AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (893.2 KB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Research Article | Open Access

An antifouling polydopamine-based fluorescent aptasensor for determination of arginine kinase

Yanbo WangHuan LiJinru ZhouFangting WangYifan QianLinglin Fu( )
Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, China

Peer review under responsibility of KeAi Communications Co., Ltd.

Show Author Information

Abstract

Simple yet efficient detection methods for food allergens are in urgent need to help people avoid the risks imposed by allergenic food. In this work, a polydopamine (PDA)-based fluorescent aptasensor was developed to detect arginine kinase (AK), one of the major allergens in shellfish. The aptamer towards AK was firstly selected via systematic evolution of ligands by exponential enrichment method and labeled with fluorescein amidite (FAM) to build a fluorescence resonance energy transfer (FRET) system with PDA particles. Polyethylene glycol (PEG) was employed to construct an antifouling surface for the aptasensor to eliminate food matrix interferences. With the presence of AK, the PDA-based aptasensor exhibited elevated fluorescent signals as the FAM-labeled aptamer bound to AK and detached from the PDA particles. The aptasensor showed great stability and resistance to nonspecific interference of background proteins and had a limit of detection (LOD) of 0.298 μg/mL. The proposed aptasensor was further proved to be feasible for quantitative analysis of AK in nine species of shrimps and five commercial processed products, which indicated its high potential in tracing the presence of AK in complex aquatic products.

References

[1]

W.W. Acker, J.M. Plasek, K.G. Blumenthal, et al., Prevalence of food allergies and intolerances documented in electronic health records, J. Allergy Clin. Immunol. 140 (2017) 1587-1591. https://doi.org/10.1016/j.jaci.2017.04.006.

[2]

Y. Wang, Z. Rao, J. Zhou, et al., A chiral assembly of gold nanoparticle trimer-based biosensors for ultrasensitive detection of the major allergen tropomyosin in shellfish, Biosens. Bioelectron. 132 (2019) 84-89. https://doi.org/10.1016/j.bios.2019.02.038.

[3]

C.K. Woo, S.L. Bahna, Not all shellfish "allergy" is allergy! Clin. Transl. Allergy 1 (2011) 3. https://doi.org/10.1186/2045-7022-1-3.

[4]

C.J. Yu, Y.F. Lin, B.L. Chiang, et al., Proteomics and immunological analysis of a novel shrimp allergen, Pen m 2, J. Immunol. 170 (2003) 445. https://doi.org/10.4049/jimmunol.170.1.445.

[5]

H.Y. Mao, M.J. Cao, S.J. Maleki, et al., Structural characterization and IgE epitope analysis of arginine kinase from Scylla paramamosain, Mol. Immunol. 56 (2013) 463-470. https://doi.org/10.1016/j.molimm.2013.04.016.

[6]

J. Shroba, N. Rath, C. Barnes, Possible role of environmental factors in the development of food allergies, Clin. Rev. Allergy Immunol. 57 (2018) 303-311. https://doi.org/10.1007/s12016-018-8703-2.

[7]

J. Heick, M. Fischer, B. Pöpping, First screening method for the simultaneous detection of seven allergens by liquid chromatography mass spectrometry, J. Chromatogr. A 1218 (2011) 938-943. https://doi.org/10.1016/j.chroma.2010.12.067.

[8]

L. Monaci, E. De Angelis, N. Montemurro, et al., Comprehensive overview and recent advances in proteomics MS based methods for food allergens analysis, TrAC, Trends Anal. Chem. 106 (2018) 21-36. https://doi.org/10.1016/j.trac.2018.06.016.

[9]

T. Mairal, P. Nadal, M. Svobodova, et al., FRET-based dimeric aptamer probe for selective and sensitive Lup an 1 allergen detection, Biosens. Bioelectron. 54 (2014) 207-210. https://doi.org/10.1016/j.bios.2013.10.070.

[10]

X. Weng, S. Neethirajan, A microfluidic biosensor using graphene oxide and aptamer-functionalized quantum dots for peanut allergen detection, Biosens. Bioelectron. 85 (2016) 649-656. https://doi.org/10.1016/j.bios.2016.05.072.

[11]

Z.S. Pehlivan, M. Torabfam, H. Kurt, Aptamer and nanomaterial based FRET biosensors: a review on recent advances (2014–2019), Microchim. Acta 186 (2019) 563. https://doi.org/10.1007/s00604-019-3659-3.

[12]

L. Ma, Z. Lei, F. Liu, et al., Cy5 labeled single-stranded DNA-polydopamine nanoparticle conjugate-based FRET assay for reactive oxygen species detection, Sensing and Bio-Sensing Research 3 (2015) 92-97. https://doi.org/10.1016/j.sbsr.2014.12.006.

[13]

S. Ma, Y.X. Qi, X.Q. Jiang, et al., Selective and sensitive monitoring of cerebral antioxidants based on the dye-Labeled DNA/polydopamine conjugates, Anal. Chem. 88 (2016) 11647-11653. https://doi.org/10.1021/acs.analchem.6b03216.

[14]

J.H. Ryu, P.B. Messersmith, H. Lee, Polydopamine surface chemistry: A decade of discovery, ACS Appl. Mater. Interfaces 10 (2018) 7523-7540. https://doi.org/10.1021/acsami.7b19865.

[15]

Y. Liu, L. Jiang, X. Fan, et al., Intracellular fluorometric determination of microRNA-21 by using a switch-on nanoprobe composed of carbon nanotubes and gold nanoclusters, Microchim. Acta 186 (2019) 447. https://doi.org/10.1007/s00604-019-3573-8.

[16]

J. Lee, A.A.S. Samson, Y. Yim, et al., A FRET assay for the quantitation of inhibitors of exonuclease EcoRV by using parchment paper inkjet-printed with graphene oxide and FAM-labelled DNA, Microchim. Acta 186 (2019) 211. https://doi.org/10.1007/s00604-019-3317-9.

[17]

C. Zhu, Z. Zeng, H. Li, et al., Single-layer MoS2-based nanoprobes for homogeneous detection of biomolecules, J. Am. Chem. Soc. 135 (2013) 5998-6001. https://doi.org/10.1021/ja4019572.

[18]

X. Zhu, H. Zheng, X. Wei, et al., Metal-organic framework (MOF): a novel sensing platform for biomolecules, Chem. Commun. 49 (2013) 1276-1278. https://doi.org/10.1039/C2CC36661D.

[19]

H. Vaisocherová-Lísalová, F. Surman, I. Víšová, et al., Copolymer brush-based ultralow-fouling biorecognition surface platform for food safety, Anal. Chem. 88 (2016) 10533-10539. https://doi.org/10.1021/acs.analchem.6b02617.

[20]

W. Nie, Q. Wang, L. Zou, et al., Low-fouling surface plasmon resonance sensor for highly sensitive detection of microRNA in a complex matrix based on the DNA tetrahedron, Anal. Chem. 90 (2018) 12584-12591. https://doi.org/10.1021/acs.analchem.8b02686.

[21]

L. Chen, S. Lv, M. Liu, et al., Low-fouling magnetic nanoparticles and evaluation of their potential application as disease markers assay in whole serum, ACS Appl. Nano. Mater. 1 (2018) 2489-2495. https://doi.org/10.1021/acsanm.8b00525.

[22]

J.V. Jokerst, T. Lobovkina, R.N. Zare, Nanoparticle PEGylation for imaging and therapy, Nanomedicine 6 (2011) 715-728. https://doi.org/10.2217/nnm.11.19.

[23]

D.T. Tran, K. Knez, K.P. Janssen, et al., Selection of aptamers against Ara h 1 protein for FO-SPR biosensing of peanut allergens in food matrices, Biosens. Bioelectron. 43 (2013) 245-251. https://doi.org/10.1016/j.bios.2012.12.022.

[24]

M. Khedri, M. Ramezani, H. Rafatpanah, et al., Detection of food-born allergens with aptamer-based biosensors, TrAC Trends Anal. Chem. 103 (2018) 126-136. https://doi.org/10.1016/j.trac.2018.04.001.

[25]

Y. Liu, K. Ai, J. Liu, et al., Dopamine-melanin colloidal nanospheres: an efficient near-infrared photothermal therapeutic agent for in vivo cancer therapy, Adv. Mater. 25 (2013) 1353-1359. https://doi.org/10.1002/adma.201204683.

[26]

L. Fu, J. Zhou, C. Wang, et al., Ion-exchange chromatography coupled with dynamic coating capillary electrophoresis for simultaneous determination of tropomyosin and arginine kinase in shellfish, Front. Chem. (2018) 6. https://doi.org/10.3389/fchem.2018.00305.

[27]

K.M. Song, E. Jeong, W. Jeon, et al., A coordination polymer nanobelt (CPNB)-based aptasensor for sulfadimethoxine, Biosens. Bioelectron. 33 (2012) 113-119. https://doi.org/10.1016/j.bios.2011.12.034.

[28]

S.Valenzano, A. De Girolamo, M.C. DeRosa, et al., Screening and identification of DNA aptamers to tyramine using in vitro selection and high-throughput sequencing, ACS Comb. Sci. 18 (2016) 302-313. https://doi.org/10.1021/acscombsci.5b00163.

[29]

L. Fu, Y. Qian, J., Zhou, Quantification of shellfish arginine kinases by double-enhanced immunoassay employing magnetic beads and gold nanoparticles as carrier, LWT-Food Sci. Technol. 122 (2019) 108916. https://doi.org/10.1016/j.lwt.2019.108916.

[30]

S. Lowe, N.M. O’Brien-Simpson, L.A. Connal, Antibiofouling polymer interfaces: poly(ethylene glycol) and other promising candidates, Poly. Chem. 6 (2015) 198-212. https://doi.org/10.1039/C4PY01356E.

[31]

X.J. Hao, X.H. Zhou, Y. Zhang, Melamine detection in dairy products by using a reusable evanescent wave fiber-optic biosensor, Sens. Actuators, B 204 (2014) 682-687. https://doi.org/10.1016/j.snb.2014.08.023.

[32]

M. Shoji, R. Adachi, H. Akiyama, Japanese food allergen labeling regulation: an update, J. AOAC Int. 101 (2017) 8-13. https://doi.org/10.5740/jaoacint.17-0389.

Food Science and Human Wellness
Pages 737-744
Cite this article:
Wang Y, Li H, Zhou J, et al. An antifouling polydopamine-based fluorescent aptasensor for determination of arginine kinase. Food Science and Human Wellness, 2023, 12(3): 737-744. https://doi.org/10.1016/j.fshw.2022.09.007

428

Views

45

Downloads

5

Crossref

5

Web of Science

5

Scopus

0

CSCD

Altmetrics

Received: 26 December 2020
Revised: 14 January 2021
Accepted: 06 February 2021
Published: 15 October 2022
© 2023 Beijing Academy of Food Sciences. Publishing services by Elsevier B.V. on behalf of KeAi Communications Co., Ltd.

This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Return