Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 14.880
Filtrar
Más filtros

Publication year range
1.
Nature ; 631(8022): 884-890, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39020178

RESUMEN

Plastic production reached 400 million tons in 2022 (ref. 1), with packaging and single-use plastics accounting for a substantial amount of this2. The resulting waste ends up in landfills, incineration or the environment, contributing to environmental pollution3. Shifting to biodegradable and compostable plastics is increasingly being considered as an efficient waste-management alternative4. Although polylactide (PLA) is the most widely used biosourced polymer5, its biodegradation rate under home-compost and soil conditions remains low6-8. Here we present a PLA-based plastic in which an optimized enzyme is embedded to ensure rapid biodegradation and compostability at room temperature, using a scalable industrial process. First, an 80-fold activity enhancement was achieved through structure-based rational engineering of a new hyperthermostable PLA hydrolase. Second, the enzyme was uniformly dispersed within the PLA matrix by means of a masterbatch-based melt extrusion process. The liquid enzyme formulation was incorporated in polycaprolactone, a low-melting-temperature polymer, through melt extrusion at 70 °C, forming an 'enzymated' polycaprolactone masterbatch. Masterbatch pellets were integrated into PLA by melt extrusion at 160 °C, producing an enzymated PLA film (0.02% w/w enzyme) that fully disintegrated under home-compost conditions within 20-24 weeks, meeting home-composting standards. The mechanical and degradation properties of the enzymated film were compatible with industrial packaging applications, and they remained intact during long-term storage. This innovative material not only opens new avenues for composters and biomethane production but also provides a feasible industrial solution for PLA degradation.


Asunto(s)
Plásticos Biodegradables , Biodegradación Ambiental , Enzimas Inmovilizadas , Hidrolasas , Poliésteres , Ingeniería de Proteínas , Plásticos Biodegradables/química , Plásticos Biodegradables/metabolismo , Enzimas Inmovilizadas/química , Enzimas Inmovilizadas/metabolismo , Hidrolasas/metabolismo , Hidrolasas/química , Poliésteres/química , Poliésteres/metabolismo , Suelo/química , Temperatura , Estabilidad de Enzimas , Compostaje
2.
Proc Natl Acad Sci U S A ; 121(30): e2403460121, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-39008666

RESUMEN

Autonomous nanorobots represent an advanced tool for precision therapy to improve therapeutic efficacy. However, current nanorobotic designs primarily rely on inorganic materials with compromised biocompatibility and limited biological functions. Here, we introduce enzyme-powered bacterial outer membrane vesicle (OMV) nanorobots. The immobilized urease on the OMV membrane catalyzes the decomposition of bioavailable urea, generating effective propulsion for nanorobots. This OMV nanorobot preserves the unique features of OMVs, including intrinsic biocompatibility, immunogenicity, versatile surface bioengineering for desired biofunctionalities, capability of cargo loading and protection. We present OMV-based nanorobots designed for effective tumor therapy by leveraging the membrane properties of OMVs. These involve surface bioengineering of robotic body with cell-penetrating peptide for tumor targeting and penetration, which is further enhanced by active propulsion of nanorobots. Additionally, OMV nanorobots can effectively safeguard the loaded gene silencing tool, small interfering RNA (siRNA), from enzymatic degradation. Through systematic in vitro and in vivo studies using a rodent model, we demonstrate that these OMV nanorobots substantially enhanced siRNA delivery and immune stimulation, resulting in the utmost effectiveness in tumor suppression when juxtaposed with static groups, particularly evident in the orthotopic bladder tumor model. This OMV nanorobot opens an inspiring avenue to design advanced medical robots with expanded versatility and adaptability, broadening their operation scope in practical biomedical domains.


Asunto(s)
Membrana Externa Bacteriana , Animales , Humanos , Membrana Externa Bacteriana/metabolismo , Ratones , Robótica/métodos , Ureasa/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Enzimas Inmovilizadas/química , Enzimas Inmovilizadas/metabolismo
3.
Nat Chem Biol ; 20(6): 732-741, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38321209

RESUMEN

Glycosylation is a critical post-translational protein modification that affects folding, half-life and functionality. Glycosylation is a non-templated and heterogeneous process because of the promiscuity of the enzymes involved. We describe a platform for sequential glycosylation reactions for tailored sugar structures (SUGAR-TARGET) that allows bespoke, controlled N-linked glycosylation in vitro enabled by immobilized enzymes produced with a one-step immobilization/purification method. We reconstruct a reaction cascade mimicking a glycosylation pathway where promiscuity naturally exists to humanize a range of proteins derived from different cellular systems, yielding near-homogeneous glycoforms. Immobilized ß-1,4-galactosyltransferase is used to enhance the galactosylation profile of three IgGs, yielding 80.2-96.3% terminal galactosylation. Enzyme recycling is demonstrated for a reaction time greater than 80 h. The platform is easy to implement, modular and reusable and can therefore produce homogeneous glycan structures derived from various hosts for functional and clinical evaluation.


