Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.128
Filter
1.
Nat Methods ; 21(7): 1340-1348, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38918604

ABSTRACT

The EMDataResource Ligand Model Challenge aimed to assess the reliability and reproducibility of modeling ligands bound to protein and protein-nucleic acid complexes in cryogenic electron microscopy (cryo-EM) maps determined at near-atomic (1.9-2.5 Å) resolution. Three published maps were selected as targets: Escherichia coli beta-galactosidase with inhibitor, SARS-CoV-2 virus RNA-dependent RNA polymerase with covalently bound nucleotide analog and SARS-CoV-2 virus ion channel ORF3a with bound lipid. Sixty-one models were submitted from 17 independent research groups, each with supporting workflow details. The quality of submitted ligand models and surrounding atoms were analyzed by visual inspection and quantification of local map quality, model-to-map fit, geometry, energetics and contact scores. A composite rather than a single score was needed to assess macromolecule+ligand model quality. These observations lead us to recommend best practices for assessing cryo-EM structures of liganded macromolecules reported at near-atomic resolution.


Subject(s)
Cryoelectron Microscopy , Models, Molecular , Cryoelectron Microscopy/methods , Ligands , SARS-CoV-2 , COVID-19/virology , Escherichia coli , beta-Galactosidase/chemistry , beta-Galactosidase/metabolism , Protein Conformation , Reproducibility of Results
2.
Int J Biol Macromol ; 273(Pt 1): 132992, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38857718

ABSTRACT

ß-D-galactosidase is a hydrolase enzyme capable of hydrolyzing lactose in milk-based foods. Its free form can be inactivated in solution during the production of low-dosage lactose foods. Then, it is important to study strategies for avoiding the free enzyme inactivation with the aim of circumventing this problem. The stabilization of ß-D-galactosidase in aqueous solution after interactions with chitosan/eucalyptus sawdust composite membrane proved to be a potential strategy when optimized by central composite rotatable (CCR) design. In this case, the best experimental conditions for ß-D-galactosidase partitioning and stability in an aqueous medium containing the chitosan-based composite membrane reinforced with eucalyptus sawdust were i) enzyme/buffer solution ratio of 0.0057, ii) pH 5.6, iii) membrane mass of 50 mg, and iv) temperature lower than 37 °C. Significance was found for the linear enzyme/buffer solution ratio, linear temperature, and quadratic pH (p < 0.05) in the interval between 0 and 60 min of study. In the interval between 60 and 120 min, there was significance (p < 0.12) for linear temperature, the temperature-enzyme/buffer solution ratio interaction and the interaction between linear pH and linear enzyme/buffer solution ratio. The Pareto charts and response surfaces clearly showed all the effects of the experimental variables on the stabilization of ß-D-galactosidase in solution after interactions with the chitosan composite membrane. In this case, industrial food reactors covered with chitosan/eucalyptus sawdust composite membrane could be a strategy for the hydrolysis of lactose during milk-producing processes.


Subject(s)
Chitosan , Enzyme Stability , beta-Galactosidase , Chitosan/chemistry , beta-Galactosidase/chemistry , beta-Galactosidase/metabolism , Hydrogen-Ion Concentration , Membranes, Artificial , Solutions , Temperature , Lactose/chemistry
3.
Biomacromolecules ; 25(5): 3055-3062, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38693874

ABSTRACT

Polymersomes, nanosized polymeric vesicles, have attracted significant interest in the areas of artificial cells and nanomedicine. Given their size, their visualization via confocal microscopy techniques is often achieved through the physical incorporation of fluorescent dyes, which however present challenges due to potential leaching. A promising alternative is the incorporation of molecules with aggregation-induced emission (AIE) behavior that are capable of fluorescing exclusively in their assembled state. Here, we report on the use of AIE polymersomes as artificial organelles, which are capable of undertaking enzymatic reactions in vitro. The ability of our polymersome-based artificial organelles to provide additional functionality to living cells was evaluated by encapsulating catalytic enzymes such as a combination of glucose oxidase/horseradish peroxidase (GOx/HRP) or ß-galactosidase (ß-gal). Via the additional incorporation of a pyridinium functionality, not only the cellular uptake is improved at low concentrations but also our platform's potential to specifically target mitochondria expands.


Subject(s)
Glucose Oxidase , Horseradish Peroxidase , beta-Galactosidase , Glucose Oxidase/chemistry , Humans , beta-Galactosidase/chemistry , beta-Galactosidase/metabolism , Horseradish Peroxidase/chemistry , Horseradish Peroxidase/metabolism , Organelles/metabolism , Fluorescent Dyes/chemistry , Polymers/chemistry , Fluorescence , HeLa Cells , Mitochondria/metabolism
4.
Food Chem ; 452: 139557, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-38728895

ABSTRACT

ß-Galactosidase (ß-gal), an enzyme related to cell wall degradation, plays an important role in regulating cell wall metabolism and reconstruction. However, activatable fluorescence probes for the detection and imaging of ß-gal fluctuations in plants have been less exploited. Herein, we report an activatable fluorescent probe based on intramolecular charge transfer (ICT), benzothiazole coumarin-bearing ß-galactoside (BC-ßgal), to achieve distinct in situ imaging of ß-gal in plant cells. It exhibits high sensitivity and selectivity to ß-gal with a fast response (8 min). BC-ßgal can be used to efficiently detect the alternations of intracellular ß-gal levels in cabbage root cells with considerable imaging integrity and imaging contrast. Significantly, BC-ßgal can assess ß-gal activity in cabbage roots under heavy metal stress (Cd2+, Cu2+, and Pb2+), revealing that ß-gal activity is negatively correlated with the severity of heavy metal stress. Our work thus facilitates the study of ß-gal biological mechanisms.


Subject(s)
Brassica , Fluorescent Dyes , Metals, Heavy , Plant Roots , beta-Galactosidase , beta-Galactosidase/metabolism , beta-Galactosidase/chemistry , Brassica/chemistry , Brassica/metabolism , Brassica/enzymology , Plant Roots/chemistry , Plant Roots/metabolism , Fluorescent Dyes/chemistry , Metals, Heavy/metabolism , Metals, Heavy/analysis , Optical Imaging , Plant Proteins/metabolism
5.
Int J Biol Macromol ; 270(Pt 1): 132101, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38734354

ABSTRACT

Aspergillus oryzae ß-D-galactosidase (ß-Gal) efficiently hydrolyzes sesaminol triglucoside into sesaminol, which has higher biological activity. However, ß-Gal is difficult to be separate from the reaction mixture and limited by stability. To resolve these problems, ß-Gal was immobilized on amino-functionalized magnetic nanoparticles mesoporous silica pre-activated with glutaraldehyde (Fe3O4@mSiO2-ß-Gal), which was used for the first time to prepare sesaminol. Under the optimal conditions, the immobilization yield and recovered activity of ß-Gal were 57.9 ± 0.3 % and 46.5 ± 0.9 %, and the enzymatic loading was 843 ± 21 Uenzyme/gsupport. The construction of Fe3O4@mSiO2-ß-Gal was confirmed by various characterization methods, and the results indicated it was suitable for heterogeneous enzyme-catalyzed reactions. Fe3O4@mSiO2-ß-Gal was readily separable under magnetic action and displayed improved activity in extreme pH and temperature conditions. After 45 days of storage at 4 °C, the activity of Fe3O4@mSiO2-ß-Gal remained at 92.3 ± 2.8 %, which was 1.29 times than that of free enzyme, and its activity remained above 85 % after 10 cycles. Fe3O4@mSiO2-ß-Gal displayed higher affinity and catalytic efficiency. The half-life was 1.41 longer than free enzymes at 55.0 °C. Fe3O4@mSiO2-ß-Gal was employed as a catalyst to prepare sesaminol, achieving a 96.7 % conversion yield of sesaminol. The excellent stability and catalytic efficiency provide broad benefits and potential for biocatalytic industry applications.


Subject(s)
Aspergillus oryzae , Enzymes, Immobilized , Glutaral , Silicon Dioxide , beta-Galactosidase , Enzymes, Immobilized/chemistry , Enzymes, Immobilized/metabolism , beta-Galactosidase/chemistry , beta-Galactosidase/metabolism , Aspergillus oryzae/enzymology , Silicon Dioxide/chemistry , Glutaral/chemistry , Dioxoles/chemistry , Dioxoles/pharmacology , Magnetite Nanoparticles/chemistry , Porosity , Temperature , Hydrogen-Ion Concentration , Enzyme Stability , Furans
6.
Int J Biol Macromol ; 270(Pt 1): 132312, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38744370

ABSTRACT

This study aimed to immobilize ß-galactosidase (ß-GAL) into enhanced polystyrene (PS) electrospun nanofiber membranes (ENMs) with functionalized graphene oxide (GO). Initially, GO sheets were functionalized by salinization with 3-aminopropyl triethoxysilane (APTES). Then the ENMs (PS, PS/GO, and PS/GO-APTES) were prepared and characterized. Then, the ß-GAL was immobilized in the different ENMs to produce the ß-GAL-bound nanocomposites (PS-GAL, PS/GO-GAL, and PS/GO-APTES-GAL). Immobilization of ß-GAL into PS/GO-APTES significantly improved enzyme adsorption by up to 87 %. Also, PS/GO-APTES-GAL improved the enzyme activity, where the highest enzyme activity was obtained at enzyme concentrations of 4 mg/L, 50 °C, and pH 4.5. Likewise, the storage stability and reusability of immobilized ß-GAL were improved. Furthermore, this process led to enhanced catalytic behavior and transgalactosylation efficiency, where GOS synthesis (72 %) and lactose conversion (81 %) increased significantly compared to the free enzyme. Overall, the immobilized ß-GAL produced in this study showed potential as an effective biocatalyst in the food industry.


Subject(s)
Enzymes, Immobilized , Graphite , Nanofibers , Oligosaccharides , beta-Galactosidase , beta-Galactosidase/chemistry , beta-Galactosidase/metabolism , Enzymes, Immobilized/chemistry , Enzymes, Immobilized/metabolism , Nanofibers/chemistry , Graphite/chemistry , Oligosaccharides/chemistry , Galactose/chemistry , Hydrogen-Ion Concentration , Enzyme Stability , Silanes/chemistry , Biocatalysis , Polystyrenes/chemistry , Temperature , Catalysis
7.
Int J Pharm ; 659: 124277, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38802027

ABSTRACT

The application of 3D printing technology in the delivery of macromolecules, such as proteins and enzymes, is limited by the lack of suitable inks. In this study, we report the development of novel inks for 3D printing of constructs containing proteins while maintaining the activity of the proteins during and after printing. Different ink formulations containing Pluronic F-127 (20-35 %, w/v), trehalose (2-10 %, w/v) or mannitol, poly (ethylene glycol) diacrylate (PEGDA) (0 or 10 %, w/w), and diphenyl(2,4,6-trimethylbenzoyl) phosphine oxide (TPO, 0 or 0.2 mg/mL) were prepared for 3D-microextrusion printing. The F2 formulation that contained ß-galactosidase (ß-gal) as a model enzyme, Pluronic F-127 (30 %), and trehalose (10 %) demonstrated the desired viscosity, printability, and dose flexibility. The shear-thinning property of the F2 formulation enabled the printing of ß-gal containing constructs with a good peak force during extrusion. After 3D printing, the enzymatic activity of the ß-gal in the constructs was maintained for an extended period, depending on the construct design and storage conditions. For instance, there was a 50 % reduction in ß-gal activity in the two-layer constructs, but only a 20 % reduction in the four-layer construct (i.e., 54.5 ± 1.2 % and 82.7 ± 9.9 %, respectively), after 4 days of storage. The ß-gal activity in constructs printed from the F2 formulation was maintained for up to 20 days when stored in sealed bags at room temperatures (21 ± 2 °C), but not when stored unsealed in the same conditions (e.g., ∼60 % activity loss within 7 days). The ß-gal from constructs printed from F2 started to release within 5 min and reached 100 % after 20 min. With the design flexibility offered by the 3D printing, the ß-gal release from the constructs was delayed to 3 h by printing a backing layer of ß-gal-free F5 ink on the constructs printed from the F2 ink. Finally, ovalbumin as an alternative protein was also incorporated in similar ink compositions. Ovalbumin exhibited a release profile like that of the ß-gal, and the release can also be modified with different shape design and/or ink composition. In conclusion, ink formulations that possess desirable properties for 3D printing of protein-containing constructs while maintaining the protein activity during and after printing were developed.


Subject(s)
Ink , Poloxamer , Polyethylene Glycols , Printing, Three-Dimensional , Trehalose , beta-Galactosidase , beta-Galactosidase/chemistry , Poloxamer/chemistry , Polyethylene Glycols/chemistry , Trehalose/chemistry , Viscosity , Excipients/chemistry , Drug Delivery Systems/methods , Mannitol/chemistry , Technology, Pharmaceutical/methods , Phosphines/chemistry
8.
Biosens Bioelectron ; 259: 116369, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38781695

ABSTRACT

Accurate and effective detection is essential to against bacterial infection and contamination. Novel biosensors, which detect bacterial bioproducts and convert them into measurable signals, are attracting attention. We developed an artificial intelligence (AI)-assisted smartphone-based colorimetric biosensor for the visualized, rapid, sensitive detection of pathogenic bacteria by measuring the bacteria secreted hyaluronidase (HAase). The biosensor consists of the chlorophenol red-ß-D-galactopyranoside (CPRG)-loaded hyaluronic acid (HA) hydrogel as the bioreactor and the ß-galactosidase (ß-gal)-loaded agar hydrogel as the signal generator. The HAase degrades the bioreactor and subsequently determines the release of CPRG, which could further react with ß-gal to generate signal colors. The self-developed YOLOv5 algorithm was utilized to analyze the signal colors acquired by smartphone. The biosensor can provide a report within 60 min with an ultra-low limit of detection (LoD) of 10 CFU/mL and differentiate between gram-positive (G+) and gram-negative (G-) bacteria. The proposed biosensor was successfully applied in various areas, especially the evaluation of infections in clinical samples with 100% sensitivity. We believe the designed biosensor has the potential to represent a new paradigm of "ASSURED" bacterial detection, applicable for broad biomedical uses.


Subject(s)
Biosensing Techniques , Colorimetry , Smartphone , Biosensing Techniques/methods , Biosensing Techniques/instrumentation , Colorimetry/instrumentation , Limit of Detection , Humans , Artificial Intelligence , beta-Galactosidase/chemistry , Bacteria/isolation & purification
9.
J Biotechnol ; 388: 35-48, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38641136

ABSTRACT

Whey protein isolate (WPI) was incorporated within calcium pectinate (CPT) beads in order to boost their anionic qualities and meliorate their glutaraldehyde (GA)-polyethyleneimine (PEI) grafting process. The Box-Behnken Design (BBD) verified that WPI inclusion significantly raised the GA-PEI-CPT-WPI beads immobilized ß-D-galactosidase (iß-GLD) activity. The BBD also revealed the optimal settings for WPI concentration, PEI pH, PEI concentration, and GA concentration, which were 2.91 %, 10.8, 3.5 %, and 2.24 %, respectively. The GA-PEI-CPT-WPI beads grafting process was scrutinized via FTIR, EDX, and SEM. The optimal GA-PEI-CPT-WPI immobilizers provided fine ß-GLD immobilization efficiencies, which reached up to 65.28 %. The free and GA-PEI-CPT-WPI iß-GLDs pH and temperature profiles were scrutinized. It was also unveiled that the thermal stability of the iß-GLD surpassed that of its free compeer as it provided lesser kd and ΔS values and larger t1/2, D-values, Ed, ΔH, and ΔG values. Furthermore, the iß-GLD provided 92.00±3.39 % activity after 42 storage days, which denoted its fine storage stability. The iß-GLD short duration (15 min) operational stability was also inspected, and 82.70±0.78 % activity was provided during the fifteenth degradation run. Moreover, the iß-GLD long duration (24 h) operational stability was inspected while degrading the lactose of buffered lactose solution (BLS) and cheese whey (CW). It was unveiled that 81.86±0.96 % and 73.58±2.24 % of the initial glucose were detected during the sixth degradation runs, respectively.


Subject(s)
Enzymes, Immobilized , Polyethyleneimine , Thermodynamics , Whey Proteins , Enzymes, Immobilized/chemistry , Enzymes, Immobilized/metabolism , Whey Proteins/chemistry , Kinetics , Polyethyleneimine/chemistry , Hydrogen-Ion Concentration , Pectins/chemistry , Pectins/metabolism , beta-Galactosidase/metabolism , beta-Galactosidase/chemistry , Glutaral/chemistry , Temperature , Enzyme Stability
10.
Bioprocess Biosyst Eng ; 47(6): 919-929, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38644439

ABSTRACT

The growing need in the current market for innovative solutions to obtain lactose-free (L-F) milk is caused by the annual increase in the prevalence of lactose intolerance inside as well as the newborn, children, and adults. Various configurations of enzymes can yield two distinct L-F products: sweet (ß-galactosidase) and unsweet (ß-galactosidase and glucose oxidase) L-F milk. In addition, the reduction of sweetness through glucose decomposition should be performed in a one-pot mode with catalase to eliminate product inhibition caused by H2O2. Both L-F products enjoy popularity among a rapidly expanding group of consumers. Although enzyme immobilization techniques are well known in industrial processes, new carriers and economic strategies are still being searched. Polymeric carriers, due to the variety of functional groups and non-toxicity, are attractive propositions for individual and co-immobilization of food enzymes. In the presented work, two strategies (with free and immobilized enzymes; ß-galactosidase NOLA, glucose oxidase from Aspergillus niger, and catalase from Serratia sp.) for obtaining sweet and unsweet L-F milk under low-temperature conditions were proposed. For free enzymes, achieving the critical assumption, lactose hydrolysis and glucose decomposition occurred after 1 and 4.3 h, respectively. The tested catalytic membranes were created on regenerated cellulose and polyamide. In both cases, the time required for lactose and glucose bioconversion was extended compared to free enzymes. However, these preparations could be reused for up to five (ß-galactosidase) and ten cycles (glucose oxidase with catalase).


Subject(s)
Enzymes, Immobilized , Glucose Oxidase , Lactose , Milk , beta-Galactosidase , beta-Galactosidase/metabolism , beta-Galactosidase/chemistry , Milk/chemistry , Lactose/metabolism , Lactose/chemistry , Glucose Oxidase/chemistry , Glucose Oxidase/metabolism , Enzymes, Immobilized/chemistry , Enzymes, Immobilized/metabolism , Animals , Aspergillus niger/enzymology , Glucose/metabolism , Glucose/chemistry , Catalase/metabolism , Catalase/chemistry , Membranes, Artificial
11.
Food Chem ; 450: 139331, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-38621310

ABSTRACT

The sensitive detection of foodborne pathogenic and rapid antibiotic susceptibility testing (AST) is of great significance. This paper reports the enzyme-triggered in situ synthesis of yellow emitting silicon nanoparticles (SiNPs) and the detection of Escherichia coli (E. coli) O157:H7 in food samples and the rapid AST. The rapid counting of E. coli O157:H7 has been achieved through direct visual observation, equipment detection, and smartphone digitalization. A simple detection platform based on smartphone senses and cotton swabs has been established. Meanwhile, rapid AST based on enzyme-catalyzed SiNPs can intuitively obtain colorimetric samples. This paper established a system for bacterial enzyme-triggered in situ synthesis of SiNPs, with high responsiveness, luminescence ratio, and specificity. The detection limit for E. coli O157:H7 can reach 100 CFU/mL during 5 h, and the recovery efficiency ranges from 90.14% to 110.16%, which makes it a promising strategy for the rapid detection of E. coli O157:H7 and AST.


Subject(s)
Escherichia coli O157 , Nanoparticles , Silicon , beta-Galactosidase , Escherichia coli O157/drug effects , Escherichia coli O157/isolation & purification , Nanoparticles/chemistry , Silicon/chemistry , Silicon/pharmacology , beta-Galactosidase/metabolism , beta-Galactosidase/chemistry , Microbial Sensitivity Tests , Food Contamination/analysis , Colorimetry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Food Microbiology
12.
Int J Biol Macromol ; 268(Pt 2): 131766, 2024 May.
Article in English | MEDLINE | ID: mdl-38657932

ABSTRACT

The biological function of terminal galactose on glycoprotein is an open field of research. Although progress had being made on enzymes that can remove the terminal galactose on glycoproteins, there is a lack of report on galactosidases that can work directly on living cells. In this study, a unique beta 1,4 galactosidase was isolated from Elizabethkingia meningoseptica (Em). It exhibited favorable stability at various temperatures (4-37 °C) and pH (5-8) levels and can remove ß-1, 4 linked galactoses directly from glycoproteins. Using Alanine scanning, we found that two acidic residues (Glu-468, and Glu-531) in the predicted active pocket are critical for galactosidase activity. In addition, we also demonstrated that it could cleave galactose residues present on living cell surface. As this enzyme has a potential application for living cell glycan editing, we named it emGalaseE or glycan-editing galactosidase I (csgeGalaseI). In summary, our findings lay the groundwork for further investigation by presenting a simple and effective approach for the removal of galactose moieties from cell surface.


Subject(s)
Flavobacteriaceae , Galactose , Flavobacteriaceae/enzymology , Galactose/metabolism , Galactose/chemistry , Hydrogen-Ion Concentration , Amino Acid Sequence , Enzyme Stability , Cell Membrane/metabolism , Galactosidases/metabolism , Galactosidases/chemistry , beta-Galactosidase/metabolism , beta-Galactosidase/chemistry , Temperature , Substrate Specificity
13.
J Sci Food Agric ; 104(11): 6769-6777, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-38563403

ABSTRACT

BACKGROUND: The general assumption that prebiotics reach the colon without any alterations has been challenged. Some in vitro and in vivo studies have demonstrated that 'non-digestible' oligosaccharides are digested to different degrees depending on their structural composition. In the present study, we compared different methods aiming to assess the digestibility of oligosaccharides synthesized by ß-galactosidase (ß-gal) of Lactobacillus delbruecki subsp. bulgaricus CRL450 (CRL450-ß-gal) from lactose, lactulose and lactitol. RESULTS: In the simulated gastrointestinal fluid method, no changes were observed. However, the oligosaccharides synthesized by CRL450-ß-gal were partially hydrolyzed in vitro, depending on their structure and composition, with rat small intestinal extract (RSIE) and small intestinal brush-border membrane vesicles (BBMV) from pig. Digestion of some oligosaccharides increased when mixtures were fed to C57BL/6 mice used as in vivo model; however, lactulose-oligosaccharides were the most resistant to the physiological conditions of mice. In general ß (1→6) linked products showed higher resistance compared to ß (1→3) oligosaccharides. CONCLUSION: In vitro digestion methods, without disaccharidases, may underestimate the importance of carbohydrates hydrolysis in the small intestine. Although BVMM and RSIE digestion assays are appropriate in vitro methods for these studies, in vivo studies remain the most reliable for understanding what actually happens in the digestion of oligosaccharides. © 2024 The Authors. Journal of The Science of Food and Agriculture published by John Wiley & Sons Ltd on behalf of Society of Chemical Industry.


Subject(s)
Digestion , Mice, Inbred C57BL , Oligosaccharides , Prebiotics , beta-Galactosidase , Prebiotics/analysis , Animals , beta-Galactosidase/metabolism , beta-Galactosidase/chemistry , Oligosaccharides/metabolism , Oligosaccharides/chemistry , Mice , Rats , Swine , Male , Lactulose/metabolism , Lactulose/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Intestine, Small/metabolism , Intestine, Small/enzymology , Lactobacillus/metabolism , Lactobacillus/enzymology , Hydrolysis , Lactose/metabolism , Lactose/chemistry
14.
Biosens Bioelectron ; 255: 116207, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38554575

ABSTRACT

Near-infrared (NIR) aggregation induced-emission luminogens (AIEgens) circumvent the noisome aggregation-caused quenching (ACQ) effect in physiological milieu, thus holding high promise for real-time and sensitive imaging of biomarkers in vivo. ß-Galactosidase (ß-Gal) is a biomarker for primary ovarian carcinoma, but current AIEgens for ß-Gal sensing display emissions in the visible region and have not been applied in vivo. We herein propose an NIR AIEgen QM-TPA-Gal and applied it for imaging ß-Gal activity in vitro and in ovarian tumor model. After being internalized by ovarian cancer cells (e.g., SKOV3), the hydrophilic nonfluorescent QM-TPA-Gal undergoes hydrolyzation by ß-Gal to yield hydrophobic QM-TPA-OH, which subsequently aggregates into nanoparticles to turn NIR fluorescence "on" through the AIE mechanism. In vitro experimental results indicate that QM-TPA-Gal has a sensitive and selective response to ß-Gal with a limit of detection (LOD) of 0.21 U/mL. Molecular docking simulation confirms that QM-TPA-Gal has a good binding ability with ß-Gal to allow efficient hydrolysis. Furthermore, QM-TPA-Gal is successfully applied for ß-Gal imaging in SKOV3 cell and SKOV3-bearing living mouse models. It is anticipated that QM-TPA-Gal could be applied for early diagnosis of ovarian cancers or other ß-Gal-associated diseases in near future.


Subject(s)
Biosensing Techniques , Ovarian Neoplasms , Animals , Humans , Mice , Female , Fluorescent Dyes/chemistry , Molecular Docking Simulation , Ovarian Neoplasms/diagnostic imaging , Optical Imaging , beta-Galactosidase/chemistry , beta-Galactosidase/metabolism
15.
Food Chem ; 448: 139082, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38537544

ABSTRACT

ß-galactosidase (lactase) is commercially important as a dietary supplement to alleviate the symptoms of lactose intolerance. This work investigated a unique activation of CMP (carboxymethylated (1 â†’ 3)-ß-d-glucan) on lactase and its mechanism by comparing it with carboxymethyl chitosan (CMCS), an inhibitor of lactase. The results illustrated that the secondary and tertiary structures of lactase were altered and its active sites exposed after complexation with CMP, and dissociation of lactase aggregates was also observed. These changes favored better accessibility of the substrate to the active sites of lactase, resulting in a maximum increase of 60.5 % in lactase activity. Furthermore, the hydrophobic and electrostatic interactions with lactase caused by the carboxymethyl group of CMP were shown to be crucial for its activation ability. Thus, the improvement of lactase activity and stability by CMP shown here is important for the development of new products in the food and pharmaceutical industries.


Subject(s)
Chitosan/analogs & derivatives , Hydrophobic and Hydrophilic Interactions , Static Electricity , beta-Galactosidase , beta-Glucans , beta-Galactosidase/chemistry , beta-Galactosidase/metabolism , beta-Glucans/chemistry , beta-Glucans/pharmacology , Chitosan/chemistry , Enzyme Stability , Kinetics , Enzyme Activation/drug effects
16.
Talanta ; 274: 125973, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38537359

ABSTRACT

Sensitive detection of copper ion (Cu2+), which is of great importance for environmental pollution and human health, is crucial. In this study, we present a highly sensitive method for measuring Cu2+ in an array of femtoliter wells. In brief, magnetic beads (MBs) modified with alkyne groups were bound to the azide groups of biotin-PEG3-azide (bio-PEG-N3) via Cu+-catalyzed click chemistry. Cu+ in the click chemistry reaction was generated by reducing Cu2+ with sodium ascorbate. Following the ligation, the surface of the MBs was modified with biotin, which could be labeled with streptavidin-ß-galactosidase (SßG). The MBs complex was then suspended in ß-galactosidase substrate fluorescein-di-ß-d-galactopyranoside (FDG), and loaded into the array of femtoliter wells. The MBs sank into the wells due to gravity, and the resulting fluorescent product, generated from the reaction between SßG on the surface of the MBs and FDG, was confined within the wells. The number of fluorescent wells increased with higher Cu2+ concentrations. The bright-field and fluorescent images of the wells were acquired using an inverted fluorescent microscope. The detection limit of this assay for Cu2+ was 1 nM without signal amplification, which was 103 times lower than that of traditional fluorescence detection assays.


Subject(s)
Azides , Click Chemistry , Copper , Copper/chemistry , Copper/analysis , Azides/chemistry , Limit of Detection , Biotin/chemistry , Polyethylene Glycols/chemistry , Streptavidin/chemistry , beta-Galactosidase/metabolism , beta-Galactosidase/chemistry , beta-Galactosidase/analysis
17.
Bioprocess Biosyst Eng ; 47(2): 249-261, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38197955

ABSTRACT

ß-galactosidase has been immobilized onto novel alginate/tea waste gel beads (Alg/TW) via covalent binding. Alg/TW beads were subjected to chemical modification through amination with polyethyleneimine (PEI) followed by activation with glutaraldehyde (GA). Chemical modification parameters including PEI concentration, PEI pH, and GA concentration were statistically optimized using Response Surface methodology (RSM) based on Box-Behnken Design (BBD). Analysis of variance (ANOVA) results confirmed the great significance of the model that had F value of 37.26 and P value < 0.05. Furthermore, the R2 value (0.9882), Adjusted R2 value (0.9617), and predicted R2 value (0.8130) referred to the high correlation between predicted and experimental values, demonstrating the fitness of the model. In addition, the coefficient of variation (CV) value was 2.90 that pointed to the accuracy of the experiments. The highest immobilization yield (IY) of ß-galactosidase (75.1%) was given under optimized conditions of PEI concentration (4%), PEI pH (9.5), and GA concentration (2.5%). Alg/TW beads were characterized by FT-IR, TGA, and SEM techniques at each step of immobilization process. Moreover, the immobilized ß-galactosidase revealed a very good reusability as it could be reused for 15 and 20 consecutive cycles keeping 99.7 and 72.1% of its initial activity, respectively. In conclusion, the environmental waste (tea waste) can be used in modern technological industries such as the food and pharmaceutical industry.


Subject(s)
Alginates , Enzymes, Immobilized , Enzymes, Immobilized/chemistry , Microspheres , Hydrogen-Ion Concentration , Alginates/chemistry , Tea , Spectroscopy, Fourier Transform Infrared , Temperature , beta-Galactosidase/chemistry
18.
Int J Biol Macromol ; 254(Pt 3): 127901, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37952798

ABSTRACT

This work describes the synthesis of fibrous nickel-based metal organic framework (Ni-ZIF) via simple solvothermal method. The material formed was calcinated at 400, 600, 800 °C to improve its surface area, porosity and enzyme binding capacity. Changes in X-ray diffraction pattern after calcination revealed the Ni-ZIF transitioned from amorphous to crystalline structure. The surface area, pore volume and pore size for Ni-ZIF@600 were found to be 312.15 m2/g, 0.88 cm3/g and 10.28 nm, with an enzyme loading capacity of 593.85 mg/g after 30 h The free (ß-Gal-LEH) and immobilized ß-Galactosidase were stable at pH 7.5, temperature 50 °C, and yielded 70.70 and 63.95 mM glucose after milk lactose hydrolysis, respectively. The Ni-ZIF@600@ß-Gal-LEH exhibited high enzyme retention capacity, maintaining 59.44 % of its original activity after 6-cycles. The enhanced magnetic property, enzyme binding capacity and easy recoverability of the calcinated Ni-ZIF could guarantee its industrial significance as immobilization module for enzyme-mediated catalysis.


Subject(s)
Enzymes, Immobilized , Nickel , Nickel/chemistry , Enzymes, Immobilized/chemistry , Temperature , beta-Galactosidase/chemistry , Magnetic Phenomena
19.
Int J Biol Macromol ; 256(Pt 2): 128418, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38029902

ABSTRACT

The objective of this study was to immobilize a recombinant ß-galactosidase (Gal) tagged with a cellulose-binding domain (CBD) onto a magnetic core-shell (CS) cellulose system. After 30 min of reaction, 4 U/capsule were immobilized (CS@Gal), resulting in levels of yield and efficiency exceeding 80 %. The optimal temperature for ß-galactosidase-CBD activity increased from 40 to 50 °C following oriented immobilization. The inhibitory effect of galactose decreased in the enzyme reactions catalyzed by CS@Gal, and Mg2+ increased the immobilized enzyme activity by 40 % in the magnetic CS cellulose system. The relative enzyme activity of the CS@Gal was 20 % higher than that of the soluble enzyme activity after 20 min at 50 °C. The CS support and CS@Gal capsules exhibited an average size of 8 ± 1 mm, with the structure of the shell (alginate-pectin-cellulose) enveloping and isolating the magnetic core. The immobilized ß-galactosidase-CBD within the magnetic CS cellulose system retained ∼80 % of its capacity to hydrolyze lactose from skim milk after 10 reuse cycles. This study unveils a novel and promising support for the oriented immobilization of recombinant ß-galactosidase using a magnetic CS system and a CBD tag. This support facilitates ß-galactosidase reuse and efficient separation, consequently enhancing the catalytic properties of the enzyme.


Subject(s)
Cellulose , Enzymes, Immobilized , Cellulose/chemistry , Enzymes, Immobilized/chemistry , Catalysis , beta-Galactosidase/chemistry , Magnetic Phenomena
20.
Bioprocess Biosyst Eng ; 47(2): 263-273, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38156992

ABSTRACT

The objective of this study was to develop a bioprocess for lactose hydrolysis in diverse dairy matrices, specifically skim milk and cheese whey, utilizing column reactors employing a core-shell enzymatic system featuring ß-galactosidase fused to a Cellulose Binding Domain (CBD) tag (ß-galactosidase-CBD). The effectiveness of reactor configurations, including ball columns and toothed columns operating in packed and fluidized-bed modes, was evaluated for catalyzing lactose hydrolysis in both skim milk and cheese whey. In a closed system, these reactors achieved lactose hydrolysis rates of approximately 50% within 5 h under all evaluated conditions. Considering the scale of the bioprocess, the developed enzymatic system was capable of continuously hydrolyzing 9.6 L of skim milk while maintaining relative hydrolysis levels of approximately 50%. The biocatalyst, created by immobilizing ß-galactosidase-CBD on magnetic core-shell capsules, exhibited exceptional operational stability, and the proposed bioprocess employing these column reactors showcases the potential for scalability.


Subject(s)
Lactose , Milk , Animals , Lactose/chemistry , Hydrolysis , Milk/chemistry , Milk/metabolism , beta-Galactosidase/chemistry , Magnetic Phenomena , Enzymes, Immobilized/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL