Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Biomacromolecules ; 2024 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-39025475

RESUMEN

Glycoside phosphorylases are enzymes that are frequently used for polysaccharide synthesis. Some of these enzymes have broad substrate specificity, enabling the synthesis of reducing-end-functionalized glucan chains. Here, we explore the potential of glycoside phosphorylases in synthesizing chromophore-conjugated polysaccharides using commercially available chromophoric model compounds as glycosyl acceptors. Specifically, we report cellulose and ß-1,3-glucan synthesis using 2-nitrophenyl ß-d-glucopyranoside, 4-nitrophenyl ß-d-glucopyranoside, and 2-methoxy-4-(2-nitrovinyl)phenyl ß-d-glucopyranoside with Clostridium thermocellum cellodextrin phosphorylase and Thermosipho africanus ß-1,3-glucan phosphorylase as catalysts. We demonstrate activity for both enzymes with all assayed chromophoric acceptors and report the crystallization-driven precipitation and detailed structural characterization of the synthesized polysaccharides, i.e., their molar mass distributions and various structural parameters, such as morphology, fibril diameter, lamellar thickness, and crystal form. Our results provide insights for the studies of chromophore-conjugated low molecular weight polysaccharides, glycoside phosphorylases, and the hierarchical assembly of crystalline cellulose and ß-1,3-glucan.

2.
Mol Pharm ; 18(8): 3125-3131, 2021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-34296616

RESUMEN

Halogenation can be utilized for the purposes of labeling and molecular imaging, providing a means to, e.g., follow drug distribution in an organism through positron emission tomography (PET) or study the molecular recognition events unfolding by nuclear magnetic resonance (NMR) spectroscopy. For cancer therapeutics, where often highly toxic substances are employed, it is of importance to be able to track the distribution of the drugs and their metabolites in order to ensure minimal side effects. Labeling should ideally have a negligible disruptive effect on the efficacy of a given drug. Using a combination of NMR spectroscopy and cytotoxicity assays, we identify a site susceptible to halogenation in monomethyl auristatin F (MMAF), a widely used cytotoxic agent in the antibody-drug conjugate (ADC) family of cancer drugs, and study the effects of fluorination and chlorination on the physiological solution structure of the auristatins and their cytotoxicity. We find that the cytotoxicity of the parent drug is retained, while the conformational equilibrium is shifted significantly toward the biologically active trans isomer, simultaneously decreasing the concentration of the inactive and potentially disruptive cis isomer by up to 50%. Our results may serve as a base for the future assembly of a multifunctional toolkit for the assessment of linker technologies and exploring bystander effects from the warhead perspective in auristatin-derived ADCs.


Asunto(s)
Antineoplásicos/química , Citotoxinas/química , Halogenación , Inmunoconjugados/química , Neoplasias/metabolismo , Oligopéptidos/química , Fenilalanina/química , Aminobenzoatos/química , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Isomerismo , Espectroscopía de Resonancia Magnética/métodos , Ratones , Conformación Molecular , Neoplasias/patología
3.
Mol Pharm ; 18(1): 285-304, 2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-33390018

RESUMEN

Boron neutron capture therapy (BNCT) is a noninvasive binary therapeutic modality applicable to the treatment of cancers. While BNCT offers a tumor-targeting selectivity that is difficult to match by other means, the last obstacles preventing the full harness of this potential come in the form of the suboptimal boron delivery strategies presently used in the clinics. To address these challenges, we have developed delivery agents that target the glucose transporter GLUT1. Here, we present the chemical synthesis of a number of ortho-carboranylmethyl-substituted glucoconjugates and the biological assessment of all positional isomers. Altogether, the study provides protocols for the synthesis and structural characterization of such glucoconjugates and insights into their essential properties, for example, cytotoxicity, GLUT1-affinity, metabolism, and boron delivery capacity. In addition to solidifying the biochemical foundations of a successful GLUT1-targeting approach to BNCT, we identify the most promising modification sites in d-glucose, which are critical in order to further develop this strategy toward clinical use.


Asunto(s)
Boro/administración & dosificación , Boro/química , Neoplasias Encefálicas/radioterapia , Transportador de Glucosa de Tipo 1/metabolismo , Compuestos de Boro/administración & dosificación , Compuestos de Boro/química , Terapia por Captura de Neutrón de Boro/métodos , Línea Celular Tumoral , Glucosa/metabolismo , Humanos
4.
J Ind Microbiol Biotechnol ; 48(5-6)2021 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-33899921

RESUMEN

Polyhydroxyalkanoates (PHAs) provide biodegradable and bio-based alternatives to conventional plastics. Incorporation of 2-hydroxy acid monomers into polymer, in addition to 3-hydroxy acids, offers possibility to tailor the polymer properties. In this study, poly(D-lactic acid) (PDLA) and copolymer P(LA-3HB) were produced and characterized for the first time in the yeast Saccharomyces cerevisiae. Expression of engineered PHA synthase PhaC1437Ps6-19, propionyl-CoA transferase Pct540Cp, acetyl-CoA acetyltransferase PhaA, and acetoacetyl-CoA reductase PhaB1 resulted in accumulation of 3.6% P(LA-3HB) and expression of engineered enzymes PhaC1Pre and PctMe resulted in accumulation of 0.73% PDLA of the cell dry weight (CDW). According to NMR, P(LA-3HB) contained D-lactic acid repeating sequences. For reference, expression of PhaA, PhaB1, and PHA synthase PhaC1 resulted in accumulation 11% poly(hydroxybutyrate) (PHB) of the CDW. Weight average molecular weights of these polymers were comparable to similar polymers produced by bacterial strains, 24.6, 6.3, and 1 130 kDa for P(LA-3HB), PDLA, and PHB, respectively. The results suggest that yeast, as a robust and acid tolerant industrial production organism, could be suitable for production of 2-hydroxy acid containing PHAs from sugars or from 2-hydroxy acid containing raw materials. Moreover, the wide substrate specificity of PHA synthase enzymes employed increases the possibilities for modifying copolymer properties in yeast in the future.


Asunto(s)
Ácido Láctico/metabolismo , Polihidroxialcanoatos/biosíntesis , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Acetil-CoA C-Acetiltransferasa/genética , Acetil-CoA C-Acetiltransferasa/metabolismo , Aciltransferasas/genética , Aciltransferasas/metabolismo , Oxidorreductasas de Alcohol/genética , Oxidorreductasas de Alcohol/metabolismo , Coenzima A Transferasas/genética , Coenzima A Transferasas/metabolismo , Escherichia coli/metabolismo , Ingeniería Genética , Hidroxibutiratos/metabolismo , Microbiología Industrial , Redes y Vías Metabólicas , Polihidroxialcanoatos/química
5.
Glycoconj J ; 37(2): 221-229, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31792892

RESUMEN

Fucosylated oligosaccharides are interesting molecules due to their bioactive properties. In particular, their application as active ingredient in milk powders is attractive for dairy industries. The objective of this study was to characterize the glycosyl hydrolase family 29 α-fucosidase produced by Aspergillus niger and test its ability to transfucosylate lactose with a view towards potential industrial applications such as the valorization of the lactose side stream produced by dairy industry. In order to reduce costs and toxicity the use of free fucose instead of environmentally questionable fucose derivatives was studied. In contrast to earlier studies, a recombinantly produced A. niger α-fucosidase was utilized. Using pNP-fucose as substrate, the optimal pH for hydrolytic activity was determined to be 3.8. The optimal temperature for a 30-min reaction was 60 °C, and considering temperature stability, the optimal temperature for a 24-h reaction was defined as 45 °C For the same hydrolysis reaction, the kinetic values were calculated to be 0.385 mM for the KM and 2.8 mmol/(mg*h) for the Vmax. Transfucosylation of lactose occurred at high substrate concentrations when reaction time was elongated to several days. The structure of the product trisaccharide was defined as 1-fucosyllactose, where fucose is α-linked to the anomeric carbon of the ß-glucose moiety of lactose. Furthermore, the enzyme was able to hydrolyze its own transfucosylation product and 2'-fucosyllactose but only poorly 3-fucosyllactose. As a conclusion, α-fucosidase from A. niger can transfucosylate lactose using free fucose as substrate producing a novel non-reducing 1-fucosyllactose.


Asunto(s)
Aspergillus niger/enzimología , Proteínas Fúngicas/metabolismo , alfa-L-Fucosidasa/metabolismo , Estabilidad de Enzimas , Fucosa/análogos & derivados , Fucosa/metabolismo , Lactosa/análogos & derivados , Lactosa/metabolismo , Especificidad por Sustrato
6.
Appl Microbiol Biotechnol ; 104(24): 10515-10529, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33147349

RESUMEN

In this work, deoxyribose-5-phosphate aldolase (Ec DERA, EC 4.1.2.4) from Escherichia coli was chosen as the protein engineering target for improving the substrate preference towards smaller, non-phosphorylated aldehyde donor substrates, in particular towards acetaldehyde. The initial broad set of mutations was directed to 24 amino acid positions in the active site or in the close vicinity, based on the 3D complex structure of the E. coli DERA wild-type aldolase. The specific activity of the DERA variants containing one to three amino acid mutations was characterised using three different substrates. A novel machine learning (ML) model utilising Gaussian processes and feature learning was applied for the 3rd mutagenesis round to predict new beneficial mutant combinations. This led to the most clear-cut (two- to threefold) improvement in acetaldehyde (C2) addition capability with the concomitant abolishment of the activity towards the natural donor molecule glyceraldehyde-3-phosphate (C3P) as well as the non-phosphorylated equivalent (C3). The Ec DERA variants were also tested on aldol reaction utilising formaldehyde (C1) as the donor. Ec DERA wild-type was shown to be able to carry out this reaction, and furthermore, some of the improved variants on acetaldehyde addition reaction turned out to have also improved activity on formaldehyde. KEY POINTS: • DERA aldolases are promiscuous enzymes. • Synthetic utility of DERA aldolase was improved by protein engineering approaches. • Machine learning methods aid the protein engineering of DERA.


Asunto(s)
Escherichia coli , Fructosa-Bifosfato Aldolasa , Aldehído-Liasas/genética , Aldehído-Liasas/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Fructosa-Bifosfato Aldolasa/genética , Aprendizaje Automático , Ingeniería de Proteínas , Especificidad por Sustrato
7.
Glycoconj J ; 35(1): 31-40, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28905280

RESUMEN

Fucosylated oligosaccharides have an important role in maintaining a healthy immune system and homeostatic gut microflora. This study employed a commercial ß-galactosidase in the production of fucose-containing galacto-oligosaccharides (fGOS) from lactose and fucose. The production was optimized using experiment design and optimal conditions for a batch production in 3-liter scale. The reaction product was analyzed and the produced galactose-fucose disaccharides were purified. The structures of these disaccharides were determined using NMR and it was verified that one major product with the structure Galß1-3Fuc and two minor products with the structures Galß1-4Fuc and Galß1-2Fuc were formed. Additionally, the product composition was defined in more detail using several different analytical methods. It was concluded that the final product contained 42% total monosaccharides, 40% disaccharides and 18% of larger oligosaccharides. 290 µmol of fGOS was produced per gram of reaction mixture and 37% of the added fucose was bound to fGOS. The fraction of fGOS from total oligosaccharides was determined as 44%. This fGOS product could be used as a new putative route to deliver fucose to the intestine.


Asunto(s)
Disacáridos/síntesis química , Fucosa/análogos & derivados , Galactosa/análogos & derivados , beta-Galactosidasa/metabolismo , Disacáridos/química , Glicosilación , Oligosacáridos/química
8.
Biochim Biophys Acta Gen Subj ; 1861(9): 2398-2405, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28591625

RESUMEN

BACKGROUND: The backbone structure of many hemicelluloses is acetylated, which presents a challenge when the objective is to convert corresponding polysaccharides to fermentable sugars or else recover hemicelluloses for biomaterial applications. Carbohydrate esterases (CE) can be harnessed to overcome these challenges. METHODS: Enzymes from different CE families, AnAcXE (CE1), OsAcXE (CE6), and MtAcE (CE16) were compared based on action and position preference towards acetyl-4-O-methylglucuronoxylan (MGX) and acetyl-galactoglucomannan (GGM). To determine corresponding positional preferences, the relative rate of acetyl group released by each enzyme was analyzed by real time 1H NMR. RESULTS: AnAcXE (CE1) showed lowest specific activity towards MGX, where OsAcXE (CE6) and MtAcE were approximately four times more active than AnAcXE (CE1). MtAcE (CE16) was further distinguished by demonstrating 100 times higher activity on GGM compared to AnAcXE (CE1) and OsAcXE (CE6), and five times higher activity on GGM than MGX. Following 24h incubation, all enzymes removed between 78 and 93% of total acetyl content from MGX and GGM, where MtAcE performed best on both substrates. MAJOR CONCLUSIONS: Considering action on MGX, all esterases showed preference for doubly substituted xylopyranosyl residues (2,3-O-acetyl-Xylp). Considering action on GGM, OsAcXE (CE6) preferentially targeted 2-O-acetyl-mannopyranosyl residues (2-O-acetyl-Manp) whereas AnAcXE (CE1) demonstrated highest activity towards 3-O-acetyl-Manp positions; regiopreference of MtAcE (CE16) on GGM was less clear. GENERAL SIGNIFICANCE: The current comparative analysis identifies options to control the position of acetyl group release at initial stages of reaction, and enzyme combinations likely to accelerate deacetylation of major hemicellulose sources.


Asunto(s)
Carbohidratos/química , Esterasas/metabolismo , Mananos/química , Xilanos/química , Acetilación , Polisacáridos/química
9.
Microb Cell Fact ; 15(1): 144, 2016 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-27538689

RESUMEN

BACKGROUND: Pectin-rich wastes, such as citrus pulp and sugar beet pulp, are produced in considerable amounts by the juice and sugar industry and could be used as raw materials for biorefineries. One possible process in such biorefineries is the hydrolysis of these wastes and the subsequent production of ethanol. However, the ethanol-producing organism of choice, Saccharomyces cerevisiae, is not able to catabolize D-galacturonic acid, which represents a considerable amount of the sugars in the hydrolysate, namely, 18 % (w/w) from citrus pulp and 16 % (w/w) sugar beet pulp. RESULTS: In the current work, we describe the construction of a strain of S. cerevisiae in which the five genes of the fungal reductive pathway for D-galacturonic acid catabolism were integrated into the yeast chromosomes: gaaA, gaaC and gaaD from Aspergillus niger and lgd1 from Trichoderma reesei, and the recently described D-galacturonic acid transporter protein, gat1, from Neurospora crassa. This strain metabolized D-galacturonic acid in a medium containing D-fructose as co-substrate. CONCLUSION: This work is the first demonstration of the expression of a functional heterologous pathway for D-galacturonic acid catabolism in Saccharomyces cerevisiae. It is a preliminary step for engineering a yeast strain for the fermentation of pectin-rich substrates to ethanol.


Asunto(s)
Ácidos Hexurónicos/metabolismo , Redes y Vías Metabólicas/genética , Pectinas/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Aspergillus niger/genética , Beta vulgaris , Citrus , Etanol/metabolismo , Fermentación , Fructosa/metabolismo , Hidrólisis , Ingeniería Metabólica/métodos , Neurospora crassa/genética , Trichoderma/genética
10.
Appl Microbiol Biotechnol ; 100(2): 673-85, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26428243

RESUMEN

We describe here the characterization of a novel enzyme called aldose-aldose oxidoreductase (Cc AAOR; EC 1.1.99) from Caulobacter crescentus. The Cc AAOR exists in solution as a dimer, belongs to the Gfo/Idh/MocA family and shows homology with the glucose-fructose oxidoreductase from Zymomonas mobilis. However, unlike other known members of this protein family, Cc AAOR is specific for aldose sugars and can be in the same catalytic cycle both oxidise and reduce a panel of monosaccharides at the C1 position, producing in each case the corresponding aldonolactone and alditol, respectively. Cc AAOR contains a tightly-bound nicotinamide cofactor, which is regenerated in this oxidation-reduction cycle. The highest oxidation activity was detected on D-glucose but significant activity was also observed on D-xylose, L-arabinose and D-galactose, revealing that both hexose and pentose sugars are accepted as substrates by Cc AAOR. The configuration at the C2 and C3 positions of the saccharides was shown to be especially important for the substrate binding. Interestingly, besides monosaccharides, Cc AAOR can also oxidise a range of 1,4-linked oligosaccharides having aldose unit at the reducing end, such as lactose, malto- and cello-oligosaccharides as well as xylotetraose. (1)H NMR used to monitor the oxidation and reduction reaction simultaneously, demonstrated that although D-glucose has the highest affinity and is also oxidised most efficiently by Cc AAOR, the reduction of D-glucose is clearly not as efficient. For the overall reaction catalysed by Cc AAOR, the L-arabinose, D-xylose and D-galactose were the most potent substrates.


Asunto(s)
Aldehído Reductasa/metabolismo , Caulobacter crescentus/enzimología , Monosacáridos/metabolismo , Biocatálisis , Caulobacter crescentus/metabolismo , Glucosa/metabolismo , Resonancia Magnética Nuclear Biomolecular , Oxidorreductasas/metabolismo , Homología de Secuencia de Aminoácido , Xilosa/metabolismo , Zymomonas/enzimología , Zymomonas/metabolismo
11.
Glycobiology ; 24(6): 494-506, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24637390

RESUMEN

O-Acetylglucuronoxylans (AcGX) in Arabidopsis thaliana carry acetyl residues on the 2-O and/or 3-O positions of the xylopyranosyl (Xylp) units, but the distribution of different O-acetylated Xylp units is partly unclear. We studied a possible correlation of xylan acetylation and the activities of different glycosyltransferases involved in xylan biosynthesis by analyzing the distribution of O-acetyl substituents on AcGX from Arabidopsis wild-type and mutants irx7, irx9-1, irx10, irx14 and gux1gux2. The relative contents of the Xylp structural units were determined with quantitative two-dimensional heteronuclear single quantum coherence nuclear magnetic resonance spectroscopy. In the wild type, the degree of acetylation (DA) was 60%. Mono- and diacetylated Xylp units constituted 44 and 6% of the AcGX backbone, respectively; while (4-O-methyl)-glucopyranosyluronic acid (1 → 2)-linked Xylp units, most of which also carry 3-O-acetylation, represented 13%. The DA was decreased in irx7, irx9-1 and irx14 due to the decrease in monoacetylation (2-O and 3-O), indicating a relationship between acetylation and other AcGX biosynthetic processes. The possible interactions that could lead to such changes have been discussed. No change in DA was observed in irx10 and gux1gux2, but monoacetylation was nonetheless elevated in gux1gux2. This indicates that acetylation occurs after addition of GlcpA to the xylan backbone. Mass fragmentation analysis suggests that the prevalent acetylation pattern is the acetyl group added on every other Xylp unit.


Asunto(s)
Glicosiltransferasas/biosíntesis , Xilanos/biosíntesis , Acetilación , Arabidopsis/enzimología , Arabidopsis/genética , Regulación de la Expresión Génica de las Plantas , Glicosiltransferasas/genética , Glicosiltransferasas/metabolismo , Mutación , Xilanos/química , Xilanos/metabolismo
12.
Metab Eng ; 25: 238-47, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25073011

RESUMEN

D-xylonate is a potential platform chemical which can be produced by engineered Saccharomyces cerevisiae strains. In order to address production constraints in more detail, we analysed the role of lactone ring opening in single cells and populations. Both D-xylono-γ-lactone and D-xylonate were produced when the Caulobacter crescentus xylB (D-xylose dehydrogenase) was expressed in S. cerevisiae, with or without co-expression of xylC (D-xylonolactonase), as seen by (1)H NMR. XylC facilitated rapid opening of the lactone and more D-xylonate was initially produced than in its absence. Using in vivo(1)H NMR analysis of cell extracts, culture media and intact cells we observed that the lactone and linear forms of D-xylonic acid were produced, accumulated intracellularly, and partially exported within 15-60min of D-xylose provision. During single-cell analysis of cells expressing the pH sensitive fluorescent probe pHluorin, pHluorin fluorescence was gradually lost from the cells during D-xylonate production, as expected for cells with decreasing intracellular pH. However, in the presence of D-xylose, only 9% of cells expressing xylB lost pHluorin fluorescence within 4.5h, whereas 99% of cells co-expressing xylB and xylC lost fluorescence, a large proportion of which also lost vitality, during this interval. Loss of vitality in the presence of D-xylose was correlated to the extracellular pH, but fluorescence was lost from xylB and xylC expressing cells regardless of the extracellular condition.


Asunto(s)
Análisis de Flujos Metabólicos/métodos , Modelos Biológicos , Saccharomyces cerevisiae/metabolismo , Azúcares Ácidos/metabolismo , Xilosidasas/metabolismo , Simulación por Computador , Regulación Enzimológica de la Expresión Génica/fisiología , Regulación Fúngica de la Expresión Génica/fisiología , Transducción de Señal/fisiología
13.
Appl Microbiol Biotechnol ; 98(23): 9653-65, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25236800

RESUMEN

Four potential dehydrogenases identified through literature and bioinformatic searches were tested for L-arabonate production from L-arabinose in the yeast Saccharomyces cerevisiae. The most efficient enzyme, annotated as a D-galactose 1-dehydrogenase from the pea root nodule bacterium Rhizobium leguminosarum bv. trifolii, was purified from S. cerevisiae as a homodimeric protein and characterised. We named the enzyme as a L-arabinose/D-galactose 1-dehydrogenase (EC 1.1.1.-), Rl AraDH. It belongs to the Gfo/Idh/MocA protein family, prefers NADP(+) but uses also NAD(+) as a cofactor, and showed highest catalytic efficiency (k cat/K m) towards L-arabinose, D-galactose and D-fucose. Based on nuclear magnetic resonance (NMR) and modelling studies, the enzyme prefers the α-pyranose form of L-arabinose, and the stable oxidation product detected is L-arabino-1,4-lactone which can, however, open slowly at neutral pH to a linear L-arabonate form. The pH optimum for the enzyme was pH 9, but use of a yeast-in-vivo-like buffer at pH 6.8 indicated that good catalytic efficiency could still be expected in vivo. Expression of the Rl AraDH dehydrogenase in S. cerevisiae, together with the galactose permease Gal2 for L-arabinose uptake, resulted in production of 18 g of L-arabonate per litre, at a rate of 248 mg of L-arabonate per litre per hour, with 86 % of the provided L-arabinose converted to L-arabonate. Expression of a lactonase-encoding gene from Caulobacter crescentus was not necessary for L-arabonate production in yeast.


Asunto(s)
Arabinosa/metabolismo , Galactosa Deshidrogenasas/metabolismo , Rhizobium leguminosarum/enzimología , Saccharomyces cerevisiae/metabolismo , Azúcares Ácidos/metabolismo , Clonación Molecular , Coenzimas/metabolismo , Estabilidad de Enzimas , Galactosa Deshidrogenasas/química , Galactosa Deshidrogenasas/genética , Galactosa Deshidrogenasas/aislamiento & purificación , Expresión Génica , Concentración de Iones de Hidrógeno , Cinética , Datos de Secuencia Molecular , NAD/metabolismo , NADP/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Rhizobium leguminosarum/metabolismo , Saccharomyces cerevisiae/enzimología , Saccharomyces cerevisiae/genética , Análisis de Secuencia de ADN
14.
Proc Natl Acad Sci U S A ; 108(14): 5891-6, 2011 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-21436041

RESUMEN

The phytohormones jasmonates (JAs) constitute an important class of elicitors for many plant secondary metabolic pathways. However, JAs do not act independently but operate in complex networks with crosstalk to several other phytohormonal signaling pathways. Here, crosstalk was detected between the JA and abscisic acid (ABA) signaling pathways in the regulation of tobacco (Nicotiana tabacum) alkaloid biosynthesis. A tobacco gene from the PYR/PYL/RCAR family, NtPYL4, the expression of which is regulated by JAs, was found to encode a functional ABA receptor. NtPYL4 inhibited the type-2C protein phosphatases known to be key negative regulators of ABA signaling in an ABA-dependent manner. Overexpression of NtPYL4 in tobacco hairy roots caused a reprogramming of the cellular metabolism that resulted in a decreased alkaloid accumulation and conferred ABA sensitivity to the production of alkaloids. In contrast, the alkaloid biosynthetic pathway was not responsive to ABA in control tobacco roots. Functional analysis of the Arabidopsis (Arabidopsis thaliana) homologs of NtPYL4, PYL4 and PYL5, indicated that also in Arabidopsis altered PYL expression affected the JA response, both in terms of biomass and anthocyanin production. These findings define a connection between a component of the core ABA signaling pathway and the JA responses and contribute to the understanding of the role of JAs in balancing tradeoffs between growth and defense.


Asunto(s)
Ácido Abscísico/metabolismo , Proteínas de Arabidopsis/metabolismo , Arabidopsis/metabolismo , Ciclopentanos/metabolismo , Nicotiana/metabolismo , Oxilipinas/metabolismo , Reguladores del Crecimiento de las Plantas/metabolismo , Receptores de Superficie Celular/metabolismo , Transducción de Señal/fisiología , Alcaloides/biosíntesis , Análisis del Polimorfismo de Longitud de Fragmentos Amplificados , Perfilación de la Expresión Génica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
J Biol Chem ; 287(21): 17662-17671, 2012 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-22493433

RESUMEN

Microorganisms use different pathways for D-galacturonate catabolism. In the known microbial oxidative pathway, D-galacturonate is oxidized to D-galactarolactone, the lactone hydrolyzed to galactarate, which is further converted to 3-deoxy-2-keto-hexarate and α-ketoglutarate. We have shown recently that Agrobacterium tumefaciens strain C58 contains an uronate dehydrogenase (At Udh) that oxidizes D-galacturonic acid to D-galactarolactone. Here we report identification of a novel enzyme from the same A. tumefaciens strain, which we named Galactarolactone cycloisomerase (At Gci) (E.C. 5.5.1.-), for the direct conversion of the D-galactarolactone to 3-deoxy-2-keto-hexarate. The At Gci enzyme is 378 amino acids long and belongs to the mandelate racemase subgroup in the enolase superfamily. At Gci was heterologously expressed in Escherichia coli, and the purified enzyme was found to exist as an octameric form. It is active both on D-galactarolactone and D-glucarolactone, but does not work on the corresponding linear hexaric acid forms. The details of the reaction mechanism were further studied by NMR and optical rotation demonstrating that the reaction product of At Gci from D-galactaro-1,4-lactone and D-glucaro-1,4-lactone conversion is in both cases the L-threo form of 3-deoxy-2-keto-hexarate.


Asunto(s)
Agrobacterium tumefaciens/enzimología , Proteínas Bacterianas/metabolismo , Liasas Intramoleculares/metabolismo , Lactonas/metabolismo , Agrobacterium tumefaciens/genética , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Liasas Intramoleculares/genética , Datos de Secuencia Molecular
16.
Fungal Genet Biol ; 49(2): 152-9, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22155165

RESUMEN

For the catabolism of D-galactose three different metabolic pathways have been described in filamentous fungi. Apart from the Leloir pathway and the oxidative pathway, there is an alternative oxido-reductive pathway. This oxido-reductive pathway has similarities to the metabolic pathway of L-arabinose, and in Trichoderma reesei (Hypocrea jecorina) and Aspergillus nidulans the same enzyme is employed for the oxidation of L-arabitol and galactitol. Here we show evidence that in Aspergillus niger L-arabitol dehydrogenase (LadA) is not involved in the D-galactose metabolism; instead another dehydrogenase encoding gene, ladB, is induced in response to D-galactose and galactitol and functions as a galactitol dehydrogenase. Deletion of ladB in A. niger results in growth arrest on galactitol and significantly slower growth on D-galactose supplemented with a small amount of D-xylose. D-galactose alone cannot be utilised by A. niger and the addition of D-xylose stimulates growth on D-galactose via transcriptional activation of the D-xylose-inducible reductase gene, xyrA. XyrA catalyses the first step of the D-galactose oxido-reductive pathway, the reduction to galactitol, which in turn seems to be an inducer of the downstream genes such as LadB. The deletion of xyrA results in reduced growth on D-galactose. The ladB gene was expressed in the heterologous host Saccharomyces cerevisiae and the tagged and purified enzyme characterised. LadB and LadA have similar in vitro activity with galactitol. It was confirmed that the reaction product of the LadB reaction from galactitol is L-xylo-3-hexulose as in the case of the T. reesei Lad1.


Asunto(s)
Aspergillus niger/enzimología , Galactosa/metabolismo , Deshidrogenasas del Alcohol de Azúcar/aislamiento & purificación , Deshidrogenasas del Alcohol de Azúcar/metabolismo , Oxidorreductasas de Alcohol/genética , Oxidorreductasas de Alcohol/metabolismo , Aspergillus niger/genética , Aspergillus niger/metabolismo , Galactitol/metabolismo , Regulación Fúngica de la Expresión Génica/efectos de los fármacos , Hexosas/metabolismo , Cetosas/metabolismo , Redes y Vías Metabólicas , Metabolismo , Oxidación-Reducción , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Deshidrogenasas del Alcohol de Azúcar/genética , Xilosa/farmacología
17.
Metab Eng ; 14(4): 427-36, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22709678

RESUMEN

An NAD(+)-dependent D-xylose dehydrogenase, XylB, from Caulobacter crescentus was expressed in Saccharomyces cerevisiae, resulting in production of 17 ± 2 g D-xylonate l(-1) at 0.23 gl(-1)h(-1) from 23 g D-xylose l(-1) (with glucose and ethanol as co-substrates). D-Xylonate titre and production rate were increased and xylitol production decreased, compared to strains expressing genes encoding T. reesei or pig liver NADP(+)-dependent D-xylose dehydrogenases. D-Xylonate accumulated intracellularly to ∼70 mgg(-1); xylitol to ∼18 mgg(-1). The aldose reductase encoding gene GRE3 was deleted to reduce xylitol production. Cells expressing D-xylonolactone lactonase xylC from C. crescentus with xylB initially produced more extracellular D-xylonate than cells lacking xylC at both pH 5.5 and pH 3, and sustained higher production at pH 3. Cell vitality and viability decreased during D-xylonate production at pH 3.0. An industrial S. cerevisiae strain expressing xylB efficiently produced 43 g D-xylonate l(-1) from 49 g D-xylose l(-1).


Asunto(s)
Ingeniería Metabólica/métodos , Saccharomyces cerevisiae/metabolismo , Ácidos Urónicos/metabolismo , Xilosa/metabolismo , Oxidorreductasas de Alcohol/biosíntesis , Oxidorreductasas de Alcohol/genética , Aldehído Reductasa/genética , Aldehído Reductasa/metabolismo , Animales , Caulobacter crescentus/enzimología , Caulobacter crescentus/genética , Etanol/metabolismo , Glucosa/metabolismo , Hígado/enzimología , Saccharomyces cerevisiae/genética , Porcinos/metabolismo
18.
Microb Cell Fact ; 11: 57, 2012 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-22569166

RESUMEN

BACKGROUND: The methylotrophic yeast Pichia pastoris has emerged as one of the most promising yeast hosts for the production of heterologous proteins. Mixed feeds of methanol and a multicarbon source instead of methanol as sole carbon source have been shown to improve product productivities and alleviate metabolic burden derived from protein production. Nevertheless, systematic quantitative studies on the relationships between the central metabolism and recombinant protein production in P. pastoris are still rather limited, particularly when growing this yeast on mixed carbon sources, thus hampering future metabolic network engineering strategies for improved protein production. RESULTS: The metabolic flux distribution in the central metabolism of P. pastoris growing on a mixed feed of glucose and methanol was analyzed by Metabolic Flux Analysis (MFA) using 13C-NMR-derived constraints. For this purpose, we defined new flux ratios for methanol assimilation pathways in P. pastoris cells growing on glucose:methanol mixtures. By using this experimental approach, the metabolic burden caused by the overexpression and secretion of a Rhizopus oryzae lipase (Rol) in P. pastoris was further analyzed. This protein has been previously shown to trigger the unfolded protein response in P. pastoris. A series of 13C-tracer experiments were performed on aerobic chemostat cultivations with a control and two different Rol producing strains growing at a dilution rate of 0.09 h(-1) using a glucose:methanol 80:20 (w/w) mix as carbon source.The MFA performed in this study reveals a significant redistribution of carbon fluxes in the central carbon metabolism when comparing the two recombinant strains vs the control strain, reflected in increased glycolytic, TCA cycle and NADH regeneration fluxes, as well as higher methanol dissimilation rates. CONCLUSIONS: Overall, a further 13C-based MFA development to characterise the central metabolism of methylotrophic yeasts when growing on mixed methanol:multicarbon sources has been implemented, thus providing a new tool for the investigation of the relationships between central metabolism and protein production. Specifically, the study points at a limited but significant impact of the conformational stress associated to secretion of recombinant proteins on the central metabolism, occurring even at modest production levels.


