733
Views
4
CrossRef citations to date
0
Altmetric
Review Articles

Enzyme hybrid nanoflowers and enzyme@metal–organic frameworks composites: fascinating hybrid nanobiocatalysts

, , , , , , , ORCID Icon & ORCID Icon show all
Pages 674-697 | Received 24 Aug 2022, Accepted 06 Feb 2023, Published online: 09 Apr 2023

References

  • Cui JD, Jia SR. Optimization protocols and improved strategies of cross-linked enzyme aggregates technology: current development and future challenges. Crit Rev Biotechnol. 2015;35:15–28.
  • Adlercreutz P. Immobilisation and application of lipases in organic media. Chem Soc Rev. 2013;42:6406–6436.
  • Zdarta J, Meyer AS, Jesionowski T, et al. A general overview of support materials for enzyme immobilization: characteristics, properties, practical utility. Catalysts. 2018;8:92.
  • Lokhande P, Kulkarni S, Chakrabarti S, et al. The progress and roadmap of metal–organic frameworks for high-performance supercapacitors. Coord Chem Rev. 2022;473:214771.
  • Bhembe YA, Lukhele LP, Hlekelele L, et al. Photocatalytic degradation of nevirapine with a heterostructure of few-layer black phosphorus coupled with niobium (V) oxide nanoflowers (FL-BP@ Nb2O5). Chemosphere. 2020;261:128159.
  • Thakur A, Kumar A, Kaya S, et al. Suppressing inhibitory compounds by nanomaterials for highly efficient biofuel production: a review. Fuel. 2022;312:122934.
  • Assad H, Fatma I, Kumar A, et al. An overview of MXene-Based nanomaterials and their potential applications towards hazardous pollutant adsorption. Chemosphere. 2022;298:134221.
  • Cui J, Jia S. Organic–inorganic hybrid nanoflowers: a novel host platform for immobilizing biomolecules. Coord Chem Rev. 2017;352:249–263.
  • Cui J, Ren S, Sun B, et al. Optimization protocols and improved strategies for metal-organic frameworks for immobilizing enzymes: current development and future challenges. Coord Chem Rev. 2018;370:22–41.
  • Feng Y, Hu H, Wang Z, et al. Three-dimensional ordered magnetic macroporous metal-organic frameworks for enzyme immobilization. J Colloid Interface Sci. 2021;590:436–445.
  • Sun B, Wang Z, Wang X, et al. Paper-based biosensor based on phenylalnine ammonia lyase hybrid nanoflowers for urinary phenylalanine measurement. Int J Biol Macromol. 2021;166:601–610.
  • Vo C-M, Cao ANT, Qazaq AS, et al. Toward syngas production from simulated biogas dry reforming. Fuel. 2022;326:125106.
  • Makola LC, Moeno S, Ouma CN, et al. Facile fabrication of a metal-free 2D-2D Nb2CTx@ g-C3N4 MXene-based Schottky-heterojunction with the potential application in photocatalytic processes. J Alloys Compd. 2022;916:165459.
  • Assad H, Kaya S, Kumar PS, et al. Insights into the role of nanotechnology on the performance of biofuel cells and the production of viable biofuels: a review. Fuel. 2022;323:124277.
  • Shen H, Shi H, Yang Y, et al. Highly efficient synergistic biocatalysis driven by stably loaded enzymes within hierarchically porous iron/cobalt metal–organic framework via biomimetic mineralization. J Mater Chem B. 2022;10:1553–1560.
  • Shen H, Shi H, Feng B, et al. A versatile biomimetic multienzyme cascade nanoplatform based on boronic acid-modified metal–organic framework for colorimetric biosensing. J Mater Chem B. 2022;10:3444–3451.
  • Ge J, Lei J, Zare RN. Protein–inorganic hybrid nanoflowers. Nat Nanotechnol. 2012;7:428–432.
  • Liu Y, Shao X, Kong D, et al. Immobilization of thermophilic lipase in inorganic hybrid nanoflower through biomimetic mineralization. Colloids Surf B. 2021;197:111450.
  • Wen H, Zhang L, Du Y, et al. Bimetal based inorganic-carbonic anhydrase hybrid hydrogel membrane for CO2 capture. J CO2 Util. 2020;39:101171.
  • Yu Y, Fei X, Tian J, et al. Self-assembled enzyme–inorganic hybrid nanoflowers and their application to enzyme purification. Colloids Surf B. 2015;130:299–304.
  • Zhang Y, Sun W, Elfeky NM, et al. Self-assembly of lipase hybrid nanoflowers with bifunctional Ca2+ for improved activity and stability. Enzyme Microb Technol. 2020;132:109408.
  • Patel SK, Otari SV, Li J, et al. Synthesis of cross-linked protein-metal hybrid nanoflowers and its application in repeated batch decolorization of synthetic dyes. J Hazard Mater. 2018;347:442–450.
  • Zhang B, Li P, Zhang H, et al. Red-blood-cell-like BSA/Zn3(PO4)2 hybrid particles: preparation and application to adsorption of heavy metal ions. Appl Surf Sci. 2016;366:328–338.
  • Ren W, Li Y, Wang J, et al. Synthesis of magnetic nanoflower immobilized lipase and its continuous catalytic application. New J Chem. 2019;43:11082–11090.
  • Hu Y, Dai L, Liu D, et al. Progress and prospect of metal-organic frameworks (MOFs) for enzyme immobilization (enzyme/MOFs). Renew Sustain Energy Rev. 2018;91:793–801.
  • Zhang M, Zhang Y, Yang C, et al. Enzyme-inorganic hybrid nanoflowers: classification, synthesis, functionalization and potential applications. Chem Eng J. 2021;415:129075.
  • Brady D, Jordaan J. Advances in enzyme immobilisation. Biotechnol Lett. 2009;31:1639.
  • Jesionowski T, Zdarta J, Krajewska B. Enzyme immobilization by adsorption: a review. Adsorption. 2014;20:801–821.
  • Liang S, Wu X-L, Xiong J, et al. Metal-organic frameworks as novel matrices for efficient enzyme immobilization: an update review. Coord Chem Rev. 2020;406:213149.
  • Rodrigues RC, Ortiz C, Berenguer-Murcia Á, et al. Modifying enzyme activity and selectivity by immobilization. Chem Soc Rev. 2013;42:6290–6307.
  • Wang X, Lan PC, Ma S. Metal–organic frameworks for enzyme immobilization: beyond host matrix materials. ACS Cent Sci. 2020;6:1497–1506.
  • Zhang Z, Yao Z-Z, Xiang S, et al. Perspective of microporous metal–organic frameworks for CO2 capture and separation. Energy Environ Sci. 2014;7:2868–2899.
  • Xu H, Liang H. Chitosan-regulated biomimetic hybrid nanoflower for efficiently immobilizing enzymes to enhance stability and by-product tolerance. Int J Biol Macromol. 2022;220:124–134.
  • Chen G, Kou X, Huang S, et al. Modulating the biofunctionality of metal-organic-framework-encapsulated enzymes through controllable embedding patterns. Angew Chem Int Ed. 2020;59:2867–2874.
  • Lyu F, Zhang Y, Zare RN, et al. One-pot synthesis of protein-embedded metal–organic frameworks with enhanced biological activities. Nano Lett. 2014;14:5761–5765.
  • Feng Y, Zhong L, Bilal M, et al. Enzymes@ZIF-8 nanocomposites with protection nanocoating: stability and acid-resistant evaluation. Polymers. 2018;11:27.
  • Feng Y, Zhong L, Jia S, et al. Acid-resistant enzyme@ MOF nanocomposites with mesoporous silica shells for enzymatic applications in acidic environments. J Biotechnol. 2019;306:54–61.
  • Zhang X, Tu R, Lu Z, et al. Hierarchical mesoporous metal–organic frameworks encapsulated enzymes: progress and perspective. Coord Chem Rev. 2021;443:214032.
  • Feng L, Yuan S, Zhang L-L, et al. Creating hierarchical pores by controlled linker thermolysis in multivariate metal–organic frameworks. J Am Chem Soc. 2018;140:2363–2372.
  • Hu C, Bai Y, Hou M, et al. Defect-induced activity enhancement of enzyme-encapsulated metal-organic frameworks revealed in microfluidic gradient mixing synthesis. Sci Adv. 2020;6:eaax5785.
  • Cheng K, Svec F, Lv Y, et al. Hierarchical micro- and mesoporous Zn-based metal-organic frameworks templated by hydrogels: their use for enzyme immobilization and catalysis of knoevenagel reaction. Small. 2019;15:e1902927.
  • Chen Y, Han S, Li X, et al. Why does enzyme not leach from metal–organic frameworks (MOFs)? Unveiling the interactions between an enzyme molecule and a MOF. Inorg Chem. 2014;53:10006–10008.
  • Gao S, Hou J, Deng Z, et al. Improving the acidic stability of zeolitic imidazolate frameworks by biofunctional molecules. Chem. 2019;5:1597–1608.
  • Jia X, Yang Z, Wang Y, et al. Hollow mesoporous silica@metal-organic framework and applications for pH-responsive drug delivery. ChemMedChem. 2018;13:400–405.
  • Wang Z, Liu Y, Li J, et al. Efficient immobilization of enzymes on amino functionalized MIL-125-NH2 metal organic framework. Biotechnol Bioprocess Eng. 2022;27:135–144.
  • Chen G, Kou X, Huang S, et al. Modulating the biofunctionality of metal-organic-framework-encapsulated enzymes through controllable embedding patterns. Angew Chem Int Ed Engl. 2020;59:2867–2874.
  • Liang J, Liang K. Nano-bio-interface engineering of metal-organic frameworks. Nano Today. 2021;40:101256.
  • Chen G, Huang S, Kou X, et al. A convenient and versatile amino-acid-boosted biomimetic strategy for the nondestructive encapsulation of biomacromolecules within metal-organic frameworks. Angew Chem Int Ed Engl. 2019;58:1463–1467.
  • Nadar SS, Varadan N, Suresh S, et al. Recent progress in nanostructured magnetic framework composites (MFCs): synthesis and applications. J Taiwan Inst Chem Eng. 2018;91:653–677.
  • Wang J, Zhao G, Yu F. Facile preparation of Fe3O4@ MOF core-shell microspheres for lipase immobilization. J Taiwan Inst Chem Eng. 2016;69:139–145.
  • Hou C, Wang Y, Ding Q, et al. Facile synthesis of enzyme-embedded magnetic metal–organic frameworks as a reusable mimic multi-enzyme system: mimetic peroxidase properties and colorimetric sensor. Nanoscale. 2015;7:18770–18779.
  • Ladole MR, Pokale PB, Patil SS, et al. Laccase immobilized peroxidase mimicking magnetic metal organic frameworks for industrial dye degradation. Bioresour Technol. 2020;317:124035.
  • Sharma N, Parhizkar M, Cong W, et al. Metal ion type significantly affects the morphology but not the activity of lipase–metal–phosphate nanoflowers. RSC Adv. 2017;7:25437–25443.
  • Ocsoy I, Dogru E, Usta S. A new generation of flowerlike horseradish peroxides as a nanobiocatalyst for superior enzymatic activity. Enzyme Microb Technol. 2015;75–76:25–29.
  • Lin Z, Xiao Y, Wang L, et al. Facile synthesis of enzyme–inorganic hybrid nanoflowers and their application as an immobilized trypsin reactor for highly efficient protein digestion. RSC Adv. 2014;4:13888–13891.
  • Somturk B, Yilmaz I, Altinkaynak C, et al. Synthesis of urease hybrid nanoflowers and their enhanced catalytic properties. Enzyme Microb Technol. 2016;86:134–142.
  • Liang L, Fei X, Li Y, et al. Hierarchical assembly of enzyme-inorganic composite materials with extremely high enzyme activity. RSC Adv. 2015;5:96997–97002.
  • Furukawa H, Cordova KE, O’Keeffe M, et al. The chemistry and applications of metal-organic frameworks. Sciecne. 2013;341:1230444.
  • Thawari AG, Rao CP. Peroxidase-like catalytic activity of copper-mediated protein–inorganic hybrid nanoflowers and nanofibers of β-lactoglobulin and α-lactalbumin: synthesis, spectral characterization, microscopic features, and catalytic activity. ACS Appl Mater Interfaces. 2016;8:10392–10402.
  • Rai SK, Narnoliya LK, Sangwan RS, et al. Self-assembled hybrid nanoflowers of manganese phosphate and L-arabinose isomerase: a stable and recyclable nanobiocatalyst for equilibrium level conversion of D-galactose to D-tagatose. ACS Sustain Chem Eng. 2018;6:6296–6304.
  • Zheng L, Sun Y, Wang J, et al. Preparation of a flower-like immobilized D-psicose 3-epimerase with enhanced catalytic performance. Catalysts. 2018;8:468.
  • Li C, Zhao J, Zhang Z, et al. Self-assembly of activated lipase hybrid nanoflowers with superior activity and enhanced stability. Biochem Eng J. 2020;158:107582.
  • Chung M, Nguyen TL, Tran TQN, et al. Ultrarapid sonochemical synthesis of enzyme-incorporated copper nanoflowers and their application to mediatorless glucose biofuel cell. Appl Surf Sci. 2018;429:203–209.
  • Batule BS, Park KS, Kim MI, et al. Ultrafast sonochemical synthesis of protein-inorganic nanoflowers. Int J Nanomed. 2015;10:137.
  • Sun J, Ge J, Liu W, et al. Multi-enzyme co-embedded organic–inorganic hybrid nanoflowers: synthesis and application as a colorimetric sensor. Nanoscale. 2014;6:255–262.
  • Li M, Luo M, Li F, et al. Biomimetic copper-based inorganic–protein nanoflower assembly constructed on the nanoscale fibrous membrane with enhanced stability and durability. J Phys Chem C. 2016;120:17348–17356.
  • Ren W, Fei X, Tian J, et al. Multiscale immobilized lipase for rapid separation and continuous catalysis. New J Chem. 2018;42:13471–13478.
  • Xu Z, Wang R, Liu C, et al. A new l-arabinose isomerase with copper ion tolerance is suitable for creating protein–inorganic hybrid nanoflowers with enhanced enzyme activity and stability. RSC Adv. 2016;6:30791–30794.
  • Nadar SS, Gawas SD, Rathod VK. Self-assembled organic inorganic hybrid glucoamylase nanoflowers with enhanced activity and stability. Int J Biol Macromol. 2016;92:660–669.
  • Hu Y, Dai L, Liu D, et al. Rationally designing hydrophobic UiO-66 support for the enhanced enzymatic performance of immobilized lipase. Green Chem. 2018;20:4500–4506.
  • Kumar A, Patel SK, Mardan B, et al. Immobilization of xylanase using a protein-inorganic hybrid system. J Microbiol Biotechnol. 2018;28:638–644.
  • He X, Chen L, He Q, et al. Cytochrome P450 enzyme-copper phosphate hybrid nano-flowers with superior catalytic performances for selective oxidation of sulfides. Chin J Chem. 2017;35:693–698.
  • Talens-Perales D, Fabra MJ, Martínez-Argente L, et al. Recyclable thermophilic hybrid protein-inorganic nanoflowers for the hydrolysis of milk lactose. Int J Biol Macromol. 2020;151:602–608.
  • Duan L, Li H, Zhang Y. Synthesis of hybrid nanoflower-based carbonic anhydrase for enhanced biocatalytic activity and stability. ACS Omega. 2018;3:18234–18241.
  • Li N, Hu B, Wang A, et al. Facile bioinspired preparation of fluorinase@fluoridated hydroxyapatite nanoflowers for the biosynthesis of 5′-fluorodeoxy adenosine. Sustainability. 2020;12:431.
  • Memon AH, Ding R, Yuan Q, et al. Facile synthesis of alcalase-inorganic hybrid nanoflowers used for soy protein isolate hydrolysis to improve its functional properties. Food Chem. 2019;289:568–574.
  • Maurya SS, Nadar SS, Rathod VK. A rapid self-assembled hybrid bio-microflowers of alpha–amylase with enhanced activity. J Biotechnol. 2020;317:27–33.
  • Ye R, Zhu C, Song Y, et al. Bioinspired synthesis of all-in-one organic-inorganic hybrid nanoflowers combined with a handheld pH meter for on-site detection of food pathogen. Small. 2016;12:3094–3100.