Asunto(s)
Enzimas Inmovilizadas , Galactosiltransferasas , Glicosilación , Enzimas Inmovilizadas/química , Enzimas Inmovilizadas/metabolismo , Humanos , Galactosiltransferasas/metabolismo , Galactosiltransferasas/química , Polisacáridos/metabolismo , Polisacáridos/química , Procesamiento Proteico-Postraduccional
4.
Acc Chem Res ; 57(1): 93-105, 2024 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-38105494

RESUMEN

Manufacturing is undergoing profound transformations, among which green biomanufacturing with low energy consumption, high efficiency, and sustainability is becoming one of the major trends. However, enzymes, as the "core chip" of biomanufacturing, are often handicapped in their application by their high cost, low operational stability, and nonreusability. Immobilization of enzymes is a technology that binds or restricts enzymes in a certain area with solid materials, allows them to still carry out their unique catalytic reaction, and allows them to be recycled and reused. Compared with free enzymes, immobilized enzymes boast numerous advantages such as enhanced storage stability, ease of separation, reusability, and controlled operation. Currently, commonly used supports for enzyme immobilization (e.g., mesoporous silica, sol-gel hydrogels, and porous polymer) can effectively improve enzyme stability and reduce product inhibition. However, they still face drawbacks such as potential leaching or conformational change during immobilization and poor machining performance. Especially, most enzyme carrier solid materials possess disordered structures, inevitably introducing deficiencies such as low loading capacity, hindered mass transfer, and unclear structure-property relationships. Additionally, it remains a notable challenge to meticulously design immobilization systems tailored to the specific characteristics of enzyme/reaction. Therefore, there is a significant demand for reliable solid materials to overcome the above challenges. Crystalline porous materials, particularly covalent organic frameworks (COFs), have garnered significant interest as a promising platform for immobilizing enzymes due to their unique properties, such as their crystalline nature, high porosity, accessible active sites, versatile synthetic conditions, and tunable structure. COFs create a stabilizing microenvironment that protects enzymes from denaturation and significantly enhances reusability. Nevertheless, some challenges still remain, including difficulties in loading large enzymes, reduced enzyme activities, and the limited functionality of carriers. Therefore, it is essential to develop innovative carriers and novel strategies to broaden the methods of immobilizing enzymes, enabling their application across a more diverse array of fields.The integration of enzymes with advanced porous materials for intensified performance and diverse applications is still in its infancy, and our group has done a series of pioneering works. This Account presents a comprehensive overview of recent research progress made by our group, including (i) the development of innovative enzyme immobilization strategies utilizing COFs to make the assembly and integration of enzymes and carriers more effective; (ii) rational design and construction of functional carriers for enzyme immobilization using COFs; and (iii) extensions of immobilized enzyme applications based on COFs from industrial catalysis to biomedicine and chiral separation. The integration of enzymes with functional crystalline materials offers mutual benefits and results in a performance that surpasses what either component can achieve individually. Additionally, immobilized enzymes exhibit enhanced functionality and intriguing characteristics that differ from those of free enzymes. Consistent with our research philosophy centered on integration, platform development, and engineering application, this Account addresses the critical challenges associated with enzyme immobilization using COFs while extending the applications of COFs and proposing future design principles for biomanufacturing and enzyme industry.


Asunto(s)
Estructuras Metalorgánicas , Enzimas Inmovilizadas , Polímeros , Catálisis , Ingeniería
5.
Nano Lett ; 24(11): 3404-3412, 2024 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-38451852

RESUMEN

Assembling metal-organic frameworks (MOFs) into ordered multidimensional porous superstructures promises the encapsulation of enzymes for heterogeneous biocatalysts. However, the full potential of this approach has been limited by the poor stability of enzymes and the uncontrolled assembly of MOF nanoparticles onto suitable supports. In this study, a novel and exceptionally robust Ni-imidazole-based MOF was synthesized in water at room temperature, enabling in situ enzyme encapsulation. Based on this MOF platform, we developed a DNA-directed assembly strategy to achieve the uniform placement of MOF nanoparticles onto bacterial cellulose nanofibers, resulting in a distinctive "branch-fruit" structure. The resulting hybrid materials demonstrated remarkable versatility across various catalytic systems, accommodating natural enzymes, nanoenzymes, and multienzyme cascades, thus showcasing enormous potential as universal microbioreactors. Furthermore, the hierarchical composites facilitated rapid diffusion of the bulky substrate while maintaining the enzyme stability, with ∼3.5-fold higher relative activity compared to the traditional enzyme@MOF immobilized in bacterial cellulose nanofibers.


Asunto(s)
Enzimas Inmovilizadas , Nanofibras , Enzimas Inmovilizadas/química , Celulosa , Frutas , ADN/química
6.
Nano Lett ; 24(15): 4682-4690, 2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38563501

RESUMEN

Multienzyme assemblies mediated by multivalent interaction play a crucial role in cellular processes. However, the three-dimensional (3D) programming of an enzyme complex with defined enzyme activity in vitro remains unexplored, primarily owing to limitations in precisely controlling the spatial topological configuration. Herein, we introduce a nanoscale 3D enzyme assembly using a tetrahedral DNA framework (TDF), enabling the replication of spatial topological configuration and maintenance of an identical edge-to-edge distance akin to natural enzymes. Our results demonstrate that 3D nanoscale enzyme assemblies in both two-enzyme systems (glucose oxidase (GOx)/horseradish peroxidase (HRP)) and three-enzyme systems (amylglucosidase (AGO)/GOx/HRP) lead to enhanced cascade catalytic activity compared to the low-dimensional structure, resulting in ∼5.9- and ∼7.7-fold enhancements over homogeneous diffusional mixtures of free enzymes, respectively. Furthermore, we demonstrate the enzyme assemblies for the detection of the metabolism biomarkers creatinine and creatine, achieving a low limit of detection, high sensitivity, and broad detection range.


Asunto(s)
Enzimas Inmovilizadas , Glucosa Oxidasa , Enzimas Inmovilizadas/química , Peroxidasa de Rábano Silvestre/química , Glucosa Oxidasa/química , ADN/química
7.
J Biol Chem ; 299(2): 102856, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36596358

RESUMEN

Heat shock protein 90 (Hsp90) is known to mediate heme insertion and activation of heme-deficient neuronal nitric oxide (NO) synthase (apo-nNOS) in cells by a highly dynamic interaction that has been extremely difficult to study mechanistically with the use of subcellular systems. In that the heme content of many critical hemeproteins is regulated by Hsp90 and the heme chaperone GAPDH, the development of an in vitro system for the study of this chaperone-mediated heme regulation would be extremely useful. Here, we show that use of an antibody-immobilized apo-nNOS led not only to successful assembly of chaperone complexes but the ability to show a clear dependence on Hsp90 and GAPDH for heme-mediated activation of apo-nNOS. The kinetics of binding for Hsp70 and Hsp90, the ATP and K+ dependence, and the absolute requirement for Hsp70 in assembly of Hsp90•apo-nNOS heterocomplexes all point to a similar chaperone machinery to the well-established canonical machine regulating steroid hormone receptors. However, unlike steroid receptors, the use of a purified protein system containing Hsp90, Hsp70, Hsp40, Hop, and p23 is unable to activate apo-nNOS. Thus, heme insertion requires a unique Hsp90-chaperone complex. With this newly developed in vitro system, which recapitulates the cellular process requiring GAPDH as well as Hsp90, further mechanistic studies are now possible to better understand the components of the Hsp90-based chaperone system as well as how this heterocomplex works with GAPDH to regulate nNOS and possibly other hemeproteins.


Asunto(s)
Gliceraldehído-3-Fosfato Deshidrogenasas , Proteínas HSP70 de Choque Térmico , Proteínas HSP90 de Choque Térmico , Hemo , Hemoproteínas , Chaperonas Moleculares , Óxido Nítrico Sintasa , Hemo/química , Hemoproteínas/química , Hemoproteínas/metabolismo , Proteínas HSP70 de Choque Térmico/química , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas HSP90 de Choque Térmico/química , Proteínas HSP90 de Choque Térmico/metabolismo , Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , Unión Proteica , Óxido Nítrico Sintasa/química , Óxido Nítrico Sintasa/metabolismo , Enzimas Inmovilizadas , Gliceraldehído-3-Fosfato Deshidrogenasas/química , Gliceraldehído-3-Fosfato Deshidrogenasas/metabolismo , Activación Enzimática
8.
J Am Chem Soc ; 146(13): 9112-9123, 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38500441

RESUMEN

Recent advances have demonstrated the promise of complex multicomponent polymeric supports to enable supra-biological enzyme performance. However, the discovery of such supports has been limited by time-consuming, low-throughput synthesis and screening. Here, we describe a novel combinatorial and high-throughput platform that enables rapid screening of complex and heterogeneous copolymer brushes as enzyme immobilization supports, named combinatorial high-throughput enzyme support screening (CHESS). Using a 384-well plate format, we synthesized arrays of three-component polymer brushes in the microwells using photoactivated surface-initiated polymerization and immobilized enzymes in situ. The utility of CHESS to identify optimal immobilization supports under thermally and chemically denaturing conditions was demonstrated usingBacillus subtilisLipase A (LipA). The identification of supports with optimal compositions was validated by immobilizing LipA on polymer-brush-modified biocatalyst particles. We further demonstrated that CHESS could be used to predict the optimal composition of polymer brushes a priori for the previously unexplored enzyme, alkaline phosphatase (AlkP). Our findings demonstrate that CHESS represents a predictable and reliable platform for dramatically accelerating the search of chemical compositions for immobilization supports and further facilitates the discovery of biocompatible and stabilizing materials.


Asunto(s)
Enzimas Inmovilizadas , Ensayos Analíticos de Alto Rendimiento , Enzimas Inmovilizadas/química , Polímeros/química
9.
J Am Chem Soc ; 146(30): 20778-20787, 2024 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-39013149

RESUMEN

The forward design of in vitro enzymatic reaction networks (ERNs) requires a detailed analysis of network kinetics and potentially hidden interactions between the substrates and enzymes. Although flow chemistry allows for a systematic exploration of how the networks adapt to continuously changing conditions, the analysis of the reaction products is often a bottleneck. Here, we report on the interface between a continuous stirred-tank reactor, in which an immobilized enzymatic network made of 12 enzymes is compartmentalized, and an ion mobility-mass spectrometer. Feeding uniformly 13C-labeled inputs to the enzymatic network generates all isotopically labeled reaction intermediates and products, which are individually detected by ion mobility-mass spectrometry (IMS-MS) based on their mass-to-charge ratios and inverse ion mobilities. The metabolic flux can be continuously and quantitatively monitored by diluting the ERN output with nonlabeled standards of known concentrations. The real-time quantitative data obtained by IMS-MS are then harnessed to train a model of network kinetics, which proves sufficiently predictive to control the ERN output after a single optimally designed experiment. The high resolution of the time-course data provided by this approach is an important stepping stone to design and control sizable and intricate ERNs.


Asunto(s)
Enzimas Inmovilizadas , Espectrometría de Masas , Espectrometría de Masas/métodos , Enzimas Inmovilizadas/química , Enzimas Inmovilizadas/metabolismo , Cinética , Espectrometría de Movilidad Iónica/métodos
10.
J Am Chem Soc ; 146(25): 17189-17200, 2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38864358

RESUMEN

Spatial immobilization of fragile enzymes using a nanocarrier is an efficient means to design heterogeneous biocatalysts, presenting superior stability and recyclability to pristine enzymes. An immobilized enzyme, however, usually compromises its catalytic activity because of inevasible mass transfer issues and the unfavorable conformation changes in a confined environment. Here, we describe a synergetic metal-organic framework pore-engineering strategy to trap lipase (an important hydrolase), which confers lipase-boosted stability and activity simultaneously. The hierarchically porous NU-1003, featuring interconnected mesopore and micropore channels, is precisely modified by chain-adjustable fatty acids on its mesopore channel, into which lipase is trapped. The interconnected pore structure ensures efficient communication between trapped lipase and exterior media, while the fatty acid-mediated hydrophobic pore can activate the opening conformation of lipase by interfacial interaction. Such dual pore compartmentalization and hydrophobization activation effects render the catalytic center of trapped lipase highly accessible, resulting in 1.57-fold and 2.46-fold activities as native lipase on ester hydrolysis and enantioselective catalysis. In addition, the feasibility of these heterogeneous biocatalysts for kinetic resolution of enantiomer is also validated, showing much higher efficiency than native lipase.


Asunto(s)
Estabilidad de Enzimas , Enzimas Inmovilizadas , Interacciones Hidrofóbicas e Hidrofílicas , Lipasa , Lipasa/química , Lipasa/metabolismo , Porosidad , Enzimas Inmovilizadas/química , Enzimas Inmovilizadas/metabolismo , Estructuras Metalorgánicas/química , Hidrólisis , Biocatálisis
11.
J Am Chem Soc ; 146(1): 858-867, 2024 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-38159294

RESUMEN

Recombinant enzymes have gained prominence due to their diverse functionalities and specificity and are often a greener alternative in biocatalysis. This context makes purifying recombinant enzymes from host cells and other impurities crucial. The primary goal is to isolate the pure enzyme of interest and ensure its stability under ambient conditions. Covalent organic frameworks (COFs), renowned for their well-ordered structure and permeability, offer a promising approach for purifying histidine-tagged (His-tagged) enzymes. Furthermore, immobilizing enzymes within COFs represents a growing field in heterogeneous biocatalysis. In this study, we have developed a flow-based technology utilizing a nickel-infused covalent organic framework (Ni-TpBpy COF) to combine two distinct processes: the purification of His-tagged enzymes and the immobilization of enzymes simultaneously. Our work primarily focuses on the purification of three His-tagged enzymes ß-glucosidase, cellobiohydrolase, and endoglucanase as well as two proteins with varying molecular weights, namely, green fluorescent protein (27 kDa) and BG Rho (88 kDa). We employed Ni-TpBpy as a column matrix to showcase the versatility of our system. Additionally, we successfully obtained a Ni-TpBpy COF immobilized with enzymes, which can serve as a heterogeneous catalyst for the hydrolysis of p-nitrophenyl-ß-d-glucopyranoside and carboxymethylcellulose. These immobilized enzymes demonstrated catalytic activity comparable to that of their free counterparts, with the added advantages of recyclability and enhanced stability under ambient conditions for an extended period, ranging from 60 to 90 days. This contrasts with the free enzymes, which do not maintain their activity as effectively over time.


Asunto(s)
Estructuras Metalorgánicas , Estructuras Metalorgánicas/química , Biocatálisis , Enzimas Inmovilizadas/química , Indicadores y Reactivos , Catálisis
12.
J Am Chem Soc ; 146(28): 18817-18822, 2024 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-38968608

RESUMEN

NAD(H)-dependent enzymes play a crucial role in the biosynthesis of pharmaceuticals and fine chemicals, but the limited recyclability of the NAD(H) cofactor hinders its more general application. Here, we report the generation of mechano-responsive PEI-modified Cry3Aa protein crystals and their use for NADH recycling over multiple reaction cycles. For demonstration of its practical utility, a complementary Cry3Aa protein particle containing genetically encoded and co-immobilized formate dehydrogenase for NADH regeneration and leucine dehydrogenase for catalyzing the NADH-dependent l-tert-leucine (l-tert-Leu) biosynthesis has been produced. When combined with the PEI-modified Cry3Aa crystal, the resultant reaction system could be used for the efficient biosynthesis of l-tert-Leu for up to 21 days with a 10.5-fold improvement in the NADH turnover number.


Asunto(s)
Formiato Deshidrogenasas , NAD , NAD/metabolismo , NAD/química , Formiato Deshidrogenasas/metabolismo , Formiato Deshidrogenasas/química , Leucina-Deshidrogenasa/metabolismo , Leucina-Deshidrogenasa/química , Cristalización , Enzimas Inmovilizadas/química , Enzimas Inmovilizadas/metabolismo , Modelos Moleculares
13.
Anal Chem ; 96(22): 9228-9235, 2024 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-38779801

RESUMEN

Open-tubular immobilized enzyme microreactors (OT-IMERs) are some of the most widely used enzyme reaction devices due to the advantages of simple preparation and fast sample processing. However, the traditional approaches for OT-IMERs preparation had some defects such as limited enzyme loading amount, susceptibility to complex sample interference, and less stability. Here, we report a strategy for the preparation of highly active and stable OT-IMERs, in which the single-stranded DNA-enzyme composites were immobilized in capillaries and then encapsulated in situ in the capillaries via zeolitic imidazolate frameworks (ZIF-L). The phosphate groups of the DNA adjusted the surface potential of the enzyme to negative values, which could attract cations, such as Zn2+, to promote the formation of ZIF-L for enzyme encapsulation. Using chymotrypsin (ChT) as a model enzyme, the prepared ChT@ZIF-L-IMER has higher activity and better affinity than the free enzyme and ChT-IMER. Moreover, the thermal stability, pH stability, and organic solvent stability of ChT@ZIF-L-IMER were much higher than those of free enzyme and ChT-IMER. Furthermore, the activity of ChT@ZIF-L-IMER was much higher than that of ChT-IMER after ten consecutive reactions. To demonstrate the versatility of this preparation method, we replaced ChT with glucose oxidase (GOx). The stability of GOx@ZIF-L-IMER was also experimentally demonstrated to be superior to that of GOx and GOx-IMER. Finally, ChT@ZIF-L-IMER was used for proteolytic digestion analysis. The results showed that ChT@ZIF-L-IMER had a short digestion time and high digestive efficiency compared with the free enzyme. The present study broadened the synthesis method of OT-IMERs, effectively integrating the advantages of metal-organic frameworks and IMER, and the prepared OT-IMERs significantly improved enzyme stability. All of the results indicated that the IMER prepared by this method had a broad application prospect in capillary electrophoresis-based high-performance enzyme analysis.


Asunto(s)
Quimotripsina , Estabilidad de Enzimas , Enzimas Inmovilizadas , Imidazoles , Zeolitas , Enzimas Inmovilizadas/química , Enzimas Inmovilizadas/metabolismo , Zeolitas/química , Imidazoles/química , Quimotripsina/metabolismo , Quimotripsina/química , Estructuras Metalorgánicas/química , Concentración de Iones de Hidrógeno
14.
Anal Chem ; 96(28): 11549-11556, 2024 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-38958207

RESUMEN

Human-borne acetone is a potent marker of lipid metabolism. Here, an enzyme immobilization method for secondary alcohol dehydrogenase (S-ADH), which is suitable for highly sensitive and selective biosensing of acetone, was developed, and then its applicability was demonstrated for spatiotemporal imaging of concentration distribution. After various investigations, S-ADH-immobilized meshes could be prepared with less than 5% variation by cross-linking S-ADH with glutaraldehyde on a cotton mesh at 40 °C for 15 min. Furthermore, high activity was obtained by adjusting the concentration of the coenzyme nicotinamide adenine dinucleotide (NADH) solution added to the S-ADH-immobilized mesh to 500 µM and the solvent to a potassium phosphate buffer solution at pH 6.5. The gas imaging system using the S-ADH-immobilized mesh was able to image the decrease in NADH fluorescence (ex 340 nm, fl 490 nm) caused by the catalytic reaction of S-ADH and the acetone distribution in the concentration range of 0.1-10 ppm-v, including the breath concentration of healthy people at rest. The exhaled breath of two healthy subjects at 6 h of fasting was quantified as 377 and 673 ppb-v, which were consistent with the values quantified by gas chromatography-mass spectrometry.


Asunto(s)
Acetona , Pruebas Respiratorias , Enzimas Inmovilizadas , Acetona/análisis , Acetona/química , Humanos , Enzimas Inmovilizadas/química , Enzimas Inmovilizadas/metabolismo , Técnicas Biosensibles , Oxidorreductasas de Alcohol/química , Oxidorreductasas de Alcohol/metabolismo , Gases/química , Gases/análisis , Espiración , NAD/análisis , NAD/química , NAD/metabolismo
15.
Anal Chem ; 96(10): 4076-4085, 2024 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-38408165

RESUMEN

In this work, direct electron transfer (DET)-type extended gate field effect transistor (EGFET) enzymatic sensors were developed by employing DET-type or quasi-DET-type enzymes to detect glucose or lactate in both 100 mM potassium phosphate buffer and artificial sweat. The system employed either a DET-type glucose dehydrogenase or a quasi-DET-type lactate oxidase, the latter of which was a mutant enzyme with suppressed oxidase activity and modified with amine-reactive phenazine ethosulfate. These enzymes were immobilized on the extended gate electrodes. Changes in the measured transistor drain current (ID) resulting from changes to the working electrode junction potential (φ) were observed as glucose and lactate concentrations were varied. Calibration curves were generated for both absolute measured ID and ΔID (normalized to a blank solution containing no substrate) to account for variations in enzyme immobilization and conjugation to the mediator and variations in reference electrode potential. This work resulted in a limit of detection of 53.9 µM (based on ID) for glucose and 2.12 mM (based on ID) for lactate, respectively. The DET-type and Quasi-DET-type EGFET enzymatic sensor was then modeled using the case of the lactate sensor as an equivalent circuit to validate the principle of sensor operation being driven through OCP changes caused by the substrate-enzyme interaction. The model showed slight deviation from collected empirical data with 7.3% error for the slope and 8.6% error for the y-intercept.


Asunto(s)
Técnicas Biosensibles , Electrones , Técnicas Biosensibles/métodos , Glucosa/metabolismo , Glucosa 1-Deshidrogenasa/metabolismo , Ácido Láctico , Enzimas Inmovilizadas/metabolismo , Electrodos
16.
Small ; 20(30): e2312220, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38344893

RESUMEN

Enzymatic activity depends on and can therefore be regulated by temperature. Selective modulation of the activity of different enzymes in one reaction pot would require temperature control local to each type of enzyme. It has been suggested previously that immobilization of enzyme on magnetic nanoparticles and exposing them to alternating magnetic field can enhance the reaction rate. This enhancement has been explained as being mediated by temperature increase caused by dissipation of the absorbed field energy in the form of heat. However, the possibility of spatially limiting this temperature increase on the microscale has been questioned. Here, it is investigated whether an activity enhancement of the enzyme sucrose phosphorylase immobilized on magnetic beads can be achieved, how this effect is related to the increase in temperature, and whether temperature differences within one reaction pot could be generated in this way. It is found that alternating magnetic field stimulation leads to increased enzymatic activity fully attributable to the increase of bulk temperature. Both theoretical analysis and experimental data indicate that no local heating near the particle surface takes place. It is further concluded that relevant increase of surface temperature can be obtained only with macroscopic, millimeter-sized, magnetic particles.


Asunto(s)
Activación Enzimática , Temperatura , Enzimas Inmovilizadas/química , Enzimas Inmovilizadas/metabolismo , Campos Magnéticos , Glucosiltransferasas/metabolismo
17.
Chembiochem ; 25(11): e202400086, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38618870

RESUMEN

Sustainable biocatalysis syntheses have gained considerable popularity over the years. However, further optimizations - notably to reduce costs - are required if the methods are to be successfully deployed in a range of areas. As part of this drive, various enzyme immobilization strategies have been studied, alongside process intensification from batch to continuous production. The flow bioreactor portfolio mainly ranges between packed bed reactors and wall-immobilized enzyme miniaturized reactors. Because of their simplicity, packed bed reactors are the most frequently encountered at lab-scale. However, at industrial scale, the growing pressure drop induced by the increase in equipment size hampers their implementation for some applications. Wall-immobilized miniaturized reactors require less pumping power, but a new problem arises due to their reduced enzyme-loading capacity. This review starts with a presentation of the current technology portfolio and a reminder of the metrics to be applied with flow bioreactors. Then, a benchmarking of the most recent relevant works is presented. The scale-up perspectives of the various options are presented in detail, highlighting key features of industrial requirements. One of the main objectives of this review is to clarify the strategies on which future study should center to maximize the performance of wall-immobilized enzyme reactors.


Asunto(s)
Biocatálisis , Reactores Biológicos , Enzimas Inmovilizadas , Enzimas Inmovilizadas/química , Enzimas Inmovilizadas/metabolismo , Miniaturización
18.
Chembiochem ; 25(4): e202300843, 2024 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-38169079

RESUMEN

Using lipases to catalyze the synthesis of the most differentiated type of compounds remains one of the major challenges among scientists. Seeking more economic and advantageous catalysts is a current goal of green chemistry. In this work, we demonstrate the potential of a chemically modified form of lipase from Thermomyces lanuginosus (cmLTL) for the synthesis of both hydrophobic (heptyl heptanoate, heptyl octanoate, heptyl decanoate, decyl heptanoate, decyl octanoate and decyl decanoate) and amphiphilic (2-(2-ethoxyethoxy)ethyl oleate and 2-(2-ethoxyethoxy)ethyl linoleate) esters, in bulk. The results were compared with its native (LTL) and immobilized (imLTL) forms. The data revealed that LTL showed poor activity for all reactions performed with n-heptane (η<20 %). ImLTL was able to synthesize all hydrophobic esters (η>60 %), with exception of the short ester, heptyl heptanoate. cmLTL was the only form of LTL capable of producing hydrophobic and amphiphilic esters, without compromising the yield when the reactions were performed under solvent-free conditions (>50 %). Molecular modeling showed that the active pocket of cmLTL is able to deeply internalize transcutol, with stronger interactions, justifying the outstanding results obtained. Furthermore, owing to the possibility of cmLTL filtration, the reusability of the catalyst is ensured for at least 6 cycles, without compromising the reaction yields.


Asunto(s)
Ésteres , Eurotiales , Lipasa , Solventes , Esterificación , Lipasa/química , Decanoatos , Heptanoatos , Enzimas Inmovilizadas/metabolismo
19.
Chembiochem ; 25(12): e202400147, 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38629211

RESUMEN

Chiral alcohols are not only important building blocks of various bioactive natural compounds and pharmaceuticals, but can serve as synthetic precursors for other valuable organic chemicals, thus the synthesis of these products is of great importance. Bio-catalysis represents one effective way to obtain these molecules, however, the weak stability and high cost of enzymes often hinder its broad application. In this work, we designed a biological nanoreactor by embedding alcohol dehydrogenase (ADH) and glucose dehydrogenase (GDH) in metal-organic-framework ZIF-8. The biocatalyst ADH&GDH@ZIF-8 could be applied to the asymmetric reduction of a series of ketones to give chiral alcohols in high yields (up to 99 %) and with excellent enantioselectivities (>99 %). In addition, the heterogeneous biocatalyst could be recycled and reused at least four times with slight activity decline. Moreover, E. coli containing ADH and GDH was immobilized by ZIF-8 to form biocatalyst E. coli@ZIF-8, which also exhibits good catalytic behaviours. Finally, the chiral alcohols are further converted to marketed drugs (R)-Fendiline, (S)-Rivastigmine and NPS R-568 respectively.


Asunto(s)
Alcohol Deshidrogenasa , Biocatálisis , Enzimas Inmovilizadas , Escherichia coli , Glucosa 1-Deshidrogenasa , Cetonas , Estructuras Metalorgánicas , Alcohol Deshidrogenasa/metabolismo , Alcohol Deshidrogenasa/química , Estructuras Metalorgánicas/química , Estructuras Metalorgánicas/metabolismo , Cetonas/química , Cetonas/metabolismo , Enzimas Inmovilizadas/metabolismo , Enzimas Inmovilizadas/química , Escherichia coli/enzimología , Escherichia coli/metabolismo , Glucosa 1-Deshidrogenasa/metabolismo , Glucosa 1-Deshidrogenasa/química , Oxidación-Reducción , Estereoisomerismo
20.
Chembiochem ; 25(15): e202400346, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38775416

RESUMEN

Multi-enzyme cascade catalysis has become an important technique for chemical reactions used in manufacturing and scientific study. In this research, we designed a four-enzyme integrated catalyst and used it to catalyse the deracemization reaction of cyclic chiral amines, where monoamine oxidase (MAO) catalyses the enantioselective oxidation of 1-methyl-1,2,3,4-tetrahydroisoquinoline (MTQ), imine reductase (IRED) catalyses the stereo selective reduction of 1-methyl-3,4-dihydroisoquinoline (MDQ), formate dehydrogenase (FDH) is used for the cyclic regeneration of cofactors, and catalase (CAT) is used for decomposition of oxidative reactions. The four enzymes were immobilized via polydopamine (PDA)-encapsulated dendritic organosilica nanoparticles (DONs) as carriers, resulting in the amphiphilic core-shell catalysts. The hydrophilic PDA shell ensures the dispersion of the catalyst in water, and the hydrophobic DON core creates a microenvironment with the spatial confinement effect of the organic substrate and the preconcentration effect to enhance the stability of the enzymes and the catalytic efficiency. The core-shell structure improves the stability and reusability of the catalyst and rationally arranges the position of different enzymes according to the reaction sequence to improve the cascade catalytic performance and cofactor recovery efficiency.


Asunto(s)
Aminas , Monoaminooxidasa , Polímeros , Aminas/química , Aminas/metabolismo , Monoaminooxidasa/metabolismo , Monoaminooxidasa/química , Polímeros/química , Polímeros/metabolismo , Formiato Deshidrogenasas/metabolismo , Formiato Deshidrogenasas/química , Catalasa/química , Catalasa/metabolismo , Indoles/química , Indoles/metabolismo , Estereoisomerismo , Enzimas Inmovilizadas/química , Enzimas Inmovilizadas/metabolismo , Oxidación-Reducción , Nanopartículas/química , Biocatálisis , Compuestos de Organosilicio/química , Oxidorreductasas/metabolismo , Oxidorreductasas/química , Catálisis
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda