Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
PLoS Genet ; 15(5): e1008149, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31067226

RESUMEN

Tomato (Solanum lycopersicum) is one of the highest-value vegetable crops worldwide. Understanding the genetic regulation of primary metabolite levels can inform efforts aimed toward improving the nutrition of commercial tomato cultivars, while maintaining key traits such as yield and stress tolerance. We identified 388 suggestive association loci (including 126 significant loci) for 92 metabolic traits including nutrition and flavor-related loci by genome-wide association study from 302 accessions in two different environments. Among them, an ascorbate quantitative trait locus TFA9 (TOMATO FRUIT ASCORBATEON CHROMOSOME 9) co-localized with SlbHLH59, which promotes high ascorbate accumulation by directly binding to the promoter of structural genes involved in the D-mannose/L-galactose pathway. The causal mutation of TFA9 is an 8-bp InDel, named InDel_8, located in the promoter region of SlbHLH59 and spanned a 5'UTR Py-rich stretch motif affecting its expression. Phylogenetic analysis revealed that differentially expressed SlbHLH59 alleles were selected during tomato domestication. Our results provide a dramatic illustration of how ascorbate biosynthesis can be regulated and was selected during the domestication of tomato. Furthermore, the findings provide novel genetic insights into natural variation of metabolites in tomato fruit, and will promote efficient utilization of metabolite traits in tomato improvement.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Solanum lycopersicum/genética , Solanum lycopersicum/metabolismo , Alelos , Ácido Ascórbico/genética , Ácido Ascórbico/metabolismo , Mapeo Cromosómico/métodos , Frutas/genética , Galactosa/biosíntesis , Galactosa/metabolismo , Regulación de la Expresión Génica de las Plantas/genética , Variación Genética/genética , Genoma de Planta/genética , Estudio de Asociación del Genoma Completo , Manosa/biosíntesis , Manosa/metabolismo , Filogenia , Regiones Promotoras Genéticas/genética , Sitios de Carácter Cuantitativo/genética
2.
Metab Eng ; 61: 215-224, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32623008

RESUMEN

Starch/cellulose has become the major feedstock for manufacturing biofuels and biochemicals because of their abundance and sustainability. In this study, we presented an artificially designed "starch-mannose-fermentation" biotransformation process through coupling the advantages of in vivo and in vitro metabolic engineering strategies together. Starch was initially converted into mannose via an in vitro metabolic engineering biosystem, and then mannose was fermented by engineered microorganisms for biomanufacturing valuable mannosyl compounds. The in vitro metabolic engineering biosystem based on phosphorylation/dephosphorylation reactions was thermodynamically favorable and the conversion rate reached 81%. The mannose production using whole-cell biocatalysts reached 75.4 g/L in a 30-L reactor, indicating the potential industrial application. Furthermore, the produced mannose in the reactor was directly served as feedstock for the fermentation process to bottom-up produced 19.2 g/L mannosyl-oligosaccharides (MOS) and 7.2 g/L mannosylglycerate (MG) using recombinant Corynebacterium glutamicum strains. Notably, such a mannose fermentation process facilitated the synthesis of MOS, which has not been achieved under glucose fermentation and improved MG production by 2.6-fold than that using the same C-mole of glucose. This approach also allowed access to produce other kinds of mannosyl derivatives from starch.


Asunto(s)
Reactores Biológicos , Corynebacterium glutamicum , Ácidos Glicéricos , Manosa/análogos & derivados , Ingeniería Metabólica , Microorganismos Modificados Genéticamente , Almidón/metabolismo , Corynebacterium glutamicum/genética , Corynebacterium glutamicum/crecimiento & desarrollo , Manosa/biosíntesis , Manosa/genética , Microorganismos Modificados Genéticamente/genética , Microorganismos Modificados Genéticamente/crecimiento & desarrollo
3.
Microb Cell Fact ; 17(1): 178, 2018 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-30445960

RESUMEN

BACKGROUND: Mannosylglycerate (MG) is one of the most widespread compatible solutes among marine microorganisms adapted to hot environments. This ionic solute holds excellent ability to protect proteins against thermal denaturation, hence a large number of biotechnological and clinical applications have been put forward. However, the current prohibitive production costs impose severe constraints towards large-scale applications. All known microbial producers synthesize MG from GDP-mannose and 3-phosphoglycerate via a two-step pathway in which mannosyl-3-phosphoglycerate is the intermediate metabolite. In an early work, this pathway was expressed in Saccharomyces cerevisiae with the goal to confirm gene function (Empadinhas et al. in J Bacteriol 186:4075-4084, 2004), but the level of MG accumulation was low. Therefore, in view of the potential biotechnological value of this compound, we decided to invest further effort to convert S. cerevisiae into an efficient cell factory for MG production. RESULTS: To drive MG production, the pathway for the synthesis of GDP-mannose, one of the MG biosynthetic precursors, was overexpressed in S. cerevisiae along with the MG biosynthetic pathway. MG production was evaluated under different cultivation modes, i.e., flask bottle, batch, and continuous mode with different dilution rates. The genes encoding mannose-6-phosphate isomerase (PMI40) and GDP-mannose pyrophosphorylase (PSA1) were introduced into strain MG01, hosting a plasmid encoding the MG biosynthetic machinery. The resulting engineered strain (MG02) showed around a twofold increase in the activity of PMI40 and PSA1 in comparison to the wild-type. In batch mode, strain MG02 accumulated 15.86 mgMG g DCW -1 , representing a 2.2-fold increase relative to the reference strain (MG01). In continuous culture, at a dilution rate of 0.15 h-1, there was a 1.5-fold improvement in productivity. CONCLUSION: In the present study, the yield and productivity of MG were increased by overexpression of the GDP-mannose pathway and optimization of the mode of cultivation. A maximum of 15.86 mgMG g DCW -1 was achieved in batch cultivation and maximal productivity of 1.79 mgMG g DCW -1  h-1 in continuous mode. Additionally, a positive correlation between MG productivity and growth rate/dilution rate was established, although this correlation is not observed for MG yield.


Asunto(s)
Biotecnología/métodos , Manosa/análogos & derivados , Ingeniería Metabólica/métodos , Saccharomyces cerevisiae/metabolismo , Técnicas de Cultivo Celular por Lotes , Biomasa , Reactores Biológicos/microbiología , Regulación Fúngica de la Expresión Génica , Ácidos Glicéricos/química , Manosa/biosíntesis , Manosa/química , Saccharomyces cerevisiae/enzimología , Saccharomyces cerevisiae/genética
4.
Angew Chem Int Ed Engl ; 57(30): 9268-9273, 2018 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-29732660

RESUMEN

O Mannosylation is a vital protein modification involved in brain and muscle development whereas the biological relevance of O-mannosyl glycans has remained largely unknown owing to the lack of structurally defined glycoforms. An efficient scaffold synthesis/enzymatic extension (SSEE) strategy was developed to prepare such structures by combining gram-scale convergent chemical syntheses of three scaffolds and strictly controlled sequential enzymatic extension catalyzed by glycosyltransferases. In total, 45 O-mannosyl glycans were obtained, covering the majority of identified mammalian structures. Subsequent glycan microarray analysis revealed fine specificities of glycan-binding proteins and specific antisera.


Asunto(s)
Glicosiltransferasas/metabolismo , Manosa/biosíntesis , Polisacáridos/biosíntesis , Conformación de Carbohidratos , Manosa/química , Polisacáridos/química
5.
Appl Microbiol Biotechnol ; 100(7): 2985-92, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26875877

RESUMEN

L-Rhamnose isomerase (L-RI, EC 5.3.1.14), catalyzing the isomerization between L-rhamnose and L-rhamnulose, plays an important role in microbial L-rhamnose metabolism and thus occurs in a wide range of microorganisms. It attracts more and more attention because of its broad substrate specificity and its great potential in enzymatic production of various rare sugars. In this article, the enzymatic properties of various reported L-RIs were compared in detail, and their applications in the production of L-rhamnulose and various rare sugars including D-allose, D-gulose, L-lyxose, L-mannose, L-talose, and L-galactose were also reviewed.


Asunto(s)
Isomerasas Aldosa-Cetosa/metabolismo , Bacillus subtilis/metabolismo , Proteínas Bacterianas/metabolismo , Escherichia coli/metabolismo , Ramnosa/metabolismo , Thermotoga maritima/metabolismo , Isomerasas Aldosa-Cetosa/genética , Bacillus subtilis/genética , Proteínas Bacterianas/genética , Biotecnología , Escherichia coli/genética , Galactosa/biosíntesis , Expresión Génica , Glucosa/biosíntesis , Hexosas/biosíntesis , Lactonas/metabolismo , Manosa/biosíntesis , Pentosas/biosíntesis , Estereoisomerismo , Thermotoga maritima/genética
6.
Appl Environ Microbiol ; 80(14): 4226-33, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24795373

RESUMEN

Marine hyperthermophiles accumulate small organic compounds, known as compatible solutes, in response to supraoptimal temperatures or salinities. Pyrococcus furiosus is a hyperthermophilic archaeon that grows optimally at temperatures near 100°C. This organism accumulates mannosylglycerate (MG) and di-myo-inositol phosphate (DIP) in response to osmotic and heat stress, respectively. It has been assumed that MG and DIP are involved in cell protection; however, firm evidence for the roles of these solutes in stress adaptation is still missing, largely due to the lack of genetic tools to produce suitable mutants of hyperthermophiles. Recently, such tools were developed for P. furiosus, making this organism an ideal target for that purpose. In this work, genes coding for the synthases in the biosynthetic pathways of MG and DIP were deleted by double-crossover homologous recombination. The growth profiles and solute patterns of the two mutants and the parent strain were investigated under optimal growth conditions and also at supraoptimal temperatures and NaCl concentrations. DIP was a suitable replacement for MG during heat stress, but substitution of MG for DIP and aspartate led to less efficient growth under conditions of osmotic stress. The results suggest that the cascade of molecular events leading to MG synthesis is tuned for osmotic adjustment, while the machinery for induction of DIP synthesis responds to either stress agent. MG protects cells against heat as effectively as DIP, despite the finding that the amount of DIP consistently increases in response to heat stress in the nine (hyper)thermophiles examined thus far.


Asunto(s)
Adaptación Fisiológica , Fosfatos de Inositol/biosíntesis , Manosa/análogos & derivados , Pyrococcus furiosus/fisiología , Estrés Fisiológico , Medios de Cultivo , Eliminación de Gen , Ácidos Glicéricos , Calor , Manosa/biosíntesis , Presión Osmótica , Pyrococcus furiosus/genética , Cloruro de Sodio/metabolismo
7.
Extremophiles ; 18(5): 835-52, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25108362

RESUMEN

Halophilic and halotolerant microorganisms adapted to thrive in hot environments accumulate compatible solutes that usually have a negative charge either associated with a carboxylic group or a phosphodiester unit. Mannosylglycerate (MG) has been detected in several members of (hyper)thermophilic bacteria and archaea, in which it responds primarily to osmotic stress. The outstanding ability of MG to stabilize protein structure in vitro as well as in vivo has been convincingly demonstrated. These findings led to an increasingly supported link between MG and microbial adaptation to high temperature. However, the accumulation of MG in many red algae has been known for a long time, and the peculiar distribution of MG in such distant lineages was intriguing. Knowledge on the biosynthetic machinery together with the rapid expansion of genome databases allowed for structural and phylogenetic analyses and provided insight into the distribution of MG. The two pathways for MG synthesis have distinct evolutionary histories and physiological roles: in red algae MG is synthesised exclusively via the single-step pathway and most probably is unrelated with stress protection. In contrast, the two-step pathway is strongly associated with osmoadaptation in (hyper)thermophilic prokaryotes. The phylogenetic analysis of the two-step pathway also reveals a second cluster composed of fungi and mesophilic bacteria, but MG has not been demonstrated in members of this cluster; we propose that the synthase is part of a more complex pathway directed at the synthesis of yet unknown molecules containing the mannosyl-glyceryl unit.


Asunto(s)
Archaea/genética , Bacterias/genética , Evolución Molecular , Manosa/análogos & derivados , Adaptación Fisiológica , Secuencia de Aminoácidos , Archaea/metabolismo , Bacterias/metabolismo , Ácidos Glicéricos , Manosa/biosíntesis , Manosa/genética , Datos de Secuencia Molecular
8.
Glycobiology ; 23(10): 1192-203, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23893788

RESUMEN

The methylotrophic yeast, Pichia pastoris, is an important organism used for the production of therapeutic proteins. Previously, we have reported the glycoengineering of this organism to produce human-like N-linked glycans but up to now no one has addressed engineering the O-linked glycosylation pathway. Typically, O-linked glycans produced by wild-type P. pastoris are linear chains of four to five α-linked mannose residues, which may be capped with ß- or phospho-mannose. Previous genetic engineering of the N-linked glycosylation pathway of P. pastoris has eliminated both of these two latter modifications, resulting in O-linked glycans which are linear α-linked mannose structures. Here, we describe a method for the co-expression of an α-1,2-mannosidase, which reduces these glycans to primarily a single O-linked mannose residue. In doing so, we have reduced the potential of these glycans to interact with carbohydrate-binding proteins, such as dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin. Furthermore, the introduction of the enzyme protein-O-linked-mannose ß-1,2-N-acetylglucosaminyltransferase 1, resulted in the capping of the single O-linked mannose residues with N-acetylglucosamine. Subsequently, this glycoform was extended into human-like sialylated glycans, similar in structure to α-dystroglycan-type glycoforms. As such, this represents the first example of sialylated O-linked glycans being produced in yeast and extends the utility of the P. pastoris production platform beyond N-linked glycosylated biotherapeutics to include molecules possessing O-linked glycans.


Asunto(s)
Manosa/biosíntesis , Ingeniería Metabólica/métodos , Pichia/metabolismo , alfa-Manosidasa/metabolismo , Pichia/crecimiento & desarrollo , Ingeniería de Proteínas , alfa-Manosidasa/genética
9.
Planta ; 237(3): 891-901, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23179444

RESUMEN

A mannosylglycerate synthase (MgS) gene detected in the genome of Selaginella moellendorffii was expressed in E. coli and the recombinant enzyme was purified and characterized. A remarkable and unprecedented feature of this enzyme was the ability to efficiently synthesize mannosylglycerate (MG) and glucosylglycerate (GG) alike, with maximal activity at 50 °C, pH 8.0 and with Mg(2+) as reaction enhancer. We have also identified a novel glycoside hydrolase gene in this plant's genome, which was functionally confirmed to be highly specific for the hydrolysis of MG and GG and named MG hydrolase (MgH), due to its homology with bacterial MgHs. The recombinant enzyme was maximally active at 40 °C and at pH 6.0-6.5. The activity was independent of cations, but Mn(2+) was a strong stimulator. Regardless of these efficient enzymatic resources we could not detect MG or GG in S. moellendorffii or in the extracts of five additional Selaginella species. Herein, we describe the properties of the first eukaryotic enzymes for the synthesis and hydrolysis of the compatible solutes, MG and GG.


Asunto(s)
Ácidos Glicéricos/metabolismo , Manosa/análogos & derivados , Selaginellaceae/enzimología , Genes de Plantas , Concentración de Iones de Hidrógeno , Hidrólisis , Cinética , Espectroscopía de Resonancia Magnética , Manosa/biosíntesis , Manosiltransferasas/genética , Proteínas Recombinantes/metabolismo , Selaginellaceae/genética , Análisis de Secuencia de Proteína , Especificidad de la Especie , Temperatura
10.
Int J Cancer ; 131(1): 117-28, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21823122

RESUMEN

Altered glycosylation in epithelial cancers may play an important role in tumour progression, as it may affect tumour cell migration and antigen presentation by antigen presenting cells. We specifically characterise the glycosylation patterns of two tumour antigens that are highly expressed in cancer tissue and often detected in their secreted form in serum: the epithelial mucin MUC1 and carcinoembryonic antigen (CEA, also called CEACAM5). We analysed 48 colorectal cancer patients, comparing normal colon and tumour epithelium within each patient. Lectin binding was studied by a standardised CEA/MUC1 capture ELISA, using several plant lectins, and the human C-type lectins MGL and DC-SIGN, and Galectin-3. Peanut agglutinin (PNA) bound to MUC1 from tumour tissue in particular, suggests increased expression of the Thomsen-Friedenreich antigen (TF-antigen) (Core 1, Galß1-3GalNAc-Ser/Thr). Only small amounts of Tn-antigen (GalNAcα-Ser/Thr) expression was observed, but the human C-type lectin MGL showed increased binding to tumour-associated MUC1. Furthermore, sialylation was greatly enhanced. In sharp contrast, tumour-associated CEA (CEACAM5) contained high levels of the blood-group related carbohydrates, Lewis X and Lewis Y. This correlated strongly with the interaction of the human C-type lectin DC-SIGN to tumour-associated CEA, suggesting that CEA can be recognized and taken up by antigen presenting cells. In addition, increased mannose expression was observed and branched N-glycans were prominent, and this correlated well with human Galectin-3 binding. These data demonstrate that individual tumour antigens contain distinct glycan structures associated with cancer and, since glycans affect cellular interactions with its microenvironment, this may have consequences for progression of the disease.


Asunto(s)
Antígeno Carcinoembrionario/metabolismo , Colon/metabolismo , Neoplasias del Colon/metabolismo , Mucosa Gástrica/metabolismo , Mucosa Intestinal/metabolismo , Mucina-1/metabolismo , Antígenos de Carbohidratos Asociados a Tumores/biosíntesis , Antígenos de Carbohidratos Asociados a Tumores/metabolismo , Moléculas de Adhesión Celular/metabolismo , Neoplasias del Colon/patología , Proteínas Ligadas a GPI/metabolismo , Galectina 3/metabolismo , Glicosilación , Humanos , Lectinas Tipo C/metabolismo , Antígenos del Grupo Sanguíneo de Lewis/biosíntesis , Antígeno Lewis X/biosíntesis , Manosa/biosíntesis , Aglutinina de Mani/metabolismo , Receptores de Superficie Celular/metabolismo
11.
Biochemistry ; 50(44): 9551-67, 2011 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-21961705

RESUMEN

Mannosyl-3-phosphoglycerate phosphatase (MpgP) is a key mediator in the physiological response to thermal and osmotic stresses, catalyzing the hydrolysis of mannosyl-3-phosphoglycerate (MPG) to the final product, α-mannosylglycerate. MpgP is a metal-dependent haloalcanoic acid dehalogenase-like (HAD-like) phosphatase, preserving the catalytic motifs I-IV of the HAD core domain, and classified as a Cof-type MPGP (HAD-IIB-MPGP family; SCOP [117505]) on the basis of its C2B cap insertion module. Herein, the crystallographic structures of Thermus thermophilus HB27 MpgP in its apo form and in complex with substrates, substrate analogues, and inhibitors are reported. Two distinct enzyme conformations, open and closed, are catalytically relevant. Apo-MpgP is primarily found in the open state, while holo-MpgP, in complex with the reaction products, is found in the closed state. Enzyme activation entails a structural rearrangement of motifs I and IV with concomitant binding of the cocatalytic Mg(2+) ion. The closure motion of the C2B domain is subsequently triggered by the anchoring of the phosphoryl group to the cocatalytic metal center, and by Arg167 fixing the mannosyl moiety inside the catalytic pocket. The results led to the proposal that in T. thermophilus HB27 MpgP the phosphoryl transfer employs a concerted D(N)S(N) mechanism with assistance of proton transfer from the general acid Asp8, forming a short-lived PO(3)(-) intermediate that is attacked by a nucleophilic water molecule. These results provide new insights into a possible continuum of phosphoryl transfer mechanisms, ranging between those purely associative and dissociative, as well as a picture of the main mechanistic aspects of phosphoryl monoester transfer catalysis, common to other members of the HAD superfamily.


Asunto(s)
Proteínas Bacterianas/química , Hidrolasas/química , Familia de Multigenes , Fosfotransferasas (Aceptor de Grupo Alcohol)/química , Thermus thermophilus/enzimología , Catálisis , Cristalografía por Rayos X , Ácidos Glicéricos/química , Manosa/análogos & derivados , Manosa/biosíntesis , Manosa/química , Modelos Moleculares , Multimerización de Proteína
12.
Infect Immun ; 79(11): 4668-73, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21896775

RESUMEN

Mycobacterium tuberculosis contains mannosylated cell wall components which are important in macrophage recognition and response. The building block for the mannosyl constituents of these components is GDP-mannose, which is synthesized through a series of enzymes involved in the mannose donor biosynthesis pathway. Nothing is known about the expression levels of the genes encoding these enzymes during the course of infection. To generate transcriptional profiles for the mannose donor biosynthesis genes from virulent M. tuberculosis and attenuated Mycobacterium bovis BCG, bacteria were grown in broth culture and within human macrophages. Our results with broth-grown bacteria show that there are differences in expression of the selected genes between M. tuberculosis and BCG, with increased expression of manC in M. tuberculosis and manA in BCG during stationary-phase growth. Results for M. tuberculosis extracted from within macrophages show that whiB2 is highly expressed and manB and manC are moderately expressed during infection. Rv3256c, Rv3258c, and ppm1 have high expression levels early and decreased expression as the infection progresses. Results with BCG show that, as in M. tuberculosis, whiB2 is highly expressed throughout infection, whereas there is either low expression or little change in expression of the remaining genes studied. Overall, our results show that there is differential regulation of expression of several genes in the mannose donor biosynthesis pathway of M. tuberculosis and BCG grown in broth and within macrophages, raising the possibility that the level of mannose donors may vary during the course of infection and thereby impact the biosynthesis of mannose-containing cell wall molecules.


Asunto(s)
Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Manosa/biosíntesis , Mycobacterium bovis/metabolismo , Mycobacterium tuberculosis/metabolismo , Transcripción Genética , Proteínas Bacterianas/genética , Perfilación de la Expresión Génica , Humanos , Macrófagos/metabolismo , Macrófagos/microbiología , Manosa/genética , Mycobacterium bovis/genética , Mycobacterium bovis/patogenicidad , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/patogenicidad , Especificidad de la Especie , Factores de Tiempo , Virulencia
13.
Environ Microbiol ; 13(8): 2056-77, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21176052

RESUMEN

A decade ago the compatible solutes mannosylglycerate (MG) and glucosylglycerate (GG) were considered to be rare in nature. Apart from two species of thermophilic bacteria, Thermus thermophilus and Rhodothermus marinus, and a restricted group of hyperthermophilic archaea, the Thermococcales, MG had only been identified in a few red algae. Glucosylglycerate was considered to be even rarer and had only been detected as an insignificant solute in two halophilic microorganisms, a cyanobacterium, as a component of a polysaccharide and of a glycolipid in two actinobacteria. Unlike the hyper/thermophilic MG-accumulating microorganisms, branching close to the root of the Tree of Life, those harbouring GG shared a mesophilic lifestyle. Exceptionally, the thermophilic bacterium Persephonella marina was reported to accumulate GG. However, and especially owing to the identification of the key-genes for MG and GG synthesis and to the escalating numbers of genomes available, a plethora of new organisms with the resources to synthesize these solutes has been recognized. The accumulation of GG as an 'emergency' compatible solute under combined salt stress and nitrogen-deficient conditions now seems to be a disseminated survival strategy from enterobacteria to marine cyanobacteria. In contrast, the thermophilic and extremely radiation-resistant bacterium Rubrobacter xylanophilus is the only actinobacterium known to accumulate MG, and under all growth conditions tested. This review addresses the environmental factors underlying the accumulation of MG, GG and derivatives in bacteria and archaea and their roles during stress adaptation or as precursors for more elaborated macromolecules. The diversity of pathways for MG and GG synthesis as well as those for some of their derivatives is also discussed. The importance of glycerate-derived organic solutes in the microbial world is only now being recognized. Their stress-dependent accumulation and the molecular aspects of their interactions with biomolecules have already fuelled several emerging applications in biotechnology and biomedicine.


Asunto(s)
Archaea/metabolismo , Bacterias/metabolismo , Vías Biosintéticas , Adaptación Fisiológica , Archaea/genética , Bacterias/genética , Vías Biosintéticas/genética , Glucósidos/biosíntesis , Glucósidos/genética , Glucósidos/metabolismo , Ácidos Glicéricos/metabolismo , Manosa/análogos & derivados , Manosa/biosíntesis , Manosa/genética , Manosa/metabolismo
14.
Microbiology (Reading) ; 156(Pt 11): 3492-3502, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20688818

RESUMEN

Lipoarabinomannan (LAM) is a major glycolipid in the mycobacterial cell envelope. LAM consists of a mannosylphosphatidylinositol (MPI) anchor, a mannan core and a branched arabinan domain. The termini of the arabinan branches can become substituted with one to three α(1→2)-linked mannosyl residues, the mannose cap, producing ManLAM. ManLAM has been associated with a range of different immunomodulatory properties of Mycobacterium tuberculosis during infection of the host. In some of these effects, the presence of the mannose cap on ManLAM appears to be crucial for its activity. So far, in the biosynthesis of the mannose cap on ManLAM, two enzymes have been reported to be involved: a mannosyltransferase that adds the first mannosyl residue of the mannose caps to the arabinan domain of LAM, and another mannosyltransferase that elongates the mannose cap up to three mannosyl residues. Here, we report that a third gene is involved, MMAR_2380, which is the Mycobacterium marinum orthologue of Rv1565c. MMAR_2380 encodes a predicted transmembrane acyltransferase. In M. marinum ΔMMAR_2380, the LAM arabinan domain is still intact, but the mutant LAM lacks the mannose cap. Additional effects of mutation of MMAR_2380 on LAM were observed: a higher degree of branching of both the arabinan domain and the mannan core, and a decreased incorporation of [1,2-(14)C]acetate into the acyl chains in mutant LAM as compared with the wild-type form. This latter effect was also observed for related lipoglycans, i.e. lipomannan (LM) and phosphatidylinositol mannosides (PIMs). Furthermore, the mutant strain showed increased aggregation in liquid cultures as compared with the wild-type strain. All phenotypic traits of M. marinum ΔMMAR_2380, the deficiency in the mannose cap on LAM and changes at the cell surface, could be reversed by complementing the mutant strain with MMAR_2380. Strikingly, membrane preparations of the mutant strain still showed enzymic activity for the arabinan mannose-capping mannosyltransferase similar to that of the wild-type strain. Although the exact function of MMAR_2380 remains unknown, we show that the protein is essential for the presence of a mannose cap on LAM.


Asunto(s)
Aciltransferasas/metabolismo , Lipopolisacáridos/biosíntesis , Manosa/biosíntesis , Mycobacterium marinum/enzimología , Acilación , Aciltransferasas/genética , Genes Bacterianos , Prueba de Complementación Genética , Lipopolisacáridos/química , Manosa/química , Manosiltransferasas/metabolismo , Mutación , Mycobacterium marinum/genética
15.
mSphere ; 5(1)2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31941812

RESUMEN

The pathogenic fungus Aspergillus fumigatus contains galactomannans localized on the surface layer of its cell walls, which are involved in various biological processes. Galactomannans comprise α-(1→2)-/α-(1→6)-mannan and ß-(1→5)-/ß-(1→6)-galactofuranosyl chains. We previously revealed that GfsA is a ß-galactofuranoside ß-(1→5)-galactofuranosyltransferase involved in the biosynthesis of ß-(1→5)-galactofuranosyl chains. In this study, we clarified the biosynthesis of ß-(1→5)-galactofuranosyl chains in A. fumigatus Two paralogs exist within A. fumigatus: GfsB and GfsC. We show that GfsB and GfsC, in addition to GfsA, are ß-galactofuranoside ß-(1→5)-galactofuranosyltransferases by biochemical and genetic analyses. GfsA, GfsB, and GfsC can synthesize ß-(1→5)-galactofuranosyl oligomers at up to lengths of 7, 3, and 5 galactofuranoses within an established in vitro highly efficient assay of galactofuranosyltransferase activity. Structural analyses of galactomannans extracted from ΔgfsB, ΔgfsC, ΔgfsAC, and ΔgfsABC strains revealed that GfsA and GfsC synthesized all ß-(1→5)-galactofuranosyl residues of fungal-type and O-mannose-type galactomannans and that GfsB exhibited limited function in A. fumigatus The loss of ß-(1→5)-galactofuranosyl residues decreased the hyphal growth rate and conidium formation ability and increased the abnormal hyphal branching structure and cell surface hydrophobicity, but this loss is dispensable for sensitivity to antifungal agents and virulence toward immunocompromised mice.IMPORTANCE ß-(1→5)-Galactofuranosyl residues are widely distributed in the subphylum Pezizomycotina of the phylum Ascomycota. Pezizomycotina includes many plant and animal pathogens. Although the structure of ß-(1→5)-galactofuranosyl residues of galactomannans in filamentous fungi was discovered long ago, it remains unclear which enzyme is responsible for biosynthesis of this glycan. Fungal cell wall formation processes are complicated, and information concerning glycosyltransferases is essential for understanding them. In this study, we showed that GfsA and GfsC are responsible for the biosynthesis of all ß-(1→5)-galactofuranosyl residues of fungal-type and O-mannose-type galactomannans. The data presented here indicate that ß-(1→5)-galactofuranosyl residues are involved in cell growth, conidiation, polarity, and cell surface hydrophobicity. Our new understanding of ß-(1→5)-galactofuranosyl residue biosynthesis provides important novel insights into the formation of the complex cell wall structure and the virulence of the members of the subphylum Pezizomycotina.


Asunto(s)
Aspergillus fumigatus/enzimología , Mananos/biosíntesis , Mananos/química , Manosa/química , Animales , Aspergillus fumigatus/genética , Pared Celular/química , Pared Celular/metabolismo , Galactosa/análogos & derivados , Glicosiltransferasas/metabolismo , Hifa , Manosa/biosíntesis , Ratones , Virulencia
16.
Enzyme Microb Technol ; 138: 109553, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32527523

RESUMEN

Recently, functional sugars, such as d-mannose, have attracted considerable attention due to their excellent physiological benefits for human health and wide applications in food and pharmaceutical industries. Therefore, d-mannose production using a sugar isomerase such as d-lyxose isomerase (d-LIase) has emerged as a research hotspot owing to its advantages over plant extraction and chemical synthesis methods. In this study, a putative d-LIase gene from Caldanaerobius polysaccharolyticus was cloned and expressed in Escherichia coli. Then, a biochemical characterization of the recombinant d-LIase was carried out and its potential use in d-mannose production also assessed. Results showed that d-LIase exhibited its maximum activity under these optimal conditions: temperature of 65 °C, a pH of 6.5, and the Mn2+ metal ion. The d-LIase was active at pH 6.0-8.0; it was also quite thermostable up to 60 °C and approximately 85 % of its maximum activity was retained after incubating for 4 h. Further, our Nano-DSC analysis determined that its melting temperature (Tm) was 70.74 °C. Using 100, 300, and 500 g L-1 of d-fructose as substrate, 25.6, 74.4, and 115 g L-1 of d-mannose were produced respectively, corresponding to a conversion rate of 25.6 %, 24.8 %, and 23.0 % under optimal conditions. Taken together, our results provide evidence for a promising candidate d-LIase for producing d-mannose directly from d-fructose.


Asunto(s)
Isomerasas Aldosa-Cetosa/metabolismo , Proteínas Bacterianas/metabolismo , Firmicutes/enzimología , Manosa/biosíntesis , Isomerasas Aldosa-Cetosa/química , Isomerasas Aldosa-Cetosa/genética , Isomerasas Aldosa-Cetosa/aislamiento & purificación , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/aislamiento & purificación , Escherichia coli/genética , Escherichia coli/metabolismo , Firmicutes/genética , Fructosa/metabolismo , Calor , Concentración de Iones de Hidrógeno , Cinética , Manganeso , Multimerización de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato
17.
Biol Pharm Bull ; 32(11): 1921-3, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19881309

RESUMEN

We detected the beta-1,2-mannosyltransferases (beta-1,2-MTs), which participate in the biosynthesis of oligomannosyl side chains in the mannan acid-labile fraction, in a particulate insoluble fractions prepared from Candida albicans NIH B-792 strain cells grown at 27 degrees C and at 37 degrees C in a yeast extract-added Sabouraud liquid medium (YSLM). The beta-1,2-MT VI-6 prepared from the cells grown at 27 degrees C exhibited the maximum activity at pH 7.0 and at 30 degrees C. The beta-1,2-MT VI-6 activity was only slightly affected by Mn2+, Mg2+, Ca2+, and ethylenediaminetetraacetic acid, but completely inhibited by Zn2+ and Ni2+. The beta-1,2-MT activities from the cells grown at 37 degrees C were lower than that from the cells grown at 27 degrees C, especially on the longer beta-1,2-mannooligosaccharides than tetraose.


Asunto(s)
Candida albicans/metabolismo , Manosa/biosíntesis , Manosiltransferasas/metabolismo , Candida albicans/enzimología , Manosa/química , Resonancia Magnética Nuclear Biomolecular
18.
Bioresour Technol ; 272: 209-216, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30340187

RESUMEN

Spent coffee grounds (SCG) or coffee residue wastes (CRW) provide excellent raw material for mannose and bioethanol production. In this study, SCG were used to produce valuable biosugars, including oligosaccharides (OSs), manno-oligosaccharides (MOSs), mannose, and bioethanol. SCG were subjected to delignification and defatting, producing SCG-derived polysaccharides. Two-stage enzymatic hydrolysis (short- and long-term) was performed to produce short-chain manno-oligosaccharides (MOSs) and monosaccharides (MSs), respectively. From 100 g dry weight (DW) amounts of SCG, approximately 77 g delignified SCG and 61 g SCG-derived polysaccharides, amounts of 15.9 g of first biosugars (mostly MOSs), 25.6 g of second biosugars (mostly MSs), and 3.1 g of bioethanol, were recovered. This technique may aid in the production of high-value mannose and OSs from SCG and other lignocellulosic biomasses that contain specific polysaccharides.


Asunto(s)
Café/metabolismo , Manosa/biosíntesis , Oligosacáridos/biosíntesis , Café/química , Hidrólisis , Polisacáridos/metabolismo
19.
FEBS Lett ; 582(4): 479-84, 2008 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-18201574

RESUMEN

GDP-N-acetyl-d-perosamine is a precursor of the LPS-O-antigen biosynthesis in Escherichia coli O157:H7. Like other GDP-6-deoxyhexoses, GDP-N-acetyl-d-perosamine is supposed to be synthesized via GDP-4-keto-6-deoxy-d-mannose, followed by a transamination- and an acetylation-reaction catalyzed by PerA and PerB. In this study, we have overproduced and purified PerA and PerB from E. coli O157:H7 in E. coli BL21. The recombinant proteins were partly characterized and the final product of the reaction catalyzed by PerB was shown to be GDP-N-acetyl-d-perosamine by chromatography, mass spectrometry, and 1H-NMR. The functional expression of PerB provides another enzymatically defined pathway for the synthesis of GDP-deoxyhexoses, which is needed to further study the corresponding glycosyltransferases in vitro.


Asunto(s)
Enzimas/metabolismo , Escherichia coli O157/enzimología , Manosa/análogos & derivados , Secuencia de Aminoácidos , Secuencia de Bases , Cromatografía Líquida de Alta Presión , Cromatografía en Capa Delgada , Clonación Molecular , Cartilla de ADN , Electroforesis en Gel de Poliacrilamida , Estabilidad de Enzimas , Enzimas/genética , Manosa/biosíntesis , Manosa/química , Espectrometría de Masas , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido
20.
Syst Appl Microbiol ; 31(3): 159-68, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18599240

RESUMEN

Mannosylglycerate (MG) is an intracellular organic solute found in some red algae, and several thermophilic bacteria and hyperthermophilic archaea. Glucosylglycerate (GG) was identified at the reducing end of a polysaccharide from mycobacteria and in a free form in a very few mesophilic bacteria and halophilic archaea. MG has a genuine role in the osmoadaptation and possibly in thermal protection of many hyper/thermophilic bacteria and archaea, but its role in red algae, where it was identified long before hyperthermophiles were even known to exist, remains to be clarified. The GG-containing polysaccharide was initially detected in Mycobacterium phlei and found to regulate fatty acid synthesis. More recently, GG has been found to be a major compatible solute under salt stress and nitrogen starvation in a few microorganisms. This review summarizes the occurrence and physiology of MG accumulation, as well as the distribution of GG, as a free solute or associated with larger macromolecules. We also focus on the recently identified pathways for the synthesis of both molecules, which were elucidated by studying hyper/thermophilic MG-accumulating organisms. The blooming era of genomics has now allowed the detection of these genes in fungi and mosses, opening a research avenue that spans the three domains of life, into the role of these two sugar derivatives.


Asunto(s)
Archaea , Bacterias , Hongos , Glucósidos/biosíntesis , Manosa/análogos & derivados , Rhodophyta , Adaptación Fisiológica , Archaea/clasificación , Archaea/fisiología , Bacterias/clasificación , Bacterias/metabolismo , Fenómenos Fisiológicos Bacterianos , Hongos/metabolismo , Glucósidos/química , Ácidos Glicéricos/química , Calor , Manosa/biosíntesis , Manosa/química , Concentración Osmolar , Rhodophyta/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA