Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.147
Filtrar
1.
J Agric Food Chem ; 72(34): 19081-19092, 2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39105795

RESUMEN

Chitosanases are valuable enzymatic tools in the food industry for converting chitosan into functional chitooligosaccharides (COSs). However, most of the chitosanases extensively characterized produced a low degree of polymerization (DP) COSs (DP = 1-3, LdpCOSs), indicating an imperative for enhancements in the product specificity for the high DP COS (DP >3, HdpCOSs) production. In this study, a chitosanase from Methanosarcina sp. 1.H.T.1A.1 (OUC-CsnA4) was cloned and expressed. Analysis of the enzyme-substrate interactions and the subsite architecture of the OUC-CsnA4 indicated that a Ser49 mutation could modify its interaction pattern with the substrate, potentially enhancing product specificity for producing HdpCOSs. Site-directed mutagenesis provided evidence that the S49I and S49P mutations in OUC-CsnA4 enabled the production of up to 24 and 26% of (GlcN)5 from chitosan, respectively─the wild-type enzyme was unable to produce detectable levels of (GlcN)5. These mutations also altered substrate binding preferences, favoring the binding of longer-chain COSs (DP >5) and enhancing (GlcN)5 production. Furthermore, molecular dynamics simulations and molecular docking studies underscored the significance of +2 subsite interactions in determining the (GlcN)4 and (GlcN)5 product specificity. These findings revealed that the positioning and interactions of the reducing end of the substrate within the catalytic cleft are crucial factors influencing the product specificity of chitosanase.


Asunto(s)
Quitosano , Glicósido Hidrolasas , Methanosarcina , Mutagénesis Sitio-Dirigida , Oligosacáridos , Polimerizacion , Oligosacáridos/química , Oligosacáridos/metabolismo , Quitosano/química , Quitosano/metabolismo , Glicósido Hidrolasas/genética , Glicósido Hidrolasas/metabolismo , Glicósido Hidrolasas/química , Especificidad por Sustrato , Methanosarcina/enzimología , Methanosarcina/genética , Methanosarcina/metabolismo , Methanosarcina/química , Proteínas Arqueales/genética , Proteínas Arqueales/metabolismo , Proteínas Arqueales/química , Quitina/metabolismo , Quitina/química , Quitina/análogos & derivados , Cinética
2.
Int J Mol Sci ; 25(15)2024 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-39126103

RESUMEN

The formation and analysis of amyloid fibers by two ß-glucosidases, BglA and BglB, belonging to the GH1 enzyme family, are reported. Both proteins have the (ß/α)8 TIM-barrel fold, which is characteristic of this family and is also the most common protein structure. BglA is an octamer, whereas BglB is a monomer. Amyloid fibrillation using pH and temperature as perturbing agents was investigated using fluorescence spectroscopy as a preliminary approach and corroborated using wide-field optical microscopy, confocal microscopy, and field-emission scanning electron microscopy. These analyses showed that both enzymes fibrillate at a wide range of acidic and alkaline conditions and at several temperature conditions, particularly at acidic pH (3-4) and at temperatures between 45 and 65 °C. Circular dichroism spectroscopy corroborated the transition from an α-helix to a ß-sheet secondary structure of both proteins in conditions where fibrillation was observed. Overall, our results suggest that fibrillation is a rather common phenomenon caused by protein misfolding, driven by a transition from an α-helix to a ß-sheet secondary structure, that many proteins can undergo if subjected to conditions that disturb their native conformation.


Asunto(s)
Amiloide , Amiloide/química , Amiloide/metabolismo , Concentración de Iones de Hidrógeno , Glicósido Hidrolasas/química , Glicósido Hidrolasas/metabolismo , Dicroismo Circular , Temperatura , Estructura Secundaria de Proteína , Pliegue de Proteína
3.
J Agric Food Chem ; 72(30): 16911-16929, 2024 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-39025827

RESUMEN

The glycoside hydrolase family 70 (GH70) contains bacterial extracellular multidomain enzymes, synthesizing α-glucans from sucrose or starch-like substrates. A few dozen have been biochemically characterized, while crystal structures cover only the core domains and lack significant parts of auxiliary domains. Here we present a systematic overview of GH70 enzymes and their 3D structural organization and bacterial origin. A representative set of 234 permuted and 25 nonpermuted GH70 enzymes was generated, covering 12 bacterial families and 3 phyla and containing 185 predicted glucansucrases (GS), 15 branching sucrases (BrS), 8 "twin" GS-BrSs, and 51 α-glucanotransferases (α-GT). Analysis of AlphaFold models of all 259 entries showed that, apart from the core domains, the structural variation regarding auxiliary domains is far greater than anticipated, with nine different domain types. We analyzed the phylogenetic distribution and discuss the possible roles of auxiliary domains as well as possible correlations between enzyme specificity, auxiliary domain type, and bacterial origin.


Asunto(s)
Bacterias , Proteínas Bacterianas , Glicósido Hidrolasas , Filogenia , Glicósido Hidrolasas/química , Glicósido Hidrolasas/genética , Glicósido Hidrolasas/metabolismo , Bacterias/enzimología , Bacterias/genética , Bacterias/clasificación , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Secuencia de Aminoácidos , Especificidad por Sustrato
4.
J Agric Food Chem ; 72(31): 17510-17523, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-39052486

RESUMEN

To convert ginsenosides Rb1, Rb2, Rb3, and Rc into Rd by a single enzyme, a putative ß-glycosidase (Pxbgl) from the xylan-degrading bacterium Petroclostridium xylanilyticum was identified and used. The kcat/Km value of Pxbgl for Rb3 was 18.18 ± 0.07 mM-1/s, which was significantly higher than those of Pxbgl for other ginsenosides. Pxbgl converted almost all Rb3 to Rd with a productivity of 5884 µM/h, which was 346-fold higher than that of only ß-xylosidase from Thermoascus aurantiacus. The productivity of Rd from the Panax ginseng root and Panax notoginseng leaf was 146 and 995 µM/h, respectively. Mutants N293 K and I447L from site-directed mutagenesis based on bioinformatics analysis showed an increase in specific activity of 29 and 7% toward Rb3, respectively. This is the first report of a ß-glycosidase that can simultaneously remove four different glycosyls at the C-20 position of natural PPD-type ginsenosides and produce Rd as the sole product from P. notoginseng leaf extracts with the highest productivity.


Asunto(s)
Proteínas Bacterianas , Ginsenósidos , Panax , Ginsenósidos/metabolismo , Ginsenósidos/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/química , Panax/química , Panax/genética , Panax/metabolismo , Especificidad por Sustrato , Glicósido Hidrolasas/genética , Glicósido Hidrolasas/metabolismo , Glicósido Hidrolasas/química , Cinética , beta-Glucosidasa/metabolismo , beta-Glucosidasa/genética , beta-Glucosidasa/química , Raíces de Plantas/química , Raíces de Plantas/metabolismo , Panax notoginseng/química , Panax notoginseng/genética , Panax notoginseng/enzimología , Panax notoginseng/metabolismo
5.
Int J Biol Macromol ; 276(Pt 1): 133642, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38964696

RESUMEN

Pullulanases are important starch-debranching enzymes that mainly hydrolyze the α-1,6-glycosidic linkages in pullulan, starch, and oligosaccharides. Nevertheless, their practical applications are constrained because of their poor activity and low thermostability. Moreover, the trade-off between activity and thermostability makes it challenging to simultaneously improve them. In this study, an engineered pullulanase was developed through reshaping the active-site tunnel and engineering the surface lysine residues using the pullulanase from Pyrococcus yayanosii CH1 (PulPY2). The specific activity of the engineered pullulanase was increased 3.1-fold, and thermostability was enhanced 1.8-fold. Moreover, the engineered pullulanase exhibited 11.4-fold improvement in catalytic efficiency (kcat/Km). Molecular dynamics simulations demonstrated an anti-correlated movement around the entrance of active-site tunnel and stronger interactions between the surface residues in the engineered pullulanase, which would be beneficial to the activity and thermostability improvement, respectively. The strategies used in this study and dynamic evidence for insight into enzyme performance improvement may provide guidance for the activity and thermostability engineering of other enzymes.


Asunto(s)
Dominio Catalítico , Estabilidad de Enzimas , Glicósido Hidrolasas , Lisina , Simulación de Dinámica Molecular , Glicósido Hidrolasas/química , Glicósido Hidrolasas/metabolismo , Lisina/química , Lisina/metabolismo , Pyrococcus/enzimología , Ingeniería de Proteínas/métodos , Cinética , Temperatura
6.
Int J Biol Macromol ; 277(Pt 1): 133960, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39029832

RESUMEN

Agarose from biomass can be used to synthesize the rare sugar 3,6-anhydro-L-galactose (L-AHG), and the new synthesis route and functional properties of L-AHG have always been the focus of research. Here we developed a novel method to co-immobilize Aga50D and BpGH117 onto streptavidin-coated magnetic nanoparticles and achieved the conversion of agarose to bioactive L-AHG in one pot. Results showed that enzymes were successfully immobilized on the carrier. The activity of co-immobilized enzymes was 2.5-fold higher than that of single immobilized enzymes. Compared with free enzymes, co-immobilized enzymes exhibited enhanced thermal stability. The co-immobilized enzymes retained 79.45 % relative activity at 40 °C for 3 h, while the free enzymes only possessed 21.40 % residual activity. After eight cycles, the co-immobilized enzymes still retained 73.47 % of the initial activity. After silica gel chromatography, the purity of L-AHG obtained by co-immobilized enzymes hydrolysis reached 83.02 %. Furthermore, bioactivity experiments demonstrated that L-AHG displayed better antioxidant and antibacterial effects than neoagarobiose. L-AHG had broad-spectrum antibacterial activity, while neoagarobiose and D-galactose did not show an obvious antibacterial effect. This study provides a feasible method for the production of L-AHG by a co-immobilized multi-enzyme system and confirms that L-AHG plays a key role in the bioactivity of neoagarobiose.


Asunto(s)
Enzimas Inmovilizadas , Galactosa , Glicósido Hidrolasas , Sefarosa , Sefarosa/química , Sefarosa/análogos & derivados , Enzimas Inmovilizadas/química , Enzimas Inmovilizadas/metabolismo , Galactosa/análogos & derivados , Galactosa/química , Glicósido Hidrolasas/metabolismo , Glicósido Hidrolasas/química , Antibacterianos/farmacología , Antibacterianos/química , Hidrólisis , Estabilidad de Enzimas , Antioxidantes/farmacología , Antioxidantes/química , Temperatura
7.
Int J Biol Macromol ; 277(Pt 1): 134093, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39053825

RESUMEN

Seaweed polysaccharides have a wide range of sources and rich content, with various biological activities such as anti-inflammatory, anti-tumor, anticoagulant, and blood pressure lowering. They can be applied in fields such as food, agriculture, and medicine. However, the poor solubility of macromolecular seaweed polysaccharides limits their further application. Reports have shown that some biological activities of seaweed oligosaccharides are more extensive and superior to that of seaweed polysaccharides. Therefore, reducing the degree of polymerization of polysaccharides will be the key to the high value utilization of seaweed polysaccharide resources. There are three main methods for degrading algal polysaccharides into algal oligosaccharides, physical, chemical and enzymatic degradation. Among them, enzymatic degradation has been a hot research topic in recent years. Various types of algal polysaccharide hydrolases and related glycosidases are powerful tools for the preparation of algal oligosaccharides, including α-agarases, ß-agaroses, α-neoagarose hydrolases and ß-galactosidases that are related to agar, κ-carrageenases, ι-carrageenases and λ-carrageenases that are related to carrageenan, ß-porphyranases that are related to porphyran, funoran hydrolases that are related to funoran, alginate lyases that are related to alginate and ulvan lyases related to ulvan. This paper describes the bioactivities of agar oligosaccharide, carrageenan oligosaccharide, porphyran oligosaccharide, funoran oligosaccharide, alginate oligosaccharide and ulvan oligosaccharide and provides a detailed review of the progress of research on the enzymatic preparation of these six oligosaccharides. At the same time, the problems and challenges faced are presented to guide and improve the preparation and application of algal oligosaccharides in the future.


Asunto(s)
Glicósido Hidrolasas , Oligosacáridos , Polisacáridos , Algas Marinas , Oligosacáridos/química , Oligosacáridos/farmacología , Algas Marinas/química , Polisacáridos/química , Polisacáridos/farmacología , Glicósido Hidrolasas/metabolismo , Glicósido Hidrolasas/química , Hidrólisis , Carragenina/química , Alginatos/química , Polisacárido Liasas/metabolismo , Polisacárido Liasas/química
8.
Int J Biol Macromol ; 277(Pt 2): 134174, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39084418

RESUMEN

The GH78 α-L-rhamnosidase from Aspergillus tubingensis (AT-Rha) was proved to be a new clade of Aspergillus α-L-rhamnosidases in the previous study. A putative α-L-rhamnosidase from A. kawachii IFO 4308 (AK-Rha) has 92 % identity in amino acid sequence with AT-Rha. In this study, AK-Rha was expressed in P. pastoris and characterized. Similar to AT-rRha, the recombinant AK-Rha (AK-rRha) showed a narrow substrate specificity to naringin. Interestingly, the enzyme activity of AK-rRha was 0.816 U/mg toward naringin, significantly lower than 125.142 U/mg of AT-rRha. Their large differences in catalytic efficiency was mainly due to their differences in kcat values between AK-rRha (0.67 s-1) and AT-rRha (4.89 × 104 s-1). The molecular dynamics simulation exhibited that the overall conformation of AK-Rha was rigid and that of AT-Rha was flexible; the Loop Y-L located above the catalytic domain formed different steric hindrances to naringin, and interacted with the flavonoid matrices at different strengths. The polar solvation energy analysis implied that the glycosidic bond was more easily hydrolysed in AT-Rha. The comparative study verified that the main feature of AK-Rha and AT-Rha represented Aspergillus α-L-rhamnosidase was the narrow substrate specificity toward naringin, and provided an insight of the relationships between their catalytic abilities and structures.


Asunto(s)
Aspergillus , Glicósido Hidrolasas , Glicósido Hidrolasas/química , Glicósido Hidrolasas/genética , Glicósido Hidrolasas/metabolismo , Especificidad por Sustrato , Aspergillus/enzimología , Aspergillus/genética , Secuencia de Aminoácidos , Simulación de Dinámica Molecular , Flavanonas/química , Cinética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido
9.
Nat Commun ; 15(1): 5510, 2024 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-38951487

RESUMEN

Co-immobilization of cells and enzymes is often essential for the cascade biocatalytic processes of industrial-scale feasibility but remains a vast challenge. Herein, we create a facile co-immobilization platform integrating enzymes and cells in covalent organic frameworks (COFs) to realize the highly efficient cascade of inulinase and E. coli for bioconversion of natural products. Enzymes can be uniformly immobilized in the COF armor, which coats on the cell surface to produce cascade biocatalysts with high efficiency, stability and recyclability. Furthermore, this one-pot in situ synthesis process facilitates a gram-scale fabrication of enzyme-cell biocatalysts, which can generate a continuous-flow device conversing inulin to D-allulose, achieving space-time yield of 161.28 g L-1 d-1 and high stability (remaining >90% initial catalytic efficiency after 7 days of continuous reaction). The created platform is applied for various cells (e.g., E. coli, Yeast) and enzymes, demonstrating excellent universality. This study paves a pathway to break the bottleneck of extra- and intracellular catalysis, creates a high-performance and customizable platform for enzyme-cell cascade biomanufacturing, and expands the scope of biocatalysis process intensification.


Asunto(s)
Biocatálisis , Células Inmovilizadas , Enzimas Inmovilizadas , Escherichia coli , Glicósido Hidrolasas , Escherichia coli/metabolismo , Enzimas Inmovilizadas/metabolismo , Enzimas Inmovilizadas/química , Glicósido Hidrolasas/metabolismo , Glicósido Hidrolasas/química , Células Inmovilizadas/metabolismo , Estructuras Metalorgánicas/química , Estructuras Metalorgánicas/metabolismo , Saccharomyces cerevisiae/metabolismo
10.
PLoS One ; 19(7): e0306410, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38990885

RESUMEN

Carbohydrate-active enzymes (CAZymes) can be found in all domains of life and play a crucial role in metabolic and physiological processes. CAZymes often possess a modular structure, comprising not only catalytic domains but also associated domains such as carbohydrate-binding modules (CBMs) and linker domains. By exploring the modular diversity of CAZy families, catalysts with novel properties can be discovered and further insight in their biological functions and evolutionary relationships can be obtained. Here we present the carbohydrate-active enzyme domain analysis tool (CANDy), an assembly of several novel scripts, tools and databases that allows users to analyze the domain architecture of all protein sequences in a given CAZy family. CANDy's usability is shown on glycoside hydrolase family 48, a small yet underexplored family containing multi-domain enzymes. Our analysis reveals the existence of 35 distinct domain assemblies, including eight known architectures, with the remaining assemblies awaiting characterization. Moreover, we substantiate the occurrence of horizontal gene transfer from prokaryotes to insect orthologs and provide evidence for the subsequent removal of auxiliary domains, likely through a gene fission event. CANDy is available at https://github.com/PyEED/CANDy.


Asunto(s)
Dominios Proteicos , Glicósido Hidrolasas/química , Glicósido Hidrolasas/metabolismo , Glicósido Hidrolasas/genética , Dominio Catalítico , Programas Informáticos , Metabolismo de los Hidratos de Carbono , Carbohidratos/química , Animales
11.
Gene ; 927: 148758, 2024 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-38977109

RESUMEN

The gut microbiota is a treasure trove of carbohydrate-active enzymes (CAZymes). To explore novel and efficient CAZymes, we analyzed the 4,142 metagenome-assembled genomes (MAGs) of the horse gut microbiota and found the MAG117.bin13 genome (Bacteroides fragilis) contains the highest number of polysaccharide utilisation loci sites (PULs), indicating its high capability for carbohydrate degradation. Bioinformatics analysis indicate that the PULs region of the MAG117.bin13 genome encodes many hypothetical proteins, which are important sources for exploring novel CAZymes. Interestingly, we discovered a hypothetical protein (595 amino acids). This protein exhibits potential CAZymes activity and has a lower similarity to CAZymes, we named it BfLac2275. We purified the protein using prokaryotic expression technology and studied its enzymatic function. The hydrolysis experiment of the polysaccharide substrate showed that the BfLac2275 protein has the ability to degrade α-lactose (156.94 U/mg), maltose (92.59 U/mg), raffinose (86.81 U/mg), and hyaluronic acid (5.71 U/mg). The enzyme activity is optimal at pH 5.0 and 30 ℃, indicating that the hypothetical protein BfLac2275 is a novel and multifunctional CAZymes in the glycoside hydrolases (GHs). These properties indicate that BfLac2275 has broad application prospects in many fields such as plant polysaccharide decomposition, food industry, animal feed additives and enzyme preparations. This study not only serves as a reference for exploring novel CAZymes encoded by gut microbiota but also provides an example for further studying the functional annotation of hypothetical genes in metagenomic assembly genomes.


Asunto(s)
Microbioma Gastrointestinal , Glicósido Hidrolasas , Metagenoma , Animales , Glicósido Hidrolasas/genética , Glicósido Hidrolasas/metabolismo , Glicósido Hidrolasas/química , Microbioma Gastrointestinal/genética , Caballos , Genoma Bacteriano , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Especificidad por Sustrato , Filogenia
12.
Glycobiology ; 34(8)2024 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-38982733

RESUMEN

Understanding the relation between enzyme domain structure and catalytic activity is crucial for optimal engineering of novel enzymes for lignocellulose bioconversion. Xylanases with varying specificities are commonly used to valorise the hemicellulose arabinoxylan (AX), yet characterization of specific arabinoxylanases remain limited. Two homologous GH5_34 arabinoxylanases, HhXyn5A and CtXyn5A, in which the two domains are connected by a 40-residue linker, exhibit distinct activity on AX, yielding different reaction product patterns, despite high sequence identity, conserved active sites and similar domain composition. In this study, the carbohydrate binding module 6 (CBM6), or the inter domain linker together with CBM6, were swapped to investigate their influence on hydrolytic activity and oligosaccharide product pattern on cereal AXs. The variants, with only CBM6 swapped, displayed reduced activity on commercial wheat and rye AX, as well as on extracted oat fibre, compared to the original enzymes. Additionally, exchange of both linker and CBM6 resulted in a reduced ratio of enzyme produced in soluble form in Escherichia coli cultivations, causing loss of activity of both HhXyn5A and CtXyn5A variants. Analysis of oligosaccharide product patterns applying HPAEC-PAD revealed a decreased number of reaction products for CtXyn5A with swapped CBM6, which resembled the product pattern of HhXyn5A. These findings emphasize the importance of the CBM6 interactions with the linker and the catalytic domain for enzyme activity and specificity, and underlines the role of the linker in enzyme structure organisation and product formation, where alterations in linker interactions with the catalytic and/or CBM6 domains, influence enzyme-substrate association and specificity.


Asunto(s)
Oligosacáridos , Xilanos , Oligosacáridos/química , Oligosacáridos/metabolismo , Xilanos/metabolismo , Xilanos/química , Glicósido Hidrolasas/química , Glicósido Hidrolasas/metabolismo , Glicósido Hidrolasas/genética , Dominio Catalítico , Dominios Proteicos , Especificidad por Sustrato , Hidrólisis , Endo-1,4-beta Xilanasas/química , Endo-1,4-beta Xilanasas/metabolismo , Endo-1,4-beta Xilanasas/genética
13.
Int J Biol Macromol ; 275(Pt 1): 133449, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38944065

RESUMEN

Glycoside hydrolases (GHs) are pivotal in the hydrolysis of the glycosidic bonds of sugars, which are the main carbon and energy sources. The genome of Marinomonas sp. ef1, an Antarctic bacterium, contains three GHs belonging to family 3. These enzymes have distinct architectures and low sequence identity, suggesting that they originated from separate horizontal gene transfer events. M-GH3_A and M-GH3_B, were found to differ in cold adaptation and substrate specificity. M-GH3_A is a bona fide cold-active enzyme since it retains 20 % activity at 10 °C and exhibits poor long-term thermal stability. On the other hand, M-GH3_B shows mesophilic traits with very low activity at 10 °C (< 5 %) and higher long-term thermal stability. Substrate specificity assays highlight that M-GH3_A is a promiscuous ß-glucosidase mainly active on cellobiose and cellotetraose, whereas M-GH3_B is a ß-xylosidase active on xylan and arabinoxylan. Structural analysis suggests that such functional differences are due to their differently shaped active sites. The active site of M-GH3_A is wider but has a narrower entrance compared to that of M-GH3_B. Genome-based prediction of metabolic pathways suggests that Marinomonas sp. ef1 can use monosaccharides derived from the GH3-catalyzed hydrolysis of oligosaccharides either as a carbon source or for producing osmolytes.


Asunto(s)
Evolución Molecular , Glicósido Hidrolasas , Oligosacáridos , Glicósido Hidrolasas/metabolismo , Glicósido Hidrolasas/genética , Glicósido Hidrolasas/química , Especificidad por Sustrato , Oligosacáridos/metabolismo , Regiones Antárticas , Polisacáridos/metabolismo , Polisacáridos/química , Filogenia , Marinomonas/enzimología , Marinomonas/genética , Organismos Acuáticos/enzimología , Estabilidad de Enzimas , Dominio Catalítico , Hidrólisis
14.
J Biosci Bioeng ; 138(2): 118-126, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38825558

RESUMEN

The α-1,3-glucanase Agl-EK14 from Flavobacterium sp. EK-14 comprises a signal peptide (SP), a catalytic domain (CAT), a first immunoglobulin-like domain (Ig1), a second immunoglobulin-like domain (Ig2), a ricin B-like lectin domain (RicinB), and a carboxy-terminal domain (CTD). SP and CTD are predicted to be involved in extracellular secretion, while the roles of Ig1, Ig2, and RicinB are unclear. To clarify their roles, domain deletion enzymes Agl-EK14ΔRicinB, Agl-EK14ΔIg2RicinB, and Agl-EK14ΔIg1Ig2RicinB were constructed. The insoluble α-1,3-glucan hydrolytic, α-1,3-glucan binding, and fungal cell wall hydrolytic activities of the deletion enzymes were almost the same and lower than those of Agl-EK14. Kinetic analysis revealed that the Km values of the deletion enzymes were similar and uniformly higher than those of Agl-EK14. These results suggest that the deletion of RicinB causes a decline in binding and hydrolytic activity and increases the Km value. To confirm the role of RicinB, Ig1, Ig2, and RicinB were fused with green fluorescent protein (GFP). As a result, RicinB-fused GFP (GFP-RicinB) showed binding to insoluble α-1,3-glucan and Aspergillus oryzae cell walls, whereas Ig1- and Ig2-fused GFP did not. These results indicated that RicinB is involved in α-1,3-glucan binding. The fusion protein GFP-Ig1Ig2RicinB was also constructed and GFP-Ig1Ig2RicinB showed strong binding to the cell wall of A. oryzae compared to GFP-RicinB. Gel filtration column chromatography suggested that the strong binding was due to GFP-Ig1Ig2RicinB loosely associated with itself.


Asunto(s)
Pared Celular , Flavobacterium , Glucanos , Dominios Proteicos , Flavobacterium/enzimología , Flavobacterium/genética , Flavobacterium/metabolismo , Pared Celular/metabolismo , Glucanos/metabolismo , Hidrólisis , Dominio Catalítico , Cinética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/química , Glicósido Hidrolasas/metabolismo , Glicósido Hidrolasas/genética , Glicósido Hidrolasas/química , Señales de Clasificación de Proteína
15.
Org Biomol Chem ; 22(28): 5783-5789, 2024 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-38938184

RESUMEN

Aspergillus fumigatus is a saprophytic fungus and opportunistic pathogen often causing fatal infections in immunocompromised patients. Recently AfKDNAse, an exoglycosidase hydrolyzing 3-deoxy-D-galacto-D-glycero-nonulosonic acid (KDN), a rare sugar from the sialic acid family, was identified and characterized. The principal function of AfKDNAse is still unclear, but a study suggests a critical role in fungal cell wall morphology and virulence. Potent AfKDNAse inhibitors are required to better probe the enzyme's biological role and as potential antivirulence factors. In this work, we developed a set of AfKDNAse inhibitors based on enzymatically stable thio-KDN motifs. C2, C9-linked heterodi-KDN were designed to fit into unusually close KDN sugar binding pockets in the protein. A polymeric compound with an average of 54 KDN motifs was also designed by click chemistry. Inhibitory assays performed on recombinant AfKDNAse showed a moderate and strong enzymatic inhibition for the two classes of compounds, respectively. The poly-KDN showed more than a nine hundred fold improved inhibitory activity (IC50 = 1.52 ± 0.37 µM, 17-fold in a KDN molar basis) compared to a monovalent KDN reference, and is to our knowledge, the best synthetic inhibitor described for a KDNase. Multivalency appears to be a relevant strategy for the design of potent KDNase inhibitors. Importantly, poly-KDN was shown to strongly decrease filamentation when co-cultured with A. fumigatus at micromolar concentrations, opening interesting perspectives in the development of antivirulence factors.


Asunto(s)
Aspergillus fumigatus , Glicósido Hidrolasas , Aspergillus fumigatus/enzimología , Aspergillus fumigatus/efectos de los fármacos , Glicósido Hidrolasas/antagonistas & inhibidores , Glicósido Hidrolasas/metabolismo , Glicósido Hidrolasas/química , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/síntesis química , Relación Estructura-Actividad , Modelos Moleculares , Estructura Molecular
16.
Arch Microbiol ; 206(7): 307, 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38884653

RESUMEN

Xylanase is the most important hydrolase in the xylan hydrolase system, the main function of which is ß-1,4-endo-xylanase, which randomly cleaves xylans to xylo-oligosaccharides and xylose. Xylanase has wide ranging of applications, but there remains little research on the cold-adapted enzymes required in some low-temperature industries. Glycoside hydrolase family 8 (GH8) xylanases have been reported to have cold-adapted enzyme activity. In this study, the xylanase gene dgeoxyn was excavated from Deinococcus geothermalis through sequence alignment. The recombinant xylanase DgeoXyn encodes 403 amino acids with a theoretical molecular weight of 45.39 kDa. Structural analysis showed that DgeoXyn has a (α/α)6-barrel fold structure typical of GH8 xylanase. At the same time, it has strict substrate specificity, is only active against xylan, and its hydrolysis products include xylobiose, xylotrinose, xytetranose, xylenanose, and a small amount of xylose. DgeoXyn is most active at 70 â„ƒ and pH 6.0. It is very stable at 10, 20, and 30 â„ƒ, retaining more than 80% of its maximum enzyme activity. The enzyme activity of DgeoXyn increased by 10% after the addition of Mn2+ and decreased by 80% after the addition of Cu2+. The Km and Vmax of dgeox were 42 mg/ml and 20,000 U/mg, respectively, at a temperature of 70 â„ƒ and pH of 6.0 using 10 mg/ml beechwood xylan as the substrate. This research on DgeoXyn will provide a theoretical basis for the development and application of low-temperature xylanase.


Asunto(s)
Deinococcus , Endo-1,4-beta Xilanasas , Estabilidad de Enzimas , Xilanos , Deinococcus/enzimología , Deinococcus/genética , Especificidad por Sustrato , Endo-1,4-beta Xilanasas/genética , Endo-1,4-beta Xilanasas/química , Endo-1,4-beta Xilanasas/metabolismo , Xilanos/metabolismo , Frío , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/química , Concentración de Iones de Hidrógeno , Glicósido Hidrolasas/genética , Glicósido Hidrolasas/metabolismo , Glicósido Hidrolasas/química , Secuencia de Aminoácidos , Hidrólisis , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Alineación de Secuencia , Clonación Molecular , Cinética , Peso Molecular , Disacáridos
17.
Food Chem ; 457: 140163, 2024 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-38924912

RESUMEN

Enzymatically modified isoquercitrin (EMIQ) is a glyco-chemically modified flavonoid exhibiting notably high biological activity, such as antioxidant, anti-inflammatory and anti-allergic properties. However, the utilization of expensive substrates such as isoquercitrin and cyclodextrin in the conventional approach has hindered the industrial-scale production of EMIQ due to high cost and low yields. Hence, the development of a cost-effective and efficient method is crucial for the biological synthesis of EMIQ. In this study, a natural cascade catalytic reaction system was constructed with α-L-rhamnosidase and amylosucrase using the inexpensive substrates rutin and sucrose. Additionally, a novel approach integrating gradient temperature regulation into biological cascade reactions was implemented. Under the optimal conditions, the rutin conversion reached a remarkable 95.39% at 24 h. Meanwhile, the productivity of quercetin-3-O-tetraglucoside with the best bioavailability reached an impressive 41.69%. This study presents promising prospects for future mass production of EMIQ directly prepared from rutin and sucrose.


Asunto(s)
Glucosiltransferasas , Quercetina , Rutina , Sacarosa , Rutina/química , Quercetina/química , Quercetina/análogos & derivados , Quercetina/metabolismo , Sacarosa/química , Sacarosa/análogos & derivados , Sacarosa/metabolismo , Glucosiltransferasas/metabolismo , Glucosiltransferasas/química , Glicósido Hidrolasas/química , Glicósido Hidrolasas/metabolismo , Temperatura , Biocatálisis
18.
Nature ; 631(8019): 199-206, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38898276

RESUMEN

The vast majority of glycosidases characterized to date follow one of the variations of the 'Koshland' mechanisms1 to hydrolyse glycosidic bonds through substitution reactions. Here we describe a large-scale screen of a human gut microbiome metagenomic library using an assay that selectively identifies non-Koshland glycosidase activities2. Using this, we identify a cluster of enzymes with extremely broad substrate specificities and thoroughly characterize these, mechanistically and structurally. These enzymes not only break glycosidic linkages of both α and ß stereochemistry and multiple connectivities, but also cleave substrates that are not hydrolysed by standard glycosidases. These include thioglycosides, such as the glucosinolates from plants, and pseudoglycosidic bonds of pharmaceuticals such as acarbose. This is achieved through a distinct mechanism of hydrolysis that involves oxidation/reduction and elimination/hydration steps, each catalysed by enzyme modules that are in many cases interchangeable between organisms and substrate classes. Homologues of these enzymes occur in both Gram-positive and Gram-negative bacteria associated with the gut microbiome and other body parts, as well as other environments, such as soil and sea. Such alternative step-wise mechanisms appear to constitute largely unrecognized but abundant pathways for glycan degradation as part of the metabolism of carbohydrates in bacteria.


Asunto(s)
Bacterias , Microbioma Gastrointestinal , Glicósido Hidrolasas , Polisacáridos , Humanos , Acarbosa/química , Acarbosa/metabolismo , Bacterias/enzimología , Bacterias/genética , Bacterias/aislamiento & purificación , Bacterias/metabolismo , Biocatálisis , Glucosinolatos/metabolismo , Glucosinolatos/química , Glicósido Hidrolasas/metabolismo , Glicósido Hidrolasas/química , Hidrólisis , Metagenoma , Oxidación-Reducción , Plantas/química , Polisacáridos/metabolismo , Polisacáridos/química , Agua de Mar/microbiología , Microbiología del Suelo , Especificidad por Sustrato , Masculino
19.
Int J Biol Macromol ; 274(Pt 1): 133262, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38901511

RESUMEN

The physicochemical properties of starch and phenolic acid (PA) complexes largely depend on the effect of non-covalent interactions on the microstructure of starch. However, whether there are differences and commonalities in the interactions between various types of PAs and starch remains unclear. The physicochemical properties and digestive characteristics of the complexes were investigated by pre-gelatinization of 16 structurally different PAs and pullulanase-modified rice starches screened. FT-IR and XRD results revealed that PA complexed with debranched rice starch (DRS) through hydrogen bonding and hydrophobic interaction. Benzoic/phenylacetic acid with polyhydroxy groups could enter the helical cavities of the starch chains to promote the formation of V-shaped crystals, and cinnamic acid with p-hydroxyl structure acted between starch chains in a bridging manner, both of which increased the relative crystallinity of DRS, with DRS-ellagic acid increasing to 20.03 %. The digestion and hydrolysis results indicated that the acidification and methoxylation of PA synergistically decreased the enzyme activity leading to a decrease in the digestibility of the complexes, and the resistant starch content of the DRS-vanillic acid complexes increased from 28.27 % to 71.67 %. Therefore, the selection of structurally appropriate PAs can be used for the targeted preparation of starch-based foods and materials.


Asunto(s)
Oryza , Almidón , Oryza/química , Almidón/química , Hidrólisis , Digestión , Fenoles/química , Fenómenos Químicos , Glicósido Hidrolasas/química , Glicósido Hidrolasas/metabolismo , Hidroxibenzoatos/química , Espectroscopía Infrarroja por Transformada de Fourier
20.
Int J Biol Macromol ; 273(Pt 1): 133026, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38852722

RESUMEN

A novel glycoside hydrolase (GH) family 16 multi-domain ß-1,3-1,4-glucanase (FsGlc16A) from Fibrobacter sp. UWP2 was identified, heterogeneously expressed, and its enzymatic properties, protein structure and application potential were characterized. Enzymological characterization showed that FsGlc16A performed the optimal catalytic activity at pH 4.5 and 50 °C with a specific activity of 3263 U/mg. FsGlc16A exhibited the substrate specificity towards oat ß-glucan, barley ß-glucan and lichenan, and in addition, it hydrolyzed oat ß-glucan and lichenan into different ß-glucooligosaccharides with polymerization degrees of 3-4, which further illustrated that it belonged to the endo-type ß-1,3-1,4-glucanase. FsGlc16A was classified in subfamily25 of GH16. A 'PXSSSS' repeats domain was identified at the C-terminus of FsGlc16A, which was distinct from the typical GH family 16 ß-1,3-1,4-glucanases. Removing the 'PXSSSS' repeats domain affected the binding of the substrate to FsGlc16A and reduced the enzyme activity. FsGlc16A displayed good potential for the applications, which hydrolyzed oat bran into ß-glucooligosaccharides, and reduced filtration time (18.89 %) and viscosity (3.64 %) in the saccharification process. This study investigated the enzymatic properties and domain function of FsGlc16A, providing new ideas and insights into the study of ß-1,3-1,4-glucanase.


Asunto(s)
Glucanos , Especificidad por Sustrato , Hidrólisis , Glucanos/química , Glucanos/metabolismo , Concentración de Iones de Hidrógeno , Secuencia de Aminoácidos , Temperatura , Dominios Proteicos , beta-Glucanos/metabolismo , beta-Glucanos/química , Glicósido Hidrolasas/química , Glicósido Hidrolasas/metabolismo , Cinética , Endo-1,3(4)-beta-Glucanasa/química , Endo-1,3(4)-beta-Glucanasa/metabolismo , Endo-1,3(4)-beta-Glucanasa/genética , Clonación Molecular , Filogenia , Estabilidad de Enzimas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...