Asunto(s)
Proteínas Fúngicas/metabolismo , Glucosa/metabolismo , Lipasa/metabolismo , Metanol/metabolismo , Pichia/metabolismo , Rhizopus/enzimología , Proteínas Fúngicas/genética , Lipasa/genética , Metaboloma , Pichia/genética , Pichia/crecimiento & desarrollo , Transporte de Proteínas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Rhizopus/genética , Vías Secretoras
19.
Biomacromolecules ; 12(2): 409-18, 2011 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-21207960

RESUMEN

Weissella confusa VTT E-90392 is an efficient producer of a dextran that is mainly composed of α-(1→6)-linked D-glucosyl units and very few α-(1→3) branch linkages. A mixture of the Chaetomium erraticum endodextranase and the Aspergillus niger α-glucosidase was used to hydrolyze W. confusa dextran to glucose and a set of enzyme-resistant isomaltooligosaccharides. Two of the oligosaccharides (tetra- and hexasaccharide) were isolated in pure form and their structures elucidated. The tetrasaccharide had a nonreducing end terminal α-(1→3)-linked glucosyl unit (α-D-Glcp-(1→3)-α-D-Glcp-(1→6)-α-D-Glcp-(1→6)-α-D-Glc), whereas the hexasaccharide had an α-(1→3)-linked isomaltosyl side group (α-D-Glcp-(1→6)[α-D-Glcp-(1→6)-α-D-Glcp-(1→3)]-α-D-Glcp-(1→6)-α-D-Glcp-(1→6)-α-D-Glc). A mixture of two isomeric oligosaccharides was also obtained in the pentasaccharide fraction, which were identified as (α-D-Glcp-(1→6)-α-D-Glcp-(1→3)-α-D-Glcp-(1→6)-α-D-Glcp-(1→6)-α-D-Glc) and (α-D-Glcp-(1→6)[α-D-Glcp-(1→3)]-α-D-Glcp-(1→6)-α-D-Glcp-(1→6)-α-D-Glc). The structures of the oligosaccharides indicated that W. confusa dextran contains both terminal and elongated α-(1→3)-branches. This is the first report evidencing the presence of elongated branches in W. confusa dextran. The (1)H and (13)C NMR spectroscopic data on the enzyme-resistant isomaltooligosaccharides with α-(1→3)-linked glucosyl and isomaltosyl groups are published here for the first time.


Asunto(s)
Dextranos/química , Leuconostocaceae/química , Oligosacáridos/química , Aspergillus niger/enzimología , Conformación de Carbohidratos , Chaetomium/enzimología , Dextranasa/antagonistas & inhibidores , Dextranasa/metabolismo , Inhibidores de Glicósido Hidrolasas , Hidrólisis , Oligosacáridos/farmacología , alfa-Glucosidasas/metabolismo
20.
Appl Microbiol Biotechnol ; 86(3): 901-9, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19921179

RESUMEN

There are at least three different pathways for the catabolism of D-galacturonate in microorganisms. In the oxidative pathway, which was described in some prokaryotic species, D-galacturonate is first oxidised to meso-galactarate (mucate) by a nicotinamide adenine dinucleotide (NAD)-dependent dehydrogenase (EC 1.1.1.203). In the following steps of the pathway mucate is converted to 2-keto-glutarate. The enzyme activities of this catabolic pathway have been described while the corresponding gene sequences are still unidentified. The D-galacturonate dehydrogenase was purified from Agrobacterium tumefaciens, and the mass of its tryptic peptides was determined using MALDI-TOF mass spectrometry. This enabled the identification of the corresponding gene udh. It codes for a protein with 267 amino acids having homology to the protein family of NAD(P)-binding Rossmann-fold proteins. The open reading frame was functionally expressed in Saccharomyces cerevisiae. The N-terminally tagged protein was not compromised in its activity and was used after purification for a kinetic characterization. The enzyme was specific for NAD and accepted D-galacturonic acid and D-glucuronic acid as substrates with similar affinities. NMR analysis showed that in water solution the substrate D-galacturonic acid is predominantly in pyranosic form which is converted by the enzyme to 1,4 lactone of galactaric acid. This lactone seems stable under intracellular conditions and does not spontaneously open to the linear meso-galactaric acid.


Asunto(s)
Agrobacterium tumefaciens/enzimología , Aldehído Oxidorreductasas/genética , Aldehído Oxidorreductasas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Ácidos Hexurónicos/metabolismo , Aldehído Oxidorreductasas/química , Aldehído Oxidorreductasas/aislamiento & purificación , Proteínas Bacterianas/química , Proteínas Bacterianas/aislamiento & purificación , Clonación Molecular , Expresión Génica , Ácido Glucurónico/metabolismo , Cinética , Espectroscopía de Resonancia Magnética , Redes y Vías Metabólicas , Modelos Biológicos , Peso Molecular , NAD/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , Saccharomyces cerevisiae/genética , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Especificidad por Sustrato , Azúcares Ácidos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA