Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 290
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Molecules ; 29(15)2024 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-39124931

RESUMEN

The study investigates the efficacy of an enzymatic preparation primarily with α-galactosidase activity for improving the quality of white sugar from poor-quality sugar beets. Focused on overcoming raffinose accumulation challenges in sugar beets, especially those harvested prematurely or stored for extended periods, an innovative exploration of enzymatic application in an industrial setting for the first time was conducted. By integrating theoretical calculations and experimental data, the findings reveal that α-galactosidase preparation notably diminishes raffinose content in beet juice, thus enhancing the sucrose yield and overall sugar quality. A reliable method to process lower-quality beets, promising enhanced efficiency in sugar production, was presented. The study also highlights the economic benefits of incorporating enzyme preparation into the production process, demonstrating a notable return on investment and underscoring the potential of enzymatic treatments to address industry challenges.


Asunto(s)
Beta vulgaris , Rafinosa , alfa-Galactosidasa , Rafinosa/química , Rafinosa/metabolismo , Beta vulgaris/química , alfa-Galactosidasa/metabolismo , alfa-Galactosidasa/química , Azúcares/química , Azúcares/metabolismo , Catálisis
2.
World J Microbiol Biotechnol ; 40(3): 91, 2024 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-38345638

RESUMEN

α-Galactosidase is an important exoglycosidase belonging to the hydrolase class of enzymes, which has therapeutic and industrial potential. It plays a crucial role in hydrolyzing α-1,6 linked terminal galacto-oligosaccharide residues such as melibiose, raffinose, and branched polysaccharides such as galacto-glucomannans and galactomannans. In this study, Actinoplanes utahensis B1 was explored for α-galactosidase production, yield improvement, and activity enhancement by purification. Initially, nine media components were screened using the Plackett-Burman design (PBD). Among these components, sucrose, soya bean flour, and sodium glutamate were identified as the best-supporting nutrients for the highest enzyme secretion by A. Utahensis B1. Later, the Central Composite Design (CCD) was implemented to fine-tune the optimization of these components. Based on sequential statistical optimization methodologies, a significant, 3.64-fold increase in α-galactosidase production, from 16 to 58.37 U/mL was achieved. The enzyme was purified by ultrafiltration-I followed by multimode chromatography and ultrafiltration-II. The purity of the enzyme was confirmed by Sodium Dodecyl Sulphate-Polyacrylamide Agarose Gel Electrophoresis (SDS-PAGE) which revealed a single distinctive band with a molecular weight of approximately 72 kDa. Additionally, it was determined that this process resulted in a 2.03-fold increase in purity. The purified α-galactosidase showed an activity of 2304 U/mL with a specific activity of 288 U/mg. This study demonstrates the isolation of Actinoplanes utahensis B1 and optimization of the process for the α-galactosidase production as well as single-step purification.


Asunto(s)
Actinoplanes , Oligosacáridos , alfa-Galactosidasa , alfa-Galactosidasa/química , Peso Molecular , Concentración de Iones de Hidrógeno
3.
Prep Biochem Biotechnol ; 53(4): 366-383, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-35801491

RESUMEN

α-Galactosidase hydrolyzes the α-1,6-linkage present at the non-reducing end of the sugars and results in the release of galactosyl residue from oligosaccharides like melibiose, raffinose, stachyose, etc. In the present study we report, α-galactosidase from Bacillus flexus isolated from Manikaran hot springs (India). Maximum enzyme production was obtained in guar gum and soybean meal after 72 h at 150 rpm. While, the temperature/pH of production was optimized at 50 °C and 7.0, respectively. Isoenzymes (α-gal I and II) were obtained and characterized based on temperature/pH optima along with their stability profile. JS27 α-Gal II was purified with a final purification fold of 11.54. Native and SDS-PAGE were used to determine the molecular weight of the enzyme as 86 and 41 kDa, respectively, indicating its homodimeric form. JS27 α-Gal II showed optimum enzyme activity at 55 °C and pH 7 (10 min). The enzyme displayed Km value of 2.3809 mM and Vmax of 2.0 × 104 µmol/min/ml with pNPG as substrate. JS27 α-Gal II demonstrated substrate hydrolysis and simultaneous formation of transgalactosylation products (α-GOS) with numerous substrates (sugar/sugar alcohols, oligosaccharides, and complex carbohydrates) which were verified by TLC and HPLC analysis. α-GOS are significant functional food ingredients and can be explored as prebiotics.


Asunto(s)
Manantiales de Aguas Termales , alfa-Galactosidasa , alfa-Galactosidasa/química , Oligosacáridos/química , Rafinosa
4.
Int J Mol Sci ; 22(19)2021 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-34639118

RESUMEN

An α-galactosidase-producing strain named Anoxybacillus vitaminiphilus WMF1, which catalyzed the reverse hydrolysis of d-galactose and glycerol to produce isofloridoside, was isolated from soil. The α-galactosidase (galV) gene was cloned and expressed in Escherichia coli. The galV was classified into the GH36 family with a molecular mass of 80 kDa. The optimum pH and temperature of galV was pH 7.5 and 60 °C, respectively, and it was highly stable at alkaline pH (6.0-9.0) and temperature below 65 °C. The specificity for p-nitrophenyl α-d-galactopyranoside was 70 U/mg, much higher than that for raffinose and stachyose. Among the metals and reagents tested, galV showed tolerance in the presence of various organic solvents. The kinetic parameters of the enzyme towards p-nitrophenyl α-d-galactopyranoside were obtained as Km (0.12 mM), Vmax (1.10 × 10-3 mM s-1), and Kcat/Km (763.92 mM-1 s-1). During the reaction of reverse hydrolysis, the enzyme exhibited high specificity towards the glycosyl donor galactose and acceptors glycerol, ethanol and ethylene glycol. Finally, the isofloridoside was synthesized using galactose as the donor and glycerol as the acceptor with a 26.6% conversion rate of galactose. This study indicated that galV might provide a potential enzyme source in producing isofloridoside because of its high thermal stability and activity.


Asunto(s)
Anoxybacillus/enzimología , Galactósidos/biosíntesis , Calor , alfa-Galactosidasa/metabolismo , Secuencia de Aminoácidos , Estabilidad de Enzimas , Concentración de Iones de Hidrógeno , Hidrólisis , Cinética , Peso Molecular , Homología de Secuencia , Especificidad por Sustrato , alfa-Galactosidasa/química
5.
Int J Mol Sci ; 22(12)2021 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-34204583

RESUMEN

Fabry disease (FD) is a lysosomal storage disease caused by mutations in the gene for the α-galactosidase A (GLA) enzyme. The absence of the enzyme or its activity results in the accumulation of glycosphingolipids, mainly globotriaosylceramide (Gb3), in different tissues, leading to a wide range of clinical manifestations. More than 1000 natural variants have been described in the GLA gene, most of them affecting proper protein folding and enzymatic activity. Currently, FD is treated by enzyme replacement therapy (ERT) or pharmacological chaperone therapy (PCT). However, as both approaches show specific drawbacks, new strategies (such as new forms of ERT, organ/cell transplant, substrate reduction therapy, or gene therapy) are under extensive study. In this review, we summarize GLA mutants described so far and discuss their putative application for the development of novel drugs for the treatment of FD. Unfavorable mutants with lower activities and stabilities than wild-type enzymes could serve as tools for the development of new pharmacological chaperones. On the other hand, GLA mutants showing improved enzymatic activity have been identified and produced in vitro. Such mutants could overcome several complications associated with current ERT, as lower-dose infusions of these mutants could achieve a therapeutic effect equivalent to that of the wild-type enzyme.


Asunto(s)
Enfermedad de Fabry/genética , Predisposición Genética a la Enfermedad , Mutación , alfa-Galactosidasa/genética , Alelos , Animales , Terapia Combinada/efectos adversos , Terapia Combinada/métodos , Manejo de la Enfermedad , Activación Enzimática , Enfermedad de Fabry/diagnóstico , Enfermedad de Fabry/metabolismo , Enfermedad de Fabry/terapia , Humanos , Relación Estructura-Actividad , Resultado del Tratamiento , alfa-Galactosidasa/química , alfa-Galactosidasa/metabolismo
6.
Molecules ; 26(5)2021 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-33669157

RESUMEN

α-Galacto-oligosaccharides (α-GOSs) have great functions as prebiotics and therapeutics. This work established the method of batch synthesis of α-GOSs by immobilized α-galactosidase for the first time, laying a foundation for industrial applications in the future. The α-galactosidase from Aspergillus niger L63 was immobilized as cross-linked enzyme aggregates (CLEAs) nano-biocatalyst through enzyme precipitating and cross-linking steps without using carriers. Among the tested agents, the ammonium sulfate showed high precipitation efficacy and induced regular structures of α-galactosidase CLEAs (Aga-CLEAs) that had been analyzed by scanning electron microscopy and Fourier-transform infrared spectroscopy. Through optimization by response surface methodology, the ammonium sulfate-induced Aga-CLEAs achieved a high activity recovery of around 90% at 0.55 U/mL of enzymes and 36.43 mM glutaraldehyde with cross-linking for 1.71 h. Aga-CLEAs showed increased thermal stability and organic solvent tolerance. The storage ability was also improved since it maintained 74.5% activity after storing at 4 °C for three months, significantly higher than that of the free enzyme (21.6%). Moreover, Aga-CLEAs exhibited excellent reusability in the α-GOSs synthesis from galactose, retaining above 66% of enzyme activity after 10 batch reactions, with product yields all above 30%.


Asunto(s)
Galactosa/biosíntesis , Oligosacáridos/biosíntesis , Prebióticos/análisis , alfa-Galactosidasa/metabolismo , Aspergillus niger/enzimología , Biocatálisis , Enzimas Inmovilizadas/química , Enzimas Inmovilizadas/metabolismo , Galactosa/química , Oligosacáridos/química , alfa-Galactosidasa/química
7.
Molecules ; 26(19)2021 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-34641615

RESUMEN

Pathogenic E. coli infection is one of the most widespread foodborne diseases, so the development of sensitive, reliable and easy operating detection tests is a key issue for food safety. Identifying bacteria with a fluorescent medium is more sensitive and faster than using chromogenic media. This study designed and synthesized a ß-galactosidase-activatable fluorescent probe BOD-Gal for the sensitive detection of E. coli. It employed a biocompatible and photostable 4,4-difluoro-3a,4a-diaza-s-indancene (BODIPY) as the fluorophore to form a ß-O-glycosidic bond with galactose, allowing the BOD-Gal to show significant on-off fluorescent signals for in vitro and in vivo bacterial detection. This work shows the potential for the use of a BODIPY based enzyme substrate for pathogen detection.


Asunto(s)
Compuestos de Boro/química , Escherichia coli/aislamiento & purificación , Colorantes Fluorescentes/química , Galactosa/metabolismo , alfa-Galactosidasa/metabolismo , Técnicas Biosensibles , Activación Enzimática , Escherichia coli/enzimología , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Microbiología de Alimentos , Galactosa/química , Sensibilidad y Especificidad , alfa-Galactosidasa/química
8.
Protein Expr Purif ; 175: 105710, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32738442

RESUMEN

Fabry disease is an X-linked lysosomal storage disorder caused by the deficiency of the enzyme, α-galactosidase A that induces the accumulation of the substrate globotriaosylceramide. Currently approved enzyme replacement therapy using recombinant human α-galactosidase A improves patient symptoms but a majority of patients experience adverse events due to the multiple infusions required for full therapeutic efficacy. Our approach is to use medicinal chemistry and phylogenic comparisons to introduce mutations into the human enzyme to increase catalytic activity and/or stability to generate an improved therapeutic enzyme that may require fewer infusions. We designed mutations at three regions of the human α-galactosidase A: the active site, the dimer interface, and a site for glycosylation. The M208E mutation, adjacent to the Y207 active site residue, increased enzyme activity 3.01-fold. This mutation introduced a charged Glu residue that is adjacent to the Y207 active site residue and close to a site of N-glycosylation. The W277C mutation, designed to promote dimer stability, introduced a strong thiol-aromatic interaction (Cys-Phe) at the dimer interface and increased activity 2.31-fold. The W277C and M208E mutations modify the structure of the enzyme into forms with enhanced thermal stability 3.7- and 3.9-fold, respectively and positive cooperativity resulting in increased Hill coefficient from 1.0 to 4.60 and 3.47, respectively. Enhanced thermal stability and positive cooperativity predict improved in vivo activity and superior therapeutic properties. Our results demonstrate the value of in vitro mutagenesis for α-galactosidase A and support future perspectives to validate these results in Fabry disease patients.


Asunto(s)
Sustitución de Aminoácidos , Enfermedad de Fabry , Mutagénesis , Multimerización de Proteína , alfa-Galactosidasa/química , Dominio Catalítico , Estabilidad de Enzimas/genética , Glicosilación , Calor , Humanos , Mutación Missense , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/uso terapéutico , alfa-Galactosidasa/genética , alfa-Galactosidasa/uso terapéutico
9.
Int J Mol Sci ; 21(19)2020 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-33036426

RESUMEN

Fabry disease, an X-linked recessive lysosomal disease, results from mutations in the GLA gene encoding lysosomal α-galactosidase A (α-Gal A). Due to these mutations, there is accumulation of globotriaosylceramide (GL-3) in plasma and in a wide range of cells throughout the body. Like other lysosomal enzymes, α-Gal A is synthesized on endoplasmic reticulum (ER) bound polyribosomes, and upon entry into the ER it undergoes glycosylation and folding. It was previously suggested that α-Gal A variants are recognized as misfolded in the ER and undergo ER-associated degradation (ERAD). In the present study, we used Drosophila melanogaster to model misfolding of α-Gal A mutants. We did so by creating transgenic flies expressing mutant α-Gal A variants and assessing development of ER stress, activation of the ER stress response and their relief with a known α-Gal A chaperone, migalastat. Our results showed that the A156V and the A285D α-Gal A mutants underwent ER retention, which led to activation of unfolded protein response (UPR) and ERAD. UPR could be alleviated by migalastat. When expressed in the fly's dopaminergic cells, misfolding of α-Gal A and UPR activation led to death of these cells and to a shorter life span, which could be improved, in a mutation-dependent manner, by migalastat.


Asunto(s)
1-Desoxinojirimicina/análogos & derivados , Lisosomas/enzimología , Pliegue de Proteína , alfa-Galactosidasa/química , 1-Desoxinojirimicina/química , 1-Desoxinojirimicina/farmacología , Animales , Animales Modificados Genéticamente , Encéfalo/metabolismo , Encéfalo/patología , Muerte Celular , Supervivencia Celular , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Drosophila melanogaster/enzimología , Retículo Endoplásmico/metabolismo , Degradación Asociada con el Retículo Endoplásmico , Enfermedad de Fabry/genética , Enfermedad de Fabry/metabolismo , Técnica del Anticuerpo Fluorescente , Pliegue de Proteína/efectos de los fármacos , Respuesta de Proteína Desplegada/efectos de los fármacos , alfa-Galactosidasa/genética
10.
Nat Chem Biol ; 13(4): 369-371, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28218913

RESUMEN

Protein folding in cells occurs in the presence of high concentrations of endogenous binding partners, and exogenous binding partners have been exploited as pharmacological chaperones. A combined mathematical modeling and experimental approach shows that a ligand improves the folding of a destabilized protein by biasing the kinetic partitioning between folding and alternative fates (aggregation or degradation). Computationally predicted inhibition of test protein aggregation and degradation as a function of ligand concentration are validated by experiments in two disparate cellular systems.


Asunto(s)
Ligandos , Chaperonas Moleculares/farmacología , Agregado de Proteínas/efectos de los fármacos , Pliegue de Proteína/efectos de los fármacos , Proteolisis/efectos de los fármacos , 1-Desoxinojirimicina/análogos & derivados , 1-Desoxinojirimicina/metabolismo , 1-Desoxinojirimicina/farmacología , Escherichia coli/química , Escherichia coli/genética , Escherichia coli/metabolismo , Células HEK293 , Humanos , Cinética , Modelos Moleculares , Reproducibilidad de los Resultados , Tetrahidrofolato Deshidrogenasa/química , Tetrahidrofolato Deshidrogenasa/metabolismo , Trimetoprim/metabolismo , Trimetoprim/farmacología , alfa-Galactosidasa/química , alfa-Galactosidasa/metabolismo
11.
Microb Cell Fact ; 18(1): 172, 2019 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-31601209

RESUMEN

BACKGROUND: α-Galactosidases are enzymes that act on galactosides present in many vegetables, mainly legumes and cereals, have growing importance with respect to our diet. For this reason, the use of their catalytic activity is of great interest in numerous biotechnological applications, especially those in the food industry directed to the degradation of oligosaccharides derived from raffinose. The aim of this work has been to optimize the recombinant production and further characterization of α-galactosidase of Saccharomyces cerevisiae. RESULTS: The MEL1 gene coding for the α-galactosidase of S. cerevisiae (ScAGal) was cloned and expressed in the S. cerevisiae strain BJ3505. Different constructions were designed to obtain the degree of purification necessary for enzymatic characterization and to improve the productive process of the enzyme. ScAGal has greater specificity for the synthetic substrate p-nitrophenyl-α-D-galactopyranoside than for natural substrates, followed by the natural glycosides, melibiose, raffinose and stachyose; it only acts on locust bean gum after prior treatment with ß-mannosidase. Furthermore, this enzyme strongly resists proteases, and shows remarkable activation in their presence. Hydrolysis of galactose bonds linked to terminal non-reducing mannose residues of synthetic galactomannan-oligosaccharides confirms that ScAGal belongs to the first group of α-galactosidases, according to substrate specificity. Optimization of culture conditions by the statistical model of Response Surface helped to improve the productivity by up to tenfold when the concentration of the carbon source and the aeration of the culture medium was increased, and up to 20 times to extend the cultivation time to 216 h. CONCLUSIONS: ScAGal characteristics and improvement in productivity that have been achieved contribute in making ScAGal a good candidate for application in the elimination of raffinose family oligosaccharides found in many products of the food industry.


Asunto(s)
Rafinosa/metabolismo , Proteínas de Saccharomyces cerevisiae/biosíntesis , Saccharomyces cerevisiae/enzimología , alfa-Galactosidasa/biosíntesis , Cinética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/química , Especificidad por Sustrato , alfa-Galactosidasa/química
12.
Microb Cell Fact ; 18(1): 180, 2019 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-31647018

RESUMEN

BACKGROUND: Structurally stable α-galactosidases are of great interest for various biotechnological applications. More thermophilic α-galactosidases with high activity and structural stability have therefore to be mined and characterized. On the other hand, few studies have been performed to prominently enhance the AOX1 promoter activity in the commonly used Pichia pastoris system, in which production of some heterologous proteins are insufficient for further study. RESULTS: ReGal2 encoding a thermoactive α-galactosidase was identified from the thermophilic (hemi)cellulolytic fungus Rasamsonia emersonii. Significantly increased production of ReGal2 was achieved when ReGal2 was expressed in an engineered Pastoris pichia expression system with a modified AOX1 promoter and simultaneous fortified expression of Mxr1 that is involved in transcriptionally activating AOX1. Purified ReGal2 exists as an oligomer and has remarkable thermo-activity and thermo-tolerance, exhibiting maximum activity of 935 U/mg towards pNPGal at 80 °C and retaining full activity after incubation at 70 °C for 60 h. ReGal2 is insensitive to treatments by many metal ions and exhibits superior tolerance to protein denaturants. Moreover, ReGal2 efficiently hydrolyzed stachyose and raffinose in soybeans at 70 °C in 3 h and 24 h, respectively. CONCLUSION: A modified P. pichia expression system with significantly enhanced AOX1 promoter activity has been established, in which ReGal2 production is markedly elevated to facilitate downstream purification and characterization. Purified ReGal2 exhibited prominent features in thermostability, catalytic activity, and resistance to protein denaturants. ReGal2 thus holds great potential in relevant biotechnological applications.


Asunto(s)
Eurotiales/enzimología , Pichia , Proteínas Recombinantes , alfa-Galactosidasa , Clonación Molecular , Proteínas Fúngicas/genética , Expresión Génica , Cinética , Pichia/genética , Pichia/metabolismo , Regiones Promotoras Genéticas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , alfa-Galactosidasa/química , alfa-Galactosidasa/genética , alfa-Galactosidasa/metabolismo
13.
J Sci Food Agric ; 99(10): 4748-4760, 2019 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-30932192

RESUMEN

BACKGROUND: Cross-linked enzyme aggregates (CLEAs) of α-galactosidase, partially purified from maize (Zea mays) flour, were prepared. The impact of various parameters on enzyme activity was examined to optimize the immobilization procedure. Biochemical characterization of the free and immobilized enzyme was carried out. Stability (thermal, pH, storage and operational stability) and reusability tests were performed. The potential use of the free enzyme and the CLEAs in hydrolysis processes of raffinose-type oligosaccharides present in soymilk was investigated. RESULTS: α-galactosidase CLEAs were prepared with 47% activity recovery under optimum conditions [1:5 (v/v) enzyme solution:saturated ammonium sulfate solution ratio; 7.5 mg protein and 0.1% (v/v) glutaraldehyde, 6 h, 4 °C, 150 rpm]. α-galactosidase CLEAs exhibited increased stability in comparison to the free enzyme. The CLEAs and the free enzyme showed a maximum activity at 40°C and their optimal pH values were5.5 and 6.0, respectively. Kinetic constants (KM , Vmax and kcat ) were calculated for the free enzyme and the CLEAs in the presence of p-nitrophenyl-α-d-galactopyranoside, stachyose, melibiose and raffinose. The effect of various chemicals and sugars on enzyme activity showed that both enzyme forms were significantly inhibited by HgCl2 and galactose. The CLEAs hydrolyzed 85% of raffinose and 96% of stachyose. CONCLUSION: The α-galactosidase CLEAs, with their satisfactory enzymatic characteristics, have much potential for use in the food and feed industry. © 2019 Society of Chemical Industry.


Asunto(s)
Oligosacáridos/química , Rafinosa/química , Leche de Soja/química , alfa-Galactosidasa/química , Biocatálisis , Reactivos de Enlaces Cruzados/química , Estabilidad de Enzimas , Enzimas Inmovilizadas/química , Galactosa/química , Concentración de Iones de Hidrógeno , Hidrólisis , Cinética , Temperatura
14.
World J Microbiol Biotechnol ; 35(2): 32, 2019 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-30701316

RESUMEN

Extremophilic microorganisms are valuable sources of enzymes for various industrial applications. In fact, given their optimal catalytic activity and operational stability under harsh physical and chemical conditions, they represent a suitable alternative to their mesophilic counterparts. For instance, extremophilic enzymes are important to foster the switch from fossil-based to lignocellulose-based industrial processes. Indeed, more stable enzymes are needed, because the conversion of the lignocellulosic biomass to a wide palette of value-added products requires extreme chemo-physical pre-treatments. Galactomannans are part of the hemicellulose fraction in lignocellulosic biomass. They are heteropolymers constituted by a ß-1,4-linked mannan backbone substituted with side chains of α-1,6-linked galactose residues. Therefore, the joint action of different hydrolytic enzymes (i.e. ß-mannanase, ß-mannosidase and α-galactosidase) is needed to accomplish their complete hydrolysis. So far, numerous galactomannan-degrading enzymes have been isolated and characterized from extremophilic microorganisms. Besides applications in biorefinery, these biocatalysts are also useful to improve the quality (i.e. digestibility and prebiotic properties) of food and feed as well as in paper industries to aid the pulp bleaching process. In this review, an overview about the structure, function and applications of galactomannans is provided. Moreover, a survey of (hyper)-thermophilic galactomannans-degrading enzymes, mainly characterized in the last decade, has been carried out. These extremozymes are described in the light of their biotechnological application in industrial processes requiring harsh conditions.


Asunto(s)
Bacterias/enzimología , Mananos/metabolismo , Manosidasas/química , alfa-Galactosidasa/química , beta-Manosidasa/química , Bacterias/química , Bacterias/genética , Biotecnología , Estabilidad de Enzimas , Galactosa/análogos & derivados , Mananos/química , Manosidasas/genética , Manosidasas/metabolismo , Plantas/química , Plantas/enzimología , Plantas/genética , Plantas/metabolismo , alfa-Galactosidasa/genética , alfa-Galactosidasa/metabolismo , beta-Manosidasa/genética , beta-Manosidasa/metabolismo
15.
Bioconjug Chem ; 29(5): 1630-1639, 2018 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-29698600

RESUMEN

The current treatment of Fabry disease by enzyme replacement therapy with commercially available recombinant human α-Galactosidase A shows a continuous deterioration of the disease patients. Human recombinant α-Galactosidase A is a homodimer with noncovalently bound subunits and is expressed in the ProCellEx plant cell-based protein expression platform to produce pegunigalsidase alfa. The effect of covalent bonding between two α-Galactosidase A subunits by PEG-based cross-linkers of various lengths was evaluated in this study. The results show that cross-linking by a bifunctional PEG polymer of 2000 Da produces a more stable protein with improved pharmacokinetic and biodistribution properties. The chemical modification did not influence the tertiary protein structure but led to an increased thermal stability and showed partial masking of immune epitopes. The developed pegunigalsidase alfa is currently tested in phase III clinical trials and has a potential to show superior efficacy versus the currently used enzyme replacement therapies in the treatment of Fabry disease patients.


Asunto(s)
Reactivos de Enlaces Cruzados/química , Polietilenglicoles/química , alfa-Galactosidasa/química , Animales , Línea Celular , Estabilidad de Enzimas , Enfermedad de Fabry/tratamiento farmacológico , Humanos , Ratones , Modelos Moleculares , Conformación Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/uso terapéutico , Distribución Tisular , Nicotiana/genética , alfa-Galactosidasa/genética , alfa-Galactosidasa/farmacocinética , alfa-Galactosidasa/uso terapéutico
16.
Bioconjug Chem ; 29(3): 649-656, 2018 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-29285931

RESUMEN

Enzymes are attractive as immunotherapeutics because they can catalyze shifts in the local availability of immunostimulatory and immunosuppressive signals. Clinical success of enzyme immunotherapeutics frequently hinges upon achieving sustained biocatalysis over relevant time scales. The time scale and location of biocatalysis are often dictated by the location of the substrate. For example, therapeutic enzymes that convert substrates distributed systemically are typically designed to have a long half-life in circulation, whereas enzymes that convert substrates localized to a specific tissue or cell population can be more effective when designed to accumulate at the target site. This Topical Review surveys approaches to improve enzyme immunotherapeutic efficacy via chemical modification, encapsulation, and immobilization that increases enzyme accumulation at target sites or extends enzyme half-life in circulation. Examples provided illustrate "replacement therapies" to restore deficient enzyme function, as well as "enhancement therapies" that augment native enzyme function via supraphysiologic doses. Existing FDA-approved enzyme immunotherapies are highlighted, followed by discussion of emerging experimental strategies such as those designed to enhance antitumor immunity or resolve inflammation.


Asunto(s)
Terapia Enzimática/métodos , Inmunoterapia/métodos , Animales , Antiinflamatorios/química , Antiinflamatorios/inmunología , Antiinflamatorios/uso terapéutico , Antineoplásicos/química , Antineoplásicos/inmunología , Antineoplásicos/uso terapéutico , Asparaginasa/química , Asparaginasa/inmunología , Asparaginasa/uso terapéutico , Biocatálisis , Enzimas Inmovilizadas/química , Enzimas Inmovilizadas/inmunología , Enzimas Inmovilizadas/uso terapéutico , Enfermedad de Fabry/inmunología , Enfermedad de Fabry/terapia , Enfermedad de Gaucher/inmunología , Enfermedad de Gaucher/terapia , Glucosilceramidasa/química , Glucosilceramidasa/inmunología , Glucosilceramidasa/uso terapéutico , Glicosilación , Humanos , Inmunoconjugados/química , Inmunoconjugados/inmunología , Inmunoconjugados/uso terapéutico , Inflamación/inmunología , Inflamación/terapia , Enfermedades por Almacenamiento Lisosomal/inmunología , Enfermedades por Almacenamiento Lisosomal/terapia , Neoplasias/inmunología , Neoplasias/terapia , alfa-Galactosidasa/química , alfa-Galactosidasa/inmunología , alfa-Galactosidasa/uso terapéutico
17.
Mar Drugs ; 16(10)2018 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-30250010

RESUMEN

A novel wild-type recombinant cold-active α-d-galactosidase (α-PsGal) from the cold-adapted marine bacterium Pseudoalteromonas sp. KMM 701, and its mutants D451A and C494N, were studied in terms of their structural, physicochemical, and catalytic properties. Homology models of the three-dimensional α-PsGal structure, its active center, and complexes with D-galactose were constructed for identification of functionally important amino acid residues in the active site of the enzyme, using the crystal structure of the α-galactosidase from Lactobacillus acidophilus as a template. The circular dichroism spectra of the wild α-PsGal and mutant C494N were approximately identical. The C494N mutation decreased the efficiency of retaining the affinity of the enzyme to standard p-nitrophenyl-α-galactopiranoside (pNP-α-Gal). Thin-layer chromatography, matrix-assisted laser desorption/ionization mass spectrometry, and nuclear magnetic resonance spectroscopy methods were used to identify transglycosylation products in reaction mixtures. α-PsGal possessed a narrow acceptor specificity. Fructose, xylose, fucose, and glucose were inactive as acceptors in the transglycosylation reaction. α-PsGal synthesized -α(1→6)- and -α(1→4)-linked galactobiosides from melibiose as well as -α(1→6)- and -α(1→3)-linked p-nitrophenyl-digalactosides (Gal2-pNP) from pNP-α-Gal. The D451A mutation in the active center completely inactivated the enzyme. However, the substitution of C494N discontinued the Gal-α(1→3)-Gal-pNP synthesis and increased the Gal-α(1→4)-Gal yield compared to Gal-α(1→6)-Gal-pNP.


Asunto(s)
Proteínas Bacterianas/metabolismo , Modelos Químicos , Pseudoalteromonas/metabolismo , alfa-Galactosidasa/metabolismo , Aclimatación , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/aislamiento & purificación , Frío , Pruebas de Enzimas , Glicosilación , Mutagénesis Sitio-Dirigida , Mutación , Pseudoalteromonas/genética , Pseudoalteromonas/fisiología , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato , alfa-Galactosidasa/química , alfa-Galactosidasa/genética , alfa-Galactosidasa/aislamiento & purificación
18.
J Sci Food Agric ; 98(4): 1502-1510, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28799644

RESUMEN

BACKGROUND: We synthesised a novel sericin peptide (SP-GI) with α-d-glucosidase inhibitory activity, which has a sequence of SEDSSEVDIDLGN. The kinetics of its peptide-induced inhibition on α-d-glucosidase activity and its interaction mechanism merging with molecular docking were both investigated. RESULTS: SP-GI exhibited significant inhibitory activity with an IC50 of 2.9 ± 0.1 µmol L-1 and this inhibition was reversible and non-competitive with a Ki value of 1.0 ± 0.1 µmol L-1 . An interaction study with SP-GI revealed it bound to α-d-glucosidase at a single binding site, resulting in alterations in α-d-glucosidase secondary structure. This led to quenching of intrinsic α-d-glucosidase fluorescence by a static quenching mechanism. Molecular docking results showed that the SP-GI binding site on α-d-glucosidase differed from acarbose, with hydrogen bonding and van der Waals forces being the main binding drivers. CONCLUSION: These findings suggest the potential use for SP-GI or other natural sericin peptides as dietary supplements for the treatment of type 2 diabetes. © 2017 Society of Chemical Industry.


Asunto(s)
Inhibidores de Glicósido Hidrolasas/química , Péptidos/química , Proteínas de Saccharomyces cerevisiae/antagonistas & inhibidores , Sericinas/química , alfa-Galactosidasa/antagonistas & inhibidores , Sitios de Unión , Cinética , Simulación del Acoplamiento Molecular , Péptidos/síntesis química , Unión Proteica , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/enzimología , Proteínas de Saccharomyces cerevisiae/química , Sericinas/síntesis química , alfa-Galactosidasa/química
19.
World J Microbiol Biotechnol ; 34(2): 19, 2018 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-29302817

RESUMEN

α-Galactosidases are assigned to the class of hydrolases and the subclass of glycoside hydrolases (GHs). They belong to six GH families and include the only characterized α-galactosidases from yeasts (GH 27, Saccharomyces cerevisiae). The present study focuses on an investigation of the lactose-inducible α-galactosidase produced by Papiliotrema flavescens. The enzyme was present on the surface of cells and in the cytosol. Its temperature optimum was about 60 °C and the pH optimum was 4.8; the pH stability ranged from 3.2 to 6.6. This α-galactosidase also exhibited transglycosylation activity. The cytosol α-galactosidase with a molecular weight about 110 kDa, was purified using a combination of liquid chromatography techniques. Three intramolecular peptides were determined by the partial structural analysis of the sequences of the protein isolated, using MALDI-TOF/TOF mass spectrometry. The data obtained recognized the first yeast α-galactosidase, which belongs to the GH 36 family. The bioinformatics analysis and homology modeling of a 210 amino acids long C-terminal sequence (derived from cDNA) confirmed the correctness of these findings. The study was also supplemented by the screening of capsular cryptococcal yeasts, which produce the surface lactose-inducible α- and ß-galactosidases. The production of the lactose-inducible α-galactosidases was not found to be a general feature within the yeast strains examined and, therefore, the existing hypothesis on the general function of this enzyme in cryptococcal capsule rearrangement cannot be confirmed.


Asunto(s)
Basidiomycota/enzimología , Proteínas Fúngicas/química , Proteínas Fúngicas/metabolismo , alfa-Galactosidasa/química , alfa-Galactosidasa/metabolismo , Secuencia de Aminoácidos , Basidiomycota/clasificación , Basidiomycota/genética , Basidiomycota/crecimiento & desarrollo , Cryptococcus , Citosol/enzimología , ADN Complementario , ADN de Hongos/genética , Estabilidad de Enzimas , Proteínas Fúngicas/genética , Proteínas Fúngicas/aislamiento & purificación , Genes Fúngicos/genética , Glicósido Hidrolasas/metabolismo , Concentración de Iones de Hidrógeno , Lactosa/metabolismo , Modelos Moleculares , Peso Molecular , Conformación Proteica , Alineación de Secuencia , Análisis de Secuencia de Proteína , Especificidad por Sustrato , Temperatura , alfa-Galactosidasa/genética , alfa-Galactosidasa/aislamiento & purificación
20.
Angew Chem Int Ed Engl ; 57(18): 5033-5036, 2018 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-29490117

RESUMEN

Self-assembly of nanoparticles provides unique opportunities as nanoplatforms for controlled delivery. By exploiting the important role of noncovalent hydrophobic interactions in the engineering of stable assemblies, nanoassemblies were formed by the self-assembly of fluorinated quantum dots in aqueous medium through fluorine-fluorine interactions. These nanoassemblies encapsulated different enzymes (laccase and α-galactosidase) with encapsulation efficiencies of ≥74 %. Importantly, the encapsulated enzymes maintained their catalytic activity, following Michaelis-Menten kinetics. Under an acidic environment the nanoassemblies were slowly disassembled, thus allowing the release of encapsulated enzymes. The effective release of the assayed enzymes demonstrated the feasibility of this nanoplatform to be used in pH-mediated enzyme delivery. In addition, the as-synthesized nanoassemblies, having a diameter of about 50 nm, presented high colloidal stability and fluorescence emission, which make them a promising multifunctional nanoplatform.


Asunto(s)
Flúor/química , Lacasa/química , Puntos Cuánticos/química , alfa-Galactosidasa/química , Flúor/metabolismo , Concentración de Iones de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Lacasa/metabolismo , Tamaño de la Partícula , Puntos Cuánticos/metabolismo , Propiedades de Superficie , alfa-Galactosidasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA