Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 279
Filtrar
Más filtros

País/Región como asunto
Intervalo de año de publicación
1.
Mol Microbiol ; 116(1): 245-259, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33629421

RESUMEN

Aspergillus fumigatus is an opportunistic mold responsible for severe life-threatening fungal infections in immunocompromised patients. The cell wall, an essential structure composed of glucan, chitin, and galactomannan, is considered to be a target for the development of antifungal drugs. The nucleotide sugar donor GDP-mannose (GDP-Man) is required for the biosynthesis of galactomannan, glycosylphosphatidylinositol (GPI) anchors, glycolipid, and protein glycosylation. Starting from fructose-6-phosphate, GDP-Man is produced by the sequential action of the enzymes phosphomannose isomerase, phosphomannomutase (Pmm), and GDP-mannose pyrophosphorylase. Here, using heterokaryon rescue and gene knockdown approaches we demonstrate that the phosphomannomutase encoding gene in A. fumigatus (pmmA) is essential for survival. Reduced expression of pmmA is associated with significant morphological defects including retarded germination, growth, reduced conidiation, and abnormal polarity. Moreover, the knockdown strain exhibited an altered cell wall organization and sensitivity toward cell wall perturbing agents. By solving the first crystal structure of A. fumigatus phosphomannomutase (AfPmmA) we identified non-conservative substitutions near the active site when compared to the human orthologues. Taken together, this work provides a genetic and structural foundation for the exploitation of AfPmmA as a potential antifungal target.


Asunto(s)
Aspergillus fumigatus/genética , Guanosina Difosfato Manosa/metabolismo , Fosfotransferasas (Fosfomutasas)/genética , Fosfotransferasas (Fosfomutasas)/metabolismo , Antifúngicos/farmacología , Aspergilosis/tratamiento farmacológico , Aspergilosis/patología , Aspergillus fumigatus/efectos de los fármacos , Aspergillus fumigatus/metabolismo , Pared Celular/metabolismo , Eliminación de Gen , Humanos , Virulencia/genética
2.
Microb Cell Fact ; 19(1): 32, 2020 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-32054531

RESUMEN

BACKGROUND: Bleomycin is a broad-spectrum glycopeptide antitumor antibiotic produced by Streptomyces verticillus. Clinically, the mixture of bleomycin A2 and bleomycin B2 is widely used in combination with other drugs for the treatment of various cancers. As a secondary metabolite, the biosynthesis of bleomycin is precisely controlled by the complex extra-/intracellular regulation mechanisms, it is imperative to investigate the global metabolic and regulatory system involved in bleomycin biosynthesis for increasing bleomycin production. RESULTS: N-acetylglucosamine (GlcNAc), the vital signaling molecule controlling the onset of development and antibiotic synthesis in Streptomyces, was found to increase the yields of bleomycins significantly in chemically defined medium. To mine the gene information relevant to GlcNAc metabolism, the DNA sequences of dasR-dasA-dasBCD-nagB and nagKA in S. verticillus were determined by chromosome walking. From the results of Real time fluorescence quantitative PCR (RT-qPCR) and electrophoretic mobility shift assays (EMSAs), the repression of the expression of nagB and nagKA by the global regulator DasR was released under induction with GlcNAc. The relief of blmT expression repression by BlmR was the main reason for increased bleomycin production. DasR, however, could not directly affect the expression of the pathway-specific repressor BlmR in the bleomycins gene cluster. With at the beginning of bleomycin synthesis, the supply of the specific precursor GDP-mannose played the key role in bleomycin production. Genetic engineering of the GDP-mannose synthesis pathway indicated that phosphomannose isomerase (ManA) and phosphomannomutase (ManB) were key enzymes for bleomycins synthesis. Here, the blmT, manA and manB co-expression strain OBlmT/ManAB was constructed. Based on GlcNAc regulation and assisted metabolic profiling analysis, the yields of bleomycin A2 and B2 were ultimately increased to 61.79 and 36.9 mg/L, respectively. CONCLUSIONS: Under GlcNAc induction, the elevated production of bleomycins was mainly associated with the alleviation of the inhibition of BlmT, so blmT and specific precursor synthesis pathways were genetically engineered for bleomycins production improvement. Combination with subsequent metabolomics analysis not only effectively increased the bleomycin yield, but also extended the utilization of chitin-derived substrates in microbial-based antibiotic production.


Asunto(s)
Acetilglucosamina/metabolismo , Bleomicina/biosíntesis , Guanosina Difosfato Manosa/metabolismo , Streptomyces/metabolismo , Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Redes y Vías Metabólicas , Metabolómica/métodos , Metabolismo Secundario
3.
Int J Mol Sci ; 20(14)2019 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-31330931

RESUMEN

GDP-mannose 3,5-epimerase (GM35E) catalyzes the conversion of GDP-mannose towards GDP-l-galactose and GDP-l-gulose. Although this reaction represents one of the few enzymatic routes towards the production of l-sugars and derivatives, it has not yet been exploited for that purpose. One of the reasons is that so far only GM35Es from plants have been characterized, yielding biocatalysts that are relatively unstable and difficult to express heterologously. Through the mining of sequence databases, we succeeded in identifying a promising bacterial homologue. The gene from the thermophilic organism Methylacidiphilum fumariolicum was codon optimized for expression in Escherichia coli, resulting in the production of 40 mg/L of recombinant protein. The enzyme was found to act as a self-sufficient GM35E, performing three chemical reactions in the same active site. Furthermore, the biocatalyst was highly stable at temperatures up to 55 °C, making it well suited for the synthesis of new carbohydrate products with application in the pharma industry.


Asunto(s)
Proteínas Bacterianas , Carbohidrato Epimerasas/química , Carbohidrato Epimerasas/metabolismo , Secuencia de Aminoácidos , Catálisis , Activación Enzimática , Estabilidad de Enzimas , Guanosina Difosfato Manosa/química , Guanosina Difosfato Manosa/metabolismo , Concentración de Iones de Hidrógeno , Modelos Moleculares , Estructura Molecular , Conformación Proteica , Proteínas Recombinantes , Relación Estructura-Actividad , Termodinámica
4.
Microbiology (Reading) ; 164(4): 614-624, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29493491

RESUMEN

Actinomycete bacteria use polyprenol phosphate mannose as a lipid linked sugar donor for extra-cytoplasmic glycosyl transferases that transfer mannose to cell envelope polymers, including glycoproteins and glycolipids. We showed recently that strains of Streptomyces coelicolor with mutations in the gene ppm1 encoding polyprenol phosphate mannose synthase were both resistant to phage φC31 and have greatly increased susceptibility to antibiotics that mostly act on cell wall biogenesis. Here we show that mutations in the genes encoding enzymes that act upstream of Ppm1 in the polyprenol phosphate mannose synthesis pathway can also confer phage resistance and antibiotic hyper-susceptibility. GDP-mannose is a substrate for Ppm1 and is synthesised by GDP-mannose pyrophosphorylase (GMP; ManC) which uses GTP and mannose-1-phosphate as substrates. Phosphomannomutase (PMM; ManB) converts mannose-6-phosphate to mannose-1-phosphate. S. coelicolor strains with knocked down GMP activity or with a mutation in sco3028 encoding PMM acquire phenotypes that resemble those of the ppm1- mutants i.e. φC31 resistant and susceptible to antibiotics. Differences in the phenotypes of the strains were observed, however. While the ppm1- strains have a small colony phenotype, the sco3028 :: Tn5062 mutants had an extremely small colony phenotype indicative of an even greater growth defect. Moreover we were unable to generate a strain in which GMP activity encoded by sco3039 and sco4238 is completely knocked out, indicating that GMP is also an important enzyme for growth. Possibly GDP-mannose is at a metabolic branch point that supplies alternative nucleotide sugar donors.


Asunto(s)
Antibacterianos/farmacología , Vías Biosintéticas , Guanosina Difosfato Manosa/metabolismo , Nucleotidiltransferasas/genética , Fosfotransferasas (Fosfomutasas)/genética , Streptomyces coelicolor/efectos de los fármacos , Streptomyces coelicolor/fisiología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Bacteriófagos/fisiología , Manosiltransferasas/genética , Manosiltransferasas/metabolismo , Mutación , Nucleotidiltransferasas/metabolismo , Fenotipo , Fosfotransferasas (Fosfomutasas)/metabolismo , Streptomyces coelicolor/virología
5.
Plant Cell ; 27(12): 3397-409, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26672069

RESUMEN

Humans are unable to synthesize l-ascorbic acid (AsA), yet it is required as a cofactor in many critical biochemical reactions. The majority of human dietary AsA is obtained from plants. In Arabidopsis thaliana, a GDP-mannose pyrophosphorylase (GMPP), VITAMIN C DEFECTIVE1 (VTC1), catalyzes a rate-limiting step in AsA synthesis: the formation of GDP-Man. In this study, we identified two nucleotide sugar pyrophosphorylase-like proteins, KONJAC1 (KJC1) and KJC2, which stimulate the activity of VTC1. The kjc1kjc2 double mutant exhibited severe dwarfism, indicating that KJC proteins are important for growth and development. The kjc1 mutation reduced GMPP activity to 10% of wild-type levels, leading to a 60% reduction in AsA levels. On the contrary, overexpression of KJC1 significantly increased GMPP activity. The kjc1 and kjc1kjc2 mutants also exhibited significantly reduced levels of glucomannan, which is also synthesized from GDP-Man. Recombinant KJC1 and KJC2 enhanced the GMPP activity of recombinant VTC1 in vitro, while KJCs did not show GMPP activity. Yeast two-hybrid assays suggested that the stimulation of GMPP activity occurs via interaction of KJCs with VTC1. These results suggest that KJCs are key factors for the generation of GDP-Man and affect AsA level and glucomannan accumulation through the stimulation of VTC1 GMPP activity.


Asunto(s)
Arabidopsis/genética , Ácido Ascórbico/metabolismo , Guanosina Difosfato Manosa/metabolismo , Mananos/metabolismo , Nucleotidiltransferasas/metabolismo , Vitaminas/metabolismo , Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Pared Celular/metabolismo , Regulación de la Expresión Génica de las Plantas , Mutación , Nucleotidiltransferasas/genética , Filogenia , Raíces de Plantas/genética , Raíces de Plantas/metabolismo , Plantas Modificadas Genéticamente , Plantones/genética , Plantones/metabolismo
6.
Biotechnol Bioeng ; 115(1): 192-205, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28922469

RESUMEN

Glycosylation of proteins is a key function of the biosynthetic-secretory pathway in the endoplasmic reticulum (ER) and Golgi apparatus. Glycosylated proteins play a crucial role in cell trafficking and signaling, cell-cell adhesion, blood-group antigenicity, and immune response. In addition, the glycosylation of proteins is an important parameter in the optimization of many glycoprotein-based drugs such as monoclonal antibodies. In vitro glycoengineering of proteins requires glycosyltransferases as well as expensive nucleotide sugars. Here, we present a designed pathway consisting of five enzymes, glucokinase (Glk), phosphomannomutase (ManB), mannose-1-phosphate-guanyltransferase (ManC), inorganic pyrophosphatase (PmPpA), and 1-domain polyphosphate kinase 2 (1D-Ppk2) expressed in E. coli for the cell-free production and regeneration of GDP-mannose from mannose and polyphosphate with catalytic amounts of GDP and ADP. It was shown that GDP-mannose is produced at various conditions, that is pH 7-8, temperature 25-35°C and co-factor concentrations of 5-20 mM MgCl2 . The maximum reaction rate of GDP-mannose achieved was 2.7 µM/min at 30°C and 10 mM MgCl2 producing 566 nmol GDP-mannose after a reaction time of 240 min. With respect to the initial GDP concentration (0.8 mM) this is equivalent to a yield of 71%. Additionally, the cascade was coupled to purified, transmembrane-deleted Alg1 (ALG1ΔTM), the first mannosyltransferase in the ER-associated lipid-linked oligosaccharide (LLO) assembly. Thereby, in a one-pot reaction, phytanyl-PP-(GlcNAc)2 -Man1 was produced with efficient nucleotide sugar regeneration for the first time. Phytanyl-PP-(GlcNAc)2 -Man1 can serve as a substrate for the synthesis of LLO for the cell-free in vitro glycosylation of proteins. A high-performance anion exchange chromatography method with UV and conductivity detection (HPAEC-UV/CD) assay was optimized and validated to determine the enzyme kinetics. The established kinetic model enabled the optimization of the GDP-mannose regenerating cascade and can further be used to study coupling of the GDP-mannose cascade with glycosyltransferases. Overall, the study envisages a first step towards the development of a platform for the cell-free production of LLOs as precursors for in vitro glycoengineering of proteins.


Asunto(s)
Enzimas/metabolismo , Escherichia coli/genética , Guanosina Difosfato Manosa/metabolismo , Lipopolisacáridos/metabolismo , Proteínas Recombinantes/metabolismo , Coenzimas/metabolismo , Enzimas/genética , Enzimas/aislamiento & purificación , Escherichia coli/metabolismo , Expresión Génica , Concentración de Iones de Hidrógeno , Cloruro de Magnesio/metabolismo , Manosa/metabolismo , Polifosfatos/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Temperatura
7.
Med Mycol ; 56(5): 621-630, 2018 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-29420778

RESUMEN

Systemic human fungal infections are increasingly common. Aspergillus species cause most of the airborne fungal infections. Life-threatening invasive aspergillosis was formerly found only in immune-suppressed patients, but recently some strains of A. fumigatus have become primary pathogens. Many fungal cell wall components are absent from mammalian systems, so they are potential drug targets. Cell-wall-targeting drugs such as echinocandins are used clinically, although echinocandin-resistant strains were discovered shortly after their introduction. Currently there are no fully effective anti-fungal drugs. Fungal cell wall glycoconjugates modulate human immune responses, as well as fungal cell adhesion, biofilm formation, and drug resistance. Guanosine diphosphate (GDP) mannose transporters (GMTs) transfer GDP-mannose from the cytosol to the Golgi lumen prior to mannosylation. Aspergillus nidulans GMTs are encoded by gmtA and gmtB. Here we elucidate the roles of A. nidulans GMTs. Strains engineered to lack either or both GMTs were assessed for hyphal and colonial morphology, cell wall ultrastructure, antifungal susceptibility, spore hydrophobicity, adherence and biofilm formation. The gmt-deleted strains had smaller colonies with reduced sporulation and with thicker hyphal walls. The gmtA deficient spores had reduced hydrophobicity and were less adherent and less able to form biofilms in vitro. Thus, gmtA not only participates in maintaining the cell wall integrity but also plays an important role in biofilm establishment and adherence of A. nidulans. These findings suggested that GMTs have roles in A. nidulans growth and cell-cell interaction and could be a potential target for new antifungals that target virulence determinants.


Asunto(s)
Aspergillus nidulans/crecimiento & desarrollo , Aspergillus nidulans/genética , Proteínas Portadoras/genética , Proteínas Fúngicas/genética , Antifúngicos/farmacología , Aspergillus nidulans/ultraestructura , Biopelículas/crecimiento & desarrollo , Adhesión Celular/fisiología , Pared Celular/ultraestructura , Equinocandinas/farmacología , Guanosina Difosfato Manosa/metabolismo , Hifa/crecimiento & desarrollo , Lipopéptidos/farmacología , Micafungina , Pruebas de Sensibilidad Microbiana , Viabilidad Microbiana/efectos de los fármacos , Morfogénesis , Eliminación de Secuencia , Esporas Fúngicas/crecimiento & desarrollo , Esporas Fúngicas/ultraestructura
8.
Biochem J ; 474(6): 897-905, 2017 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-28104756

RESUMEN

The obligate intracellular lifestyle of Plasmodium falciparum and the difficulties in obtaining sufficient amounts of biological material have hampered the study of specific metabolic pathways in the malaria parasite. Thus, for example, the pools of sugar nucleotides required to fuel glycosylation reactions have never been studied in-depth in well-synchronized asexual parasites or in other stages of its life cycle. These metabolites are of critical importance, especially considering the renewed interest in the presence of N-, O-, and other glycans in key parasite proteins. In this work, we adapted a liquid chromatography tandem mass spectrometry (LC-MS/MS) method based on the use of porous graphitic carbon (PGC) columns and MS-friendly solvents to quantify sugar nucleotides in the malaria parasite. We report the thorough quantification of the pools of these metabolites throughout the intraerythrocytic cycle of P. falciparum The sensitivity of the method enabled, for the first time, the targeted analysis of these glycosylation precursors in gametocytes, the parasite sexual stages that are transmissible to the mosquito vector.


Asunto(s)
Guanosina Difosfato Fucosa/metabolismo , Guanosina Difosfato Manosa/metabolismo , Azúcares de Guanosina Difosfato/metabolismo , Plasmodium falciparum/metabolismo , Uridina Difosfato Galactosa/metabolismo , Uridina Difosfato Glucosa/metabolismo , Uridina Difosfato N-Acetilgalactosamina/metabolismo , Cromatografía Liquida , Eritrocitos/parasitología , Gametogénesis/fisiología , Guanosina Difosfato Fucosa/análisis , Guanosina Difosfato Manosa/análisis , Azúcares de Guanosina Difosfato/análisis , Humanos , Estadios del Ciclo de Vida/fisiología , Plasmodium falciparum/crecimiento & desarrollo , Espectrometría de Masas en Tándem , Uridina Difosfato Galactosa/análisis , Uridina Difosfato Glucosa/análisis , Uridina Difosfato N-Acetilgalactosamina/análisis
9.
Am J Hum Genet ; 93(4): 727-34, 2013 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-24035193

RESUMEN

In guanosine diphosphate (GDP)-mannose pyrophosphorylase A (GMPPA), we identified a homozygous nonsense mutation that segregated with achalasia and alacrima, delayed developmental milestones, and gait abnormalities in a consanguineous Pakistani pedigree. Mutations in GMPPA were subsequently found in ten additional individuals from eight independent families affected by the combination of achalasia, alacrima, and neurological deficits. This autosomal-recessive disorder shows many similarities with triple A syndrome, which is characterized by achalasia, alacrima, and variable neurological deficits in combination with adrenal insufficiency. GMPPA is a largely uncharacterized homolog of GMPPB. GMPPB catalyzes the formation of GDP-mannose, which is an essential precursor of glycan moieties of glycoproteins and glycolipids and is associated with congenital and limb-girdle muscular dystrophies with hypoglycosylation of α-dystroglycan. Surprisingly, GDP-mannose pyrophosphorylase activity was unchanged and GDP-mannose levels were strongly increased in lymphoblasts of individuals with GMPPA mutations. This suggests that GMPPA might serve as a GMPPB regulatory subunit mediating feedback inhibition of GMPPB instead of displaying catalytic enzyme activity itself. Thus, a triple-A-like syndrome can be added to the growing list of congenital disorders of glycosylation, in which dysregulation rather than mere enzyme deficiency is the basal pathophysiological mechanism.


Asunto(s)
Codón sin Sentido , Genes Recesivos/genética , Guanosina Difosfato Manosa/genética , Discapacidad Intelectual/genética , Nucleotidiltransferasas/genética , Adolescente , Insuficiencia Suprarrenal/genética , Adulto , Niño , Consanguinidad , Acalasia del Esófago/genética , Enfermedades Hereditarias del Ojo/genética , Glicosilación , Guanosina Difosfato Manosa/metabolismo , Homocigoto , Humanos , Discapacidad Intelectual/enzimología , Enfermedades del Aparato Lagrimal/genética , Enfermedades del Sistema Nervioso/genética , Nucleotidiltransferasas/metabolismo , Linaje , Adulto Joven
10.
Am J Hum Genet ; 93(1): 29-41, 2013 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-23768512

RESUMEN

Congenital muscular dystrophies with hypoglycosylation of α-dystroglycan (α-DG) are a heterogeneous group of disorders often associated with brain and eye defects in addition to muscular dystrophy. Causative variants in 14 genes thought to be involved in the glycosylation of α-DG have been identified thus far. Allelic mutations in these genes might also cause milder limb-girdle muscular dystrophy phenotypes. Using a combination of exome and Sanger sequencing in eight unrelated individuals, we present evidence that mutations in guanosine diphosphate mannose (GDP-mannose) pyrophosphorylase B (GMPPB) can result in muscular dystrophy variants with hypoglycosylated α-DG. GMPPB catalyzes the formation of GDP-mannose from GTP and mannose-1-phosphate. GDP-mannose is required for O-mannosylation of proteins, including α-DG, and it is the substrate of cytosolic mannosyltransferases. We found reduced α-DG glycosylation in the muscle biopsies of affected individuals and in available fibroblasts. Overexpression of wild-type GMPPB in fibroblasts from an affected individual partially restored glycosylation of α-DG. Whereas wild-type GMPPB localized to the cytoplasm, five of the identified missense mutations caused formation of aggregates in the cytoplasm or near membrane protrusions. Additionally, knockdown of the GMPPB ortholog in zebrafish caused structural muscle defects with decreased motility, eye abnormalities, and reduced glycosylation of α-DG. Together, these data indicate that GMPPB mutations are responsible for congenital and limb-girdle muscular dystrophies with hypoglycosylation of α-DG.


Asunto(s)
Distroglicanos/metabolismo , Distrofia Muscular de Cinturas/genética , Mutación Missense , Nucleotidiltransferasas/metabolismo , Animales , Preescolar , Análisis Mutacional de ADN/métodos , Distroglicanos/genética , Anomalías del Ojo/patología , Femenino , Fibroblastos/enzimología , Fibroblastos/patología , Estudios de Asociación Genética/métodos , Glicosilación , Guanosina Difosfato Manosa/metabolismo , Heterocigoto , Humanos , Lactante , Recién Nacido , Masculino , Músculo Esquelético/enzimología , Músculo Esquelético/patología , Distrofia Muscular de Cinturas/enzimología , Nucleotidiltransferasas/genética , Pez Cebra/genética , Pez Cebra/metabolismo
11.
BMC Plant Biol ; 16: 62, 2016 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-26956020

RESUMEN

BACKGROUND: Saccharina japonica is an important commercial brown seaweed, its main product is alginate, which is used in food, textile and by the cosmetic and pharmaceutical industries. GDP-mannose dehydrogenase (GMD) is the key enzyme involved in the synthesis of alginate. However, little is known about GMD in S. japonica. Here we report comparative biochemical analysis of two GMD genes in S. japonica. RESULTS: Two GMD genes from S. japonica (Sjgmd1, Sjgmd2) were cloned. The open reading frame lengths of Sjgmd1, Sjgmd2 are 963 bp and 948 bp, respectively. Alignment analysis showed that the two SjGMD sequences shared 79.38 % identity. Both proteins possess the GGxCLPKDV and GxGxVG sequence motifs characteristic of the short-chain dehydrogenase/reductase superfamily. The optimum temperatures for SjGMDs were 30 °C (SjGMD1) and 20 °C (SjGMD2), and the optimum pH values were 8.0 (SjGMD1) and 8.25 (SjGMD2). Kinetic analysis demonstrated the Km values for the substrate GDP-mannose were 289 µM (SjGMD1) and 177 µM (SjGMD2), and the Km values for the cofactor NAD(+) were 139 µM (SjGMD1) and 195 µM (SjGMD2). The metal iron Zn(2+) is a potent inhibitor of SjGMD1 and SjGMD2. Real-time PCR analysis showed that heat and desiccation treatments resulted in a significant increase in Sjgmd1 and Sjgmd2 transcript abundance, suggesting that the SjGMDs are directly involved in the acclimitisation of S. japonica to abiotic stresses. CONCLUSION: Our work identified two novel genes encoding GMD in S. japonica, comparatively characterized their structural characteristics and enzyme kinetics, and revealed the function of GMD in the stress adaptability of S. japonica. The knowledge obtained here enriched our understanding of the alginate synthesis mechanism in S. japonica, and may promote further research on functional differences between GMD genes.


Asunto(s)
Deshidrogenasas de Carbohidratos/genética , Phaeophyceae/genética , Secuencia de Aminoácidos , Deshidrogenasas de Carbohