© The Author(s) 2024.

Acknowledgements

Acknowledgements

This work was supported by the National Key R&D Program of China (2022YFA1205801), and the National Natural Science Foundation of China (T2225026, 82172087, and 8210054010).

Rights and permissions

This is an open-access article distributed under  the  terms  of  the  Creative  Commons  Attribution  4.0 International  License (CC BY) (http://creativecommons.org/licenses/by/4.0/), which  permits  unrestricted  use,  distribution,  and reproduction in any medium, provided the original author and source are credited.

Return