  • Zeinhom MMA, Wang Y, Sheng L, et al. Smart phone based immunosensor coupled with nanoflower signal amplification for rapid detection of Salmonella enteritidis in milk, cheese and water. Sens Actuators B. 2018;261:75–82.
  • Ye R, Zhu C, Song Y, et al. One-pot bioinspired synthesis of all-inclusive protein–protein nanoflowers for point-of-care bioassay: detection of E. coli O157: H7 from milk. Nanoscale. 2016;8:18980–18986.
  • Yu J, Wang C, Wang A, et al. Dual-cycle immobilization to reuse both enzyme and support by reblossoming enzyme–inorganic hybrid nanoflowers. RSC Adv. 2018;8:16088–16094.
  • Zhang B, Li P, Zhang H, et al. Preparation of lipase/Zn3(PO4)2 hybrid nanoflower and its catalytic performance as an immobilized enzyme. Chem Eng J. 2016;291:287–297.
  • Zhang B, Li P, Zhang H, et al. Papain/Zn3(PO4)2 hybrid nanoflower: preparation, characterization and its enhanced catalytic activity as an immobilized enzyme. RSC Adv. 2016;6:46702–46710.
  • Patel SK, Gupta RK, Kumar V, et al. Influence of metal ions on the immobilization of β-glucosidase through protein-inorganic hybrids. Indian J Microbiol. 2019;59:370–374.
  • Guo J, Wang Y, Zhao M. A self-activated nanobiocatalytic cascade system based on an enzyme-inorganic hybrid nanoflower for colorimetric and visual detection of glucose in human serum. Sens Actuators B Chem. 2019;284:45–54.
  • Cao X, Guo Y, Zhao M, et al. An efficient multi-enzyme cascade platform based on mesoporous metal-organic frameworks for the detection of organophosphorus and glucose. Food Chem. 2022;381:132282.
  • Soni S, Dwivedee BP, Banerjee UC. An ultrafast sonochemical strategy to synthesize lipase-manganese phosphate hybrid nanoflowers with promoted biocatalytic performance in the kinetic resolution of β-Aryloxyalcohols. ChemNanoMat. 2018;4:1007–1020.
  • Song Y, Gao J, He Y, et al. Preparation of a flowerlike nanobiocatalyst system via biomimetic mineralization of cobalt phosphate with enzyme. Ind Eng Chem Res. 2017;56:14923–14930.
  • Wu K, Zhang Y, Sun Q, et al. Preparation of cytochrome P450 enzyme-cobalt phosphate hybrid nano-flowers for oxidative coupling of benzylamine. Enzyme Microb Technol. 2019;131:109386.
  • López-Gallego F, Yate L. Selective biomineralization of Co 3 (PO 4) 2-sponges triggered by His-tagged proteins: efficient heterogeneous biocatalysts for redox processes. Chem Commun. 2015;51:8753–8756.
  • Patel SK, Choi H, Lee J-K. Multimetal-based inorganic–protein hybrid system for enzyme immobilization. ACS Sustain Chem Eng. 2019;7:13633–13638.
  • Zhu L, Gong L, Zhang Y, et al. Rapid detection of phenol using a membrane containing laccase nanoflowers. Chem Asian J. 2013;8:2358–2360.
  • Huang Y, Ran X, Lin Y, et al. Self-assembly of an organic–inorganic hybrid nanoflower as an efficient biomimetic catalyst for self-activated tandem reactions. Chem Commun. 2015;51:4386–4389.
  • Altinkaynak C, Yilmaz I, Koksal Z, et al. Preparation of lactoperoxidase incorporated hybrid nanoflower and its excellent activity and stability. Int J Biol Macromol. 2016;84:402–409.
  • Altinkaynak C, Tavlasoglu S, Ocsoy I. A new generation approach in enzyme immobilization: organic-inorganic hybrid nanoflowers with enhanced catalytic activity and stability. Enzyme Microb Technol. 2016;93:105–112.
  • Ke C, Fan Y, Chen Y, et al. A new lipase–inorganic hybrid nanoflower with enhanced enzyme activity. RSC Adv. 2016;6:19413–19416.
  • Wang L-B, Wang Y-C, He R, et al. A new nanobiocatalytic system based on allosteric effect with dramatically enhanced enzymatic performance. J Am Chem Soc. 2013;135:1272–1275.
  • Zhang Z, Zhang Y, He L, et al. A feasible synthesis of Mn3(PO4)2@ BSA nanoflowers and its application as the support nanomaterial for Pt catalyst. J Power Sources. 2015;284:170–177.
  • Zhang Z, Zhang Y, Song R, et al. Manganese (II) phosphate nanoflowers as electrochemical biosensors for the high-sensitivity detection of ractopamine. Sens Actuators B Chem. 2015;211:310–317.
  • Gao J, Liu H, Tong C, et al. Hemoglobin-Mn3(PO4)2 hybrid nanoflower with opulent electroactive centers for high-performance hydrogen peroxide electrochemical biosensor. Sens Actuators B Chem. 2020;307:127628.
  • Mohammad M, Ahmadpoor F, Shojaosadati SA. Mussel-inspired magnetic nanoflowers as an effective nanozyme and antimicrobial agent for biosensing and catalytic reduction of organic dyes. ACS Omega. 2020;5:18766–18777.
  • Zhao F, Wang Q, Dong J, et al. Enzyme-inorganic nanoflowers/alginate microbeads: an enzyme immobilization system and its potential application. Process Biochem. 2017;57:87–94.
  • Zhong L, Jiao X, Hu H, et al. Activated magnetic lipase-inorganic hybrid nanoflowers: a highly active and recyclable nanobiocatalyst for biodiesel production. Renew Energy. 2021;171:825–832.
  • Liu Y, Zhang Y, Li X, et al. Self-repairing metal–organic hybrid complexes for reinforcing immobilized chloroperoxidase reusability. Chem Commun. 2017;53:3216–3219.
  • Yang L, Wu N, Bai R, et al. A novel strategy for the detection of pyruvate in fermentation processes based on well-distributed enzyme-inorganic hybrid nanoflowers on thiol graphene modified gold electrodes. Electrochim Acta. 2022;427:140855.
  • Wang Q, Lian X, Fang Y, et al. Applications of immobilized bio-catalyst in metal-organic frameworks. Catalysts. 2018;8:166.
  • Li S, Liu X, Chai H, et al. Recent advances in the construction and analytical applications of metal-organic frameworks-based nanozymes. Trends Anal Chem. 2018;105:391–403.
  • Kempahanumakkagari S, Kumar V, Samaddar P, et al. Biomolecule-embedded metal-organic frameworks as an innovative sensing platform. Biotechnol Adv. 2018;36:467–481.
  • Wang J. Electrochemical glucose biosensors. Chem Rev. 2008;108:814–825.
  • Feng Y, Du Y, Kuang G, et al. Hierarchical micro-and mesoporous ZIF-8 with core–shell superstructures using colloidal metal sulfates as soft templates for enzyme immobilization. J Colloid Interface Sci. 2022;610:709–718.
  • Ahmad R, Shanahan J, Rizaldo S, et al. Co-immobilization of an enzyme system on a metal-organic framework to produce a more effective biocatalyst. Catalysts. 2020;10:499.
  • Zhu X, Huang J, Liu J, et al. A dual enzyme–inorganic hybrid nanoflower incorporated microfluidic paper-based analytic device (μPAD) biosensor for sensitive visualized detection of glucose. Nanoscale. 2017;9:5658–5663.
  • Ning D, Liu Q, Wang Q, et al. Luminescent MOF nanosheets for enzyme assisted detection of H2O2 and glucose and activity assay of glucose oxidase. Sens Actuators B. 2019;282:443–448.
  • Gao C, Zhu H, Chen J, et al. Facile synthesis of enzyme functional metal-organic framework for colorimetric detecting H2O2 and ascorbic acid. Chin Chem Lett. 2017;28:1006–1012.
  • Yin Y, Gao C, Xiao Q, et al. Protein-metal organic framework hybrid composites with intrinsic peroxidase-like activity as a colorimetric biosensing platform. ACS Appl Mater Interfaces. 2016;8:29052–29061.
  • Liu X, Qi W, Wang Y, et al. A facile strategy for enzyme immobilization with highly stable hierarchically porous metal–organic frameworks. Nanoscale. 2017;9:17561–17570.
  • Xie S, Ye J, Yuan Y, et al. A multifunctional hemin@ metal–organic framework and its application to construct an electrochemical aptasensor for thrombin detection. Nanoscale. 2015;7:18232–18238.
  • Wang X, Lu X, Wu L, et al. 3D metal-organic framework as highly efficient biosensing platform for ultrasensitive and rapid detection of bisphenol A. Biosens Bioelectron. 2015;65:295–301.
  • Lu X, Wang X, Wu L, et al. Response characteristics of bisphenols on a metal–organic framework-based tyrosinase nanosensor. ACS Appl Mater Interfaces. 2016;8:16533–16539.
  • Mehta J, Dhaka S, Bhardwaj N, et al. Application of an enzyme encapsulated metal-organic framework composite for convenient sensing and degradation of methyl parathion. Sens Actuators B. 2019;290:267–274.
  • Mehta J, Dhaka S, Paul AK, et al. Organophosphate hydrolase conjugated UiO-66-NH2 MOF based highly sensitive optical detection of methyl parathion. Environ Res. 2019;174:46–53.
  • Lei Z, Gao C, Chen L, et al. Recent advances in biomolecule immobilization based on self-assembly: organic–inorganic hybrid nanoflowers and metal–organic frameworks as novel substrates. J Mater Chem B. 2018;6:1581–1594.
  • Duan F, Feng X, Yang X, et al. A simple and powerful co-delivery system based on pH-responsive metal-organic frameworks for enhanced cancer immunotherapy. Biomaterials. 2017;122:23–33.
  • Zheng H, Zhang Y, Liu L, et al. One-pot synthesis of metal–organic frameworks with encapsulated target molecules and their applications for controlled drug delivery. J Am Chem Soc. 2016;138:962–968.
  • Tang L, Shi J, Wang X, et al. Coordination polymer nanocapsules prepared using metal–organic framework templates for pH-responsive drug delivery. Nanotechnology. 2017;28:275601.
  • Zhang Y, Wang F, Ju E, et al. Metal-organic-framework-based vaccine platforms for enhanced systemic immune and memory response. Adv Funct Mater. 2016;26:6454–6461.
  • Lian X, Huang Y, Zhu Y, et al. Enzyme-MOF nanoreactor activates nontoxic paracetamol for cancer therapy. Angew Chem. 2018;130:5827–5832.
  • Li P, Moon S-Y, Guelta MA, et al. Encapsulation of a nerve agent detoxifying enzyme by a mesoporous zirconium metal–organic framework engenders thermal and long-term stability. J Am Chem Soc. 2016;138:8052–8055.
  • Zhang L, Wang Z, Zhang Y, et al. Erythrocyte membrane cloaked metal–organic framework nanoparticle as biomimetic nanoreactor for starvation-activated colon cancer therapy. ACS Nano. 2018;12:10201–10211.
  • Chen W-H, Luo G-F, Vazquez-Gonzalez M, et al. Glucose-responsive metal–organic-framework nanoparticles act as “smart” sense-and-treat carriers. ACS Nano. 2018;12:7538–7545.
  • Baldemir A, Köse NB, Ildız N, et al. Synthesis and characterization of green tea (Camellia sinensis (L.) Kuntze) extract and its major components-based nanoflowers: a new strategy to enhance antimicrobial activity. RSC Adv. 2017;7:44303–44308.
  • Baldemir Kılıç A, Altinkaynak C, Ildız N, et al. A new approach for green synthesis and characterization of Artemisia L. (Asteraceae) genotype extracts-Cu2 nanocomplexes (nanoflower) and their effective antimicrobial activity. Med Sci. 2020;9:191.
  • Zhang M, Peltier R, Zhang M, et al. In situ reduction of silver nanoparticles on hybrid polydopamine–copper phosphate nanoflowers with enhanced antimicrobial activity. J Mater Chem B. 2017;5:5311–5317.
  • Hua X, Xing Y, Zhang X. Enhanced promiscuity of lipase-inorganic nanocrystal composites in the epoxidation of fatty acids in organic media. ACS Appl Mater Interfaces. 2016;8:16257–16261.
  • Yu J, Chen X, Jiang M, et al. Efficient promiscuous Knoevenagel condensation catalyzed by papain confined in Cu 3(PO4)2 nanoflowers. RSC Adv. 2018;8:2357–2364.
  • Jiang W, Wang X, Yang J, et al. Lipase-inorganic hybrid nanoflower constructed through biomimetic mineralization: a new support for biodiesel synthesis. J Colloid Interface Sci. 2018;514:102–107.
  • Patel S, Otari S, Kang Y, et al. Protein-inorganic hybrid system for efficient his-tagged enzymes immobilization and its application in L-xylulose production. RSC Adv. 2017;7:3488–3494.
  • Ahmed IN, Yang X-L, Dubale AA, et al. Hydrolysis of cellulose using cellulase physically immobilized on highly stable zirconium based metal-organic frameworks. Bioresour Technol. 2018;270:377–382.
  • Noma SAA, Yılmaz BS, Ulu A, et al. Development of L-Asparaginase@hybrid nanoflowers (ASNase@HNFs) reactor system with enhanced enzymatic reusability and stability. Catal Lett. 2021;151:1191–1201.
  • Zhong L, Feng Y, Hu H, et al. Enhanced enzymatic performance of immobilized lipase on metal organic frameworks with superhydrophobic coating for biodiesel production. J Colloid Interface Sci. 2021;602:426–436.
  • Li J, Lei X, Qiao Y, et al. The water status in China and an adaptive governance frame for water management. Int J Environ Res Public Health. 2020;17:2085.
  • Briamonte L, Zucaro R, Luzzi S. Sustainable management of water resources: agricultural sector and environmental protection. Econ Agro-Aliment. 2020;22:1–7.
  • Routoula E, Patwardhan SV. Degradation of anthraquinone dyes from effluents: a review focusing on enzymatic dye degradation with industrial potential. Environ Sci Technol. 2020;54:647–664.
  • Fu M, Xing J, Ge Z. Preparation of laccase-loaded magnetic nanoflowers and their recycling for efficient degradation of bisphenol A. Sci Total Environ. 2019;651:2857–2865.
  • Wu E, Li Y, Huang Q, et al. Laccase immobilization on amino-functionalized magnetic metal organic framework for phenolic compound removal. Chemosphere. 2019;233:327–335.
  • Alhayali NI, Özpozan NK, Dayan S, et al. Catalase/Fe3O4@ Cu2+ hybrid biocatalytic nanoflowers fabrication and efficiency in the reduction of organic pollutants. Polyhedron. 2021;194:114888.
  • Ranjan S, Dasgupta N, Chakraborty AR, et al. Nanoscience and nanotechnologies in food industries: opportunities and research trends. J Nanopart Res. 2014;16:1–23.
  • Terry LA, White SF, Tigwell LJ. The application of biosensors to fresh produce and the wider food industry. J Agric Food Chem. 2005;53:1309–1316.
  • Adley CC. Past, present and future of sensors in food production. Foods. 2014;3:491–510.
  • Xie J, Zhang Y, Simpson B. Food enzymes immobilization: novel carriers, techniques and applications. Curr Opin Food Sci. 2022;43:27–35.
  • Atiroglu V, Atiroglu A, Ozacar M. Immobilization of alpha-amylase enzyme on a protein @metal-organic framework nanocomposite: a new strategy to develop the reusability and stability of the enzyme. Food Chem. 2021;349:129127.
  • Han J, Feng H, Wu J, et al. Construction of multienzyme co-immobilized hybrid nanoflowers for an efficient conversion of cellulose into glucose in a cascade reaction. J Agric Food Chem. 2021;69:7910–7921.
  • Zhu L, Shen B, Song Z, et al. Permeabilized TreS-expressing Bacillus subtilis cells decorated with glucose Isomerase and a shell of ZIF-8 as a reusable biocatalyst for the coproduction of trehalose and fructose. J Agric Food Chem. 2020;68:4464–4472.
  • Fotiadou R, Patila M, Hammami MA, et al. Development of effective lipase-hybrid nanoflowers enriched with carbon and magnetic nanomaterials for biocatalytic transformations. Nanomaterials. 2019;9:808.
  • Hu H, Li P, Wang Z, et al. Glutamate oxidase-integrated biomimetic metal–organic framework hybrids as cascade nanozymes for ultrasensitive glutamate detection. J Agric Food Chem. 2022;70(12):3785–3794.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.