Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 372
Filtrar
1.
J Agric Food Chem ; 72(33): 18585-18593, 2024 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-39133835

RESUMEN

d-Tagatose is a highly promising functional sweetener known for its various physiological functions. In this study, a novel tagatose 4-epimerase from Thermoprotei archaeon (Thar-T4Ease), with the ability to convert d-fructose to d-tagatose, was discovered through a combination of structure similarity search and sequence-based protein clustering. The recombinant Thar-T4Ease exhibited optimal activity at pH 8.5 and 85 °C, in the presence of 1 mM Ni2+. Its kcat and kcat/Km values toward d-fructose were measured to be 248.5 min-1 and 2.117 mM-1·min-1, respectively. Notably, Thar-T4Ease exhibited remarkable thermostability, with a t1/2 value of 198 h at 80 °C. Moreover, it achieved a conversion ratio of 18.9% using 100 g/L d-fructose as the substrate. Finally, based on sequence and structure analysis, crucial residues for the catalytic activity of Thar-T4Ease were identified by molecular docking and site-directed mutagenesis. This research expands the repertoire of enzymes with C4-epimerization activity and opens up new possibilities for the cost-effective production of d-tagatose from d-fructose.


Asunto(s)
Estabilidad de Enzimas , Hexosas , Simulación del Acoplamiento Molecular , Hexosas/química , Hexosas/metabolismo , Cinética , Proteínas Arqueales/genética , Proteínas Arqueales/química , Proteínas Arqueales/metabolismo , Fructosa/química , Fructosa/metabolismo , Carbohidrato Epimerasas/genética , Carbohidrato Epimerasas/química , Carbohidrato Epimerasas/metabolismo , Concentración de Iones de Hidrógeno , Especificidad por Sustrato , Calor , Secuencia de Aminoácidos , Racemasas y Epimerasas/genética , Racemasas y Epimerasas/química , Racemasas y Epimerasas/metabolismo
2.
Int J Biol Macromol ; 277(Pt 3): 134202, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39089546

RESUMEN

Cellobiose 2-epimerase (CE) catalyzes the conversion of the lactose into its high-value derivatives, epilactose and lactulose, which has great prospects in food applications. In this study, CE sequences from the Qinghai-Tibet Plateau gene catalogue, we screened these for structural flexibility through molecular dynamics simulation to identify potential psychrophilic CE candidates. One such psychrophilic CE we termed psyCE demonstrated exceptional epimerization activity, achieving an optimum activity of 122.2 ± 1.6 U/mg. Its kinetic parameters (Kcat and Km) for epimerization activity were 219.9 ± 5.6 s-1 and 261.9 ± 18.1 mM, respectively, representing the highest Kcat recorded among known cold-active CEs. Notably, this is the first report of a psychrophilic CE. The psyCE can effectively produce epilactose at 8 °C, converting 20.3 % of 200 mM lactose into epilactose within four hours. These findings suggest that psyCE is highly suitable for cryogenic food processing, and glaciers may serve as a valuable repository of psychrophilic enzymes.


Asunto(s)
Carbohidrato Epimerasas , Celobiosa , Carbohidrato Epimerasas/genética , Carbohidrato Epimerasas/química , Carbohidrato Epimerasas/metabolismo , Celobiosa/química , Celobiosa/metabolismo , Cinética , Tibet , Simulación de Dinámica Molecular , Lactosa/metabolismo , Lactosa/química , Secuencia de Aminoácidos , Disacáridos
3.
Biotechnol J ; 19(8): e2400280, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39167550

RESUMEN

BACKGROUND: D-Allulose is one of the most well-known rare sugars widely used in food, cosmetics, and pharmaceutical industries. The most popular method for D-allulose production is the conversion from D-fructose catalyzed by D-allulose 3-epimerase (DAEase). To address the general problem of low catalytic efficiency and poor thermostability of wild-type DAEase, D-allulose biosensor was adopted in this study to develop a convenient and efficient method for high-throughput screening of DAEase variants. RESULTS: The catalytic activity and thermostability of DAEase from Caballeronia insecticola were simultaneously improved by semi-rational molecular modification. Compared with the wild-type enzyme, DAEaseS37N/F157Y variant exhibited 14.7% improvement in the catalytic activity and the half-time value (t1/2) at 65°C increased from 1.60 to 27.56 h by 17.23-fold. To our delight, the conversion rate of D-allulose was 33.6% from 500-g L-1 D-fructose in 1 h by Bacillus subtilis WB800 whole cells expressing this DAEase variant. Furthermore, the practicability of cell immobilization was evaluated and more than 80% relative activity of the immobilized cells was maintained from the second to seventh cycle. CONCLUSION: All these results indicated that the DAEaseS37N/F157Y variant would be a potential candidate for the industrial production of D-allulose.


Asunto(s)
Bacillus subtilis , Técnicas Biosensibles , Estabilidad de Enzimas , Fructosa , Técnicas Biosensibles/métodos , Fructosa/metabolismo , Bacillus subtilis/enzimología , Bacillus subtilis/genética , Carbohidrato Epimerasas/genética , Carbohidrato Epimerasas/metabolismo , Carbohidrato Epimerasas/química , Ingeniería de Proteínas/métodos , Racemasas y Epimerasas/genética , Racemasas y Epimerasas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/química , Temperatura
4.
Int J Mol Sci ; 25(12)2024 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-38928068

RESUMEN

As a low-calorie sugar, D-allulose is produced from D-fructose catalyzed by D-allulose 3-epimerase (DAE). Here, to improve the catalytic activity, stability, and processability of DAE, we reported a novel method by forming organic-inorganic hybrid nanoflowers (NF-DAEs) and co-immobilizing them on resins to form composites (Re-NF-DAEs). NF-DAEs were prepared by combining DAE with metal ions (Co2+, Cu2+, Zn2+, Ca2+, Ni2+, Fe2+, and Fe3+) in PBS buffer, and were analyzed by scanning electron microscopy (SEM), Fourier transform infrared spectroscopy, and X-ray diffraction. All of the NF-DAEs showed higher catalytic activities than free DAE, and the NF-DAE with Ni2+ (NF-DAE-Ni) reached the highest relative activity of 218%. The NF-DAEs improved the thermal stability of DAE, and the longest half-life reached 228 min for NF-DAE-Co compared with 105 min for the free DAE at 55 °C. To further improve the recycling performance of the NF-DAEs in practical applications, we combined resins and NF-DAEs to form Re-NF-DAEs. Resins and NF-DAEs co-effected the performance of the composites, and ReA (LXTE-606 neutral hydrophobic epoxy-based polypropylene macroreticular resins)-based composites (ReA-NF-DAEs) exhibited outstanding relative activities, thermal stabilities, storage stabilities, and processabilities. The ReA-NF-DAEs were able to be reused to catalyze the conversion from D-fructose to D-allulose, and kept more than 60% of their activities after eight cycles.


Asunto(s)
Estabilidad de Enzimas , Enzimas Inmovilizadas , Enzimas Inmovilizadas/química , Carbohidrato Epimerasas/química , Carbohidrato Epimerasas/metabolismo , Nanoestructuras/química , Fructosa/química , Espectroscopía Infrarroja por Transformada de Fourier , Difracción de Rayos X
5.
Int J Biol Macromol ; 273(Pt 1): 133027, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38857717

RESUMEN

D-allulose, a low-calorie rare sugar catalyzed by D-allulose 3-epimerase (DAE), is highly sought after for its potential health benefits. However, poor reusability and stability of DAE limited its popularization in industrial applications. Although metal-organic frameworks (MOFs) offer a promising enzyme platform for enzyme immobilization, developing customized strategies for MOF immobilization of enzymes remains challenging. In this study, we introduce a designable strategy involving the construction of bimetal-organic frameworks (ZnCo-MOF) based on metal ions compatibility. The DAE@MOFs materials were prepared and characterized, and the immobilization of DAE and the enzymatic characteristics of the MOF-immobilized DAE were subsequently evaluated. Remarkably, DAE@ZnCo-MOF exhibited superior recyclability which could maintain 95 % relative activity after 8 consecutive cycles. The storage stability is significantly improved compared to the free form, with a relative activity of 116 % remaining after 30 days. Molecular docking was also employed to investigate the interaction between DAE and the components of MOFs synthesis. The results demonstrate that the DAE@ZnCo-MOF exhibited enhanced catalytic efficiency and increased stability. This study introduces a viable and adaptable MOF-based immobilization strategy for enzymes, which holds the potential to expand the implementation of enzyme biocatalysts in a multitude of disciplines.


Asunto(s)
Enzimas Inmovilizadas , Estructuras Metalorgánicas , Simulación del Acoplamiento Molecular , Estructuras Metalorgánicas/química , Enzimas Inmovilizadas/química , Enzimas Inmovilizadas/metabolismo , Carbohidrato Epimerasas/química , Carbohidrato Epimerasas/metabolismo , Estabilidad de Enzimas , Iones/química , Fructosa
6.
Enzyme Microb Technol ; 179: 110465, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38852283

RESUMEN

Enzymatic production of D-mannose attracts increasing attention because of the health effects and commercial values of D-mannose. Several kinds of epimerases or isomerases have been used for enzymatic production of D-mannose from D-glucose or D-fructose. D-Mannose epimerase (MEase), belonging to N-acyl-D-glucosamine 2-epimerase superfamily enzymes, catalyzes the C-2 epimerization between D-glucose and D-mannose. In this study, a novel MEase was identified from Cytophagaceae bacterium SJW1-29. Sequence and structure alignments indicate that it is highly conserved with the reported R. slithyformis MEase with the known crystal structure. It was a metal-independent enzyme, with an optimal pH of 8.0 and an optimal temperature of 40 °C. The specific activities on D-glucose and D-mannose were 2.90 and 2.96 U/mg, respectively. The Km, kcat, and kcat/Km on D-glucose were measured to be 194.9 mM, 2.72 s-1, and 0.014 mM-1 s-1, respectively. The purified enzyme produced 23.15 g/L of D-mannose from 100 g/L of D-glucose at pH 8.0 and 40 °C for 8 h, with a conversion rate of 23.15 %.


Asunto(s)
Carbohidrato Epimerasas , Glucosa , Manosa , Manosa/metabolismo , Glucosa/metabolismo , Especificidad por Sustrato , Cinética , Carbohidrato Epimerasas/metabolismo , Carbohidrato Epimerasas/genética , Carbohidrato Epimerasas/química , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Concentración de Iones de Hidrógeno , Secuencia de Aminoácidos , Clonación Molecular , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/química , Temperatura , Modelos Moleculares , Alineación de Secuencia
7.
Int J Biol Macromol ; 269(Pt 1): 131986, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38697423

RESUMEN

D-allulose, a highly desirable sugar substitute, is primarily produced using the D-allulose 3-epimerase (DAE). However, the availability of usable DAE enzymes is limited. In this study, we discovered and engineered a novel DAE Rum55, derived from a human gut bacterium Ruminococcus sp. CAG55. The activity of Rum55 was strictly dependent on the presence of Co2+, and it exhibited an equilibrium conversion rate of 30.6 % and a half-life of 4.5 h at 50 °C. To enhance its performance, we engineered the interface interaction of Rum55 to stabilize its tetramer structure, and the best variant E268R was then attached with a self-assembling peptide to form active enzyme aggregates as carrier-free immobilization. The half-life of the best variant E268R-EKL16 at 50 °C was dramatically increased 30-fold to 135.3 h, and it maintained 90 % of its activity after 13 consecutive reaction cycles. Additionally, we identified that metal ions played a key role in stabilizing the tetramer structure of Rum55, and the dependence on metal ions for E268R-EKL16 was significantly reduced. This study provides a useful route for improving the thermostability of DAEs, opening up new possibilities for the industrial production of D-allulose.


Asunto(s)
Estabilidad de Enzimas , Ingeniería de Proteínas , Ruminococcus , Ruminococcus/enzimología , Ruminococcus/genética , Ingeniería de Proteínas/métodos , Péptidos/química , Péptidos/metabolismo , Carbohidrato Epimerasas/química , Carbohidrato Epimerasas/genética , Carbohidrato Epimerasas/metabolismo , Cinética , Modelos Moleculares , Fructosa/metabolismo , Fructosa/química
8.
Nat Commun ; 15(1): 3897, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38719841

RESUMEN

Understanding enzyme catalysis as connected to protein motions is a major challenge. Here, based on temperature kinetic studies combined with isotope effect measurements, we obtain energetic description of C-H activation in NAD-dependent UDP-glucuronic acid C4 epimerase. Approach from the ensemble-averaged ground state (GS) to the transition state-like reactive conformation (TSRC) involves, alongside uptake of heat ( Δ H ‡ = 54 kJ mol-1), significant loss in entropy ( - T Δ S ‡ = 20 kJ mol-1; 298 K) and negative activation heat capacity ( Δ C p ‡ = -0.64 kJ mol-1 K-1). Thermodynamic changes suggest the requirement for restricting configurational freedom at the GS to populate the TSRC. Enzyme variants affecting the electrostatic GS preorganization reveal active-site interactions important for precise TSRC sampling and H-transfer. Collectively, our study captures thermodynamic effects associated with TSRC sampling and establishes rigid positioning for C-H activation in an enzyme active site that requires conformational flexibility in fulfillment of its natural epimerase function.


Asunto(s)
Dominio Catalítico , Termodinámica , Cinética , Conformación Proteica , Carbohidrato Epimerasas/química , Carbohidrato Epimerasas/metabolismo , Carbohidrato Epimerasas/genética , Biocatálisis , Catálisis , Modelos Moleculares
9.
Bioprocess Biosyst Eng ; 47(6): 841-850, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38676737

RESUMEN

D-Allulose 3-epimerase (DAE) is a vital biocatalyst for the industrial synthesis of D-allulose, an ultra-low calorie rare sugar. However, limited thermostability of DAEs hinders their use at high-temperature production. In this research, hyperthermophilic TI-DAE (Tm = 98.4 ± 0.7 ℃) from Thermotoga sp. was identified via in silico screening. A comparative study of the structure and function of site-directed saturation mutagenesis mutants pinpointed the residue I100 as pivotal in maintaining the high-temperature activity and thermostability of TI-DAE. Employing TI-DAE as a biocatalyst, D-allulose was produced from D-fructose with a conversion rate of 32.5%. Moreover, TI-DAE demonstrated excellent catalytic synergy with glucose isomerase CAGI, enabling the one-step conversion of D-glucose to D-allulose with a conversion rate of 21.6%. This study offers a promising resource for the enzyme engineering of DAEs and a high-performance biocatalyst for industrial D-allulose production.


Asunto(s)
Thermotoga , Thermotoga/enzimología , Thermotoga/genética , Carbohidrato Epimerasas/genética , Carbohidrato Epimerasas/química , Carbohidrato Epimerasas/metabolismo , Carbohidrato Epimerasas/biosíntesis , Racemasas y Epimerasas/genética , Racemasas y Epimerasas/metabolismo , Racemasas y Epimerasas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/biosíntesis , Fructosa/metabolismo , Fructosa/biosíntesis , Fructosa/química , Estabilidad de Enzimas , Biocatálisis , Mutagénesis Sitio-Dirigida , Calor
10.
Enzyme Microb Technol ; 178: 110448, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38657401

RESUMEN

D-allulose is a naturally occurring rare sugar and beneficial to human health. However, the efficient biosynthesis of D-allulose remains a challenge. Here, we mined a new D-tagatose 3-epimerase from Kroppenstedtia eburnean (KeDt3e) with high catalytic efficiency. Initially, crucial factors contributing to the low conversion of KeDt3e were identified through crystal structure analysis, density functional theory calculations (DFT), and molecular dynamics (MD) simulations. Subsequently, based on the mechanism, combining restructuring the flexible region, proline substitution based onconsensus sequence analysis, introducing disulfide bonds, and grafting properties, and reshaping the active center, the optimal mutant M5 of KeDt3e was obtained with enhanced thermostability and activity. The optimal mutant M5 exhibited an enzyme activity of 130.8 U/mg, representing a 1.2-fold increase; Tm value increased from 52.7 °C to 71.2 °C; and half-life at 55 °C extended to 273.7 min, representing a 58.2-fold improvement, and the detailed mechanism of performance improvement was analyzed. Finally, by screening the ribosome-binding site (RBS) of the optimal mutant M5 recombinant bacterium (G01), the engineered strain G08 with higher expression levels was obtained. The engineered strain G08 catalyzed 500 g/L D-fructose to produce 172.4 g/L D-allulose, with a conversion of 34.4% in 0.5 h and productivity of 344.8 g/L/h on a 1 L scale. This study presents a promising approach for industrial-scale production of D-allulose.


Asunto(s)
Carbohidrato Epimerasas , Estabilidad de Enzimas , Hexosas , Hexosas/metabolismo , Carbohidrato Epimerasas/genética , Carbohidrato Epimerasas/metabolismo , Carbohidrato Epimerasas/química , Simulación de Dinámica Molecular , Fructosa/metabolismo , Cinética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/química , Especificidad por Sustrato , Ingeniería de Proteínas , Racemasas y Epimerasas/metabolismo , Racemasas y Epimerasas/genética , Racemasas y Epimerasas/química
11.
FEBS Lett ; 598(11): 1422-1437, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38649293

RESUMEN

Among the epimerases specific to alginate, some of them in Azotobacter genera convert ß-d-mannuronic acid to α-l-guluronic acid but also have lyase activity to degrade alginate. The remarkable characteristics of these epimerases make it a promising enzyme for tailoring alginates to meet specific demands. Here, we determined the structure of the bifunctional mannuronan C-5 epimerase AlgE3 from Azotobacter chroococcum (AcAlgE3) in complex with several mannuronic acid oligomers as well as in apo form, which allowed us to elucidate the binding manner of each mannuronic acid oligomer, and the structural plasticity, which is dependent on calcium ions. Moreover, a comprehensive analysis of the lyase activity profiles of AcAlgE3 combined with structural characteristics explained the preference for different chain length oligomers.


Asunto(s)
Alginatos , Azotobacter , Carbohidrato Epimerasas , Azotobacter/enzimología , Azotobacter/metabolismo , Alginatos/química , Alginatos/metabolismo , Carbohidrato Epimerasas/química , Carbohidrato Epimerasas/metabolismo , Carbohidrato Epimerasas/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Ácidos Hexurónicos/química , Ácidos Hexurónicos/metabolismo , Especificidad por Sustrato , Calcio/metabolismo , Calcio/química , Modelos Moleculares , Cristalografía por Rayos X , Unión Proteica , Dominio Catalítico
12.
Chembiochem ; 24(24): e202300555, 2023 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-37769151

RESUMEN

Uridine diphosphate N-acetylglucosamine 2-epimerase (GNE) is a key enzyme in the sialic acid biosynthesis pathway. Sialic acids are primarily terminal carbohydrates on glycans and play fundamental roles in health and disease. In search of effective GNE inhibitors not based on a carbohydrate scaffold, we performed a high-throughput screening campaign of 68,640 drug-like small molecules against recombinant GNE using a UDP detection assay. We validated nine of the primary actives with an orthogonal real-time NMR assay and verified their IC50 values in the low micromolar to nanomolar range manually. Stability and solubility studies revealed three compounds for further evaluation. Thermal shift assays, analytical size exclusion, and interferometric scattering microscopy demonstrated that the GNE inhibitors acted on the oligomeric state of the protein. Finally, hydrogen-deuterium exchange mass spectrometry (HDX-MS) revealed which sections of GNE were shifted upon the addition of the inhibitors. In summary, we have identified three small molecules as GNE inhibitors with high potency in vitro, which serve as promising candidates to modulate sialic acid biosynthesis in more complex systems.


Asunto(s)
Carbohidrato Epimerasas , Ácido N-Acetilneuramínico , Humanos , Carbohidrato Epimerasas/química , Carbohidrato Epimerasas/metabolismo , Ácidos Siálicos/química , Carbohidratos , Polisacáridos
13.
J Biol Chem ; 299(10): 105200, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37660908

RESUMEN

The sugar, 2,3-diacetamido-2,3-dideoxy-d-mannuronic acid, was first identified ∼40 years ago in the O-antigen of Pseudomonas aeruginosa O:3,a,d. Since then, it has been observed on the O-antigens of various pathogenic Gram-negative bacteria including Bordetella pertussis, Escherichia albertii, and Pseudomonas mediterranea. Previous studies have established that five enzymes are required for its biosynthesis beginning with uridine dinucleotide (UDP)-N-acetyl-d-glucosamine (UDP-GlcNAc). The final step in the pathway is catalyzed by a 2-epimerase, which utilizes UDP-2,3-diacetamido-2,3-dideoxy-d-glucuronic acid as its substrate. Curious as to whether this biochemical pathway is found in extreme thermophiles, we examined the published genome sequence for Thermus thermophilus HB27 and identified five ORFs that could possibly encode for the required enzymes. The focus of this investigation is on the ORF WP_011172736, which we demonstrate encodes for a 2-epimerase. For this investigation, ten high resolution X-ray crystallographic structures were determined to resolutions of 2.3 Å or higher. The models have revealed the manner in which the 2-epimerase anchors its UDP-sugar substrate as well as its UDP-sugar product into the active site. In addition, this study reveals for the first time the manner in which any sugar 2-epimerase can simultaneously bind UDP-sugars in both the active site and the allosteric binding region. We have also demonstrated that the T. thermophilus enzyme is allosterically regulated by UDP-GlcNAc. Whereas the sugar 2-epimerases that function on UDP-GlcNAc have been the focus of past biochemical and structural analyses, this is the first detailed investigation of a 2-epimerase that specifically utilizes UDP-2,3-diacetamido-2,3-dideoxy-d-glucuronic acid as its substrate.


Asunto(s)
Racemasas y Epimerasas , Azúcares , Thermus thermophilus , Carbohidrato Epimerasas/química , Dominio Catalítico , Antígenos O , Racemasas y Epimerasas/metabolismo , Azúcares de Uridina Difosfato , Thermus thermophilus/enzimología , Biocatálisis
14.
Acta Crystallogr D Struct Biol ; 79(Pt 7): 585-595, 2023 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-37314406

RESUMEN

Mannose 2-epimerase (ME), a member of the acylglucosamine 2-epimerase (AGE) superfamily that catalyzes epimerization of D-mannose and D-glucose, has recently been characterized to have potential for D-mannose production. However, the substrate-recognition and catalytic mechanism of ME remains unknown. In this study, structures of Runella slithyformis ME (RsME) and its D254A mutant [RsME(D254A)] were determined in their apo forms and as intermediate-analog complexes [RsME-D-glucitol and RsME(D254A)-D-glucitol]. RsME possesses the (α/α)6-barrel of the AGE superfamily members but has a unique pocket-covering long loop (loopα7-α8). The RsME-D-glucitol structure showed that loopα7-α8 moves towards D-glucitol and closes the active pocket. Trp251 and Asp254 in loopα7-α8 are only conserved in MEs and interact with D-glucitol. Kinetic analyses of the mutants confirmed the importance of these residues for RsME activity. Moreover, the structures of RsME(D254A) and RsME(D254A)-D-glucitol revealed that Asp254 is vital for binding the ligand in a correct conformation and for active-pocket closure. Docking calculations and structural comparison with other 2-epimerases show that the longer loopα7-α8 in RsME causes steric hindrance upon binding to disaccharides. A detailed substrate-recognition and catalytic mechanism for monosaccharide-specific epimerization in RsME has been proposed.


Asunto(s)
Manosa , Racemasas y Epimerasas , Manosa/metabolismo , Especificidad por Sustrato , Carbohidrato Epimerasas/química
15.
Essays Biochem ; 67(3): 615-627, 2023 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-36876890

RESUMEN

Alginate is a polysaccharide consisting of ß-D-mannuronate (M) and α-L-guluronate (G) produced by brown algae and some bacterial species. Alginate has a wide range of industrial and pharmaceutical applications, owing mainly to its gelling and viscosifying properties. Alginates with high G content are considered more valuable since the G residues can form hydrogels with divalent cations. Alginates are modified by lyases, acetylases, and epimerases. Alginate lyases are produced by alginate-producing organisms and by organisms that use alginate as a carbon source. Acetylation protects alginate from lyases and epimerases. Following biosynthesis, alginate C-5 epimerases convert M to G residues at the polymer level. Alginate epimerases have been found in brown algae and alginate-producing bacteria, predominantly Azotobacter and Pseudomonas species. The best characterised epimerases are the extracellular family of AlgE1-7 from Azotobacter vinelandii(Av). AlgE1-7 all consist of combinations of one or two catalytic A-modules and one to seven regulatory R-modules, but even though they are sequentially and structurally similar, they create different epimerisation patterns. This makes the AlgE enzymes promising for tailoring of alginates to have the desired properties. The present review describes the current state of knowledge regarding alginate-active enzymes with focus on epimerases, characterisation of the epimerase reaction, and how alginate epimerases can be used in alginate production.


Asunto(s)
Azotobacter vinelandii , Liasas , Racemasas y Epimerasas , Alginatos/química , Carbohidrato Epimerasas/química
16.
Acta Crystallogr D Struct Biol ; 78(Pt 9): 1180-1191, 2022 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-36048157

RESUMEN

D-Allulose, a low-calorie rare sugar with various physiological functions, is mainly produced through the isomerization of D-fructose by ketose 3-epimerases (KEases), which exhibit various substrate specificities. A novel KEase from a Clostridia bacterium (CDAE) was identified to be a D-allulose 3-epimerase and was further characterized as thermostable and metal-dependent. In order to explore its structure-function relationship, the crystal structure of CDAE was determined using X-ray diffraction at 2.10 Šresolution, revealing a homodimeric D-allulose 3-epimerase structure with extensive interactions formed at the dimeric interface that contribute to structure stability. Structural analysis identified the structural features of CDAE, which displays a common (ß/α)8-TIM barrel and an ordered Mn2+-binding architecture at the active center, which may explain the positive effects of Mn2+ on the activity and stability of CDAE. Furthermore, comparison of CDAE and other KEase structures revealed several structural differences, highlighting the remarkable differences in enzyme-substrate binding at the O4, O5 and O6 sites of the bound substrate, which are mainly induced by distinct hydrophobic pockets in the active center. The shape and hydrophobicity of this pocket appear to produce the differences in specificity and affinity for substrates among KEase family enzymes. Exploration of the crystal structure of CDAE provides a better understanding of its structure-function relationship, which might provide a basis for molecular modification of CDAE and further provides a reference for other KEases.


Asunto(s)
Carbohidrato Epimerasas , Racemasas y Epimerasas , Carbohidrato Epimerasas/química , Fructosa/química , Especificidad por Sustrato
17.
Appl Environ Microbiol ; 88(3): e0183621, 2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-34878812

RESUMEN

The structure and functional properties of alginates are dictated by the monomer composition and molecular weight distribution. Mannuronan C-5-epimerases determine the monomer composition by catalyzing the epimerization of ß-d-mannuronic acid (M) residues into α-l-guluronic acid (G) residues. The molecular weight is affected by alginate lyases, which catalyze a ß-elimination mechanism that cleaves alginate chains. The reaction mechanisms for the epimerization and lyase reactions are similar, and some enzymes can perform both reactions. These dualistic enzymes share high sequence identity with mannuronan C-5-epimerases without lyase activity. The mechanism behind their activity and the amino acid residues responsible for it are still unknown. We investigate mechanistic determinants involved in the bifunctional epimerase and lyase activity of AlgE7 from Azotobacter vinelandii. Based on sequence analyses, a range of AlgE7 variants were constructed and subjected to activity assays and product characterization by nuclear magnetic resonance (NMR) spectroscopy. Our results show that calcium promotes lyase activity, whereas NaCl reduces the lyase activity of AlgE7. By using defined polymannuronan (polyM) and polyalternating alginate (polyMG) substrates, the preferred cleavage sites of AlgE7 were found to be M|XM and G|XM, where X can be either M or G. From the study of AlgE7 mutants, R148 was identified as an important residue for the lyase activity, and the point mutant R148G resulted in an enzyme with only epimerase activity. Based on the results obtained in the present study, we suggest a unified catalytic reaction mechanism for both epimerase and lyase activities where H154 functions as the catalytic base and Y149 functions as the catalytic acid. IMPORTANCE Postharvest valorization and upgrading of algal constituents are promising strategies in the development of a sustainable bioeconomy based on algal biomass. In this respect, alginate epimerases and lyases are valuable enzymes for tailoring the functional properties of alginate, a polysaccharide extracted from brown seaweed with numerous applications in food, medicine, and material industries. By providing a better understanding of the catalytic mechanism and of how the two enzyme actions can be altered by changes in reaction conditions, this study opens further applications of bacterial epimerases and lyases in the enzymatic tailoring of alginate polymers.


Asunto(s)
Azotobacter vinelandii , Alginatos/metabolismo , Azotobacter vinelandii/genética , Carbohidrato Epimerasas/química , Ácidos Hexurónicos/metabolismo , Polisacárido Liasas/metabolismo
18.
Carbohydr Res ; 510: 108445, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34607125

RESUMEN

The synthesis of analogues of natural enzyme substrates can be used to help deduce enzymatic mechanisms. N-Acetylmannosamine-6-phosphate 2-epimerase is an enzyme in the bacterial sialic acid catabolic pathway. To investigate whether the mechanism of this enzyme involves a re-protonation mechanism by the same neighbouring lysine that performed the deprotonation or a unique substrate-assisted proton displacement mechanism involving the substrate C5 hydroxyl, the syntheses of two analogues of the natural substrate, N-acetylmannosamine-6-phosphate, are described. In these novel analogues, the C5 hydroxyl has been replaced with a proton and a methyl ether respectively. As recently reported, Staphylococcus aureus N-acetylmannosamine-6-phosphate 2-epimerase was co-crystallized with these two compounds. The 5-deoxy variant bound to the enzyme active site in a different orientation to the natural substrate, while the 5-methoxy variant did not bind, adding to the evidence that this enzyme uses a substrate-assisted proton displacement mechanism. This mechanistic information may help in the design of potential antibacterial drug candidates.


Asunto(s)
Proteínas Bacterianas/metabolismo , Carbohidrato Epimerasas/metabolismo , Hexosaminas/biosíntesis , Fosfatos de Azúcar/biosíntesis , Proteínas Bacterianas/química , Conformación de Carbohidratos , Carbohidrato Epimerasas/química , Hexosaminas/química , Staphylococcus aureus/enzimología , Fosfatos de Azúcar/química
19.
Int J Mol Sci ; 22(19)2021 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-34639151

RESUMEN

Thermal stability is a limiting factor for effective application of D-psicose 3-epimerase (DPEase) enzyme. Recently, it was reported that the thermal stability of DPEase was improved by immobilizing enzymes on graphene oxide (GO) nanoparticles. However, the detailed mechanism is not known. In this study, we investigated interaction details between GO and DPEase by performing molecular dynamics (MD) simulations. The results indicated that the domain (K248 to D268) of DPEase was an important anchor for immobilizing DPEase on GO surface. Moreover, the strong interactions between DPEase and GO can prevent loop α1'-α1 and ß4-α4 of DPEase from the drastic fluctuation. Since these two loops contained active site residues, the geometry of the active pocket of the enzyme remained stable at high temperature after the DPEase was immobilized by GO, which facilitated efficient catalytic activity of the enzyme. Our research provided a detailed mechanism for the interaction between GO and DPEase at the nano-biology interface.


Asunto(s)
Agrobacterium tumefaciens/enzimología , Carbohidrato Epimerasas/química , Enzimas Inmovilizadas/química , Grafito/química , Calor , Carbohidrato Epimerasas/metabolismo , Dominio Catalítico , Estabilidad de Enzimas , Concentración de Iones de Hidrógeno , Simulación de Dinámica Molecular , Conformación Proteica
20.
ACS Appl Mater Interfaces ; 13(41): 49433-49444, 2021 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-34612033

RESUMEN

Multienzymatic cascade reactions are a powerful strategy for straightforward and highly specific synthesis of complex materials, such as active substances in drugs. Cross-inhibitions and incompatible reaction steps, however, often limit enzymatic activity and thus the conversion. Such limitations occur, e.g., in the enzymatic synthesis of the biologically active sialic acid cytidine monophosphate N-acetylneuraminic acid (CMP-Neu5Ac). We addressed this challenge by developing a confinement and compartmentalization concept of hydrogel-immobilized enzymes for improving the efficiency of the enzyme cascade reaction. The three enzymes required for the synthesis of CMP-Neu5Ac, namely, N-acyl-d-glucosamine 2-epimerase (AGE), N-acetylneuraminate lyase (NAL), and CMP-sialic acid synthetase (CSS), were immobilized into bulk hydrogels and microstructured hydrogel-enzyme-dot arrays, which were then integrated into microfluidic devices. To overcome the cytidine triphosphate (CTP) cross-inhibition of AGE and NAL, only a low CTP concentration was applied and continuously conveyed through the device. In a second approach, the enzymes were compartmentalized in separate reaction chambers of the microfluidic device to completely avoid cross-inhibitions and enable the use of higher substrate concentrations. Immobilization efficiencies of up to 25% and pronounced long-term activity of the immobilized enzymes for several weeks were realized. Moreover, immobilized enzymes were less sensitive to inhibition and the substrate-channeling effect between immobilized enzymes promoted the overall conversion in the trienzymatic cascade reaction. Based on this, CMP-Neu5Ac was successfully synthesized by immobilized enzymes in noncompartmentalized and compartmentalized microfluidic devices. This study demonstrates the high potential of immobilizing enzymes in (compartmentalized) microfluidic devices to perform multienzymatic cascade reactions despite cross-inhibitions under continuous flow conditions. Due to the ease of enzyme immobilization in hydrogels, this concept is likely applicable for many cascade reactions with or without cross-inhibition characteristics.


Asunto(s)
Citidina Monofosfato/análogos & derivados , Enzimas Inmovilizadas/química , Hidrogeles/química , Ácidos Siálicos/síntesis química , Carbohidrato Epimerasas/química , Proteínas Portadoras/química , Citidina Monofosfato/síntesis química , Dispositivos Laboratorio en un Chip , Microfluídica/instrumentación , Microfluídica/métodos , N-Acilneuraminato Citidililtransferasa/química , Oxo-Ácido-Liasas/química , Polietilenglicoles/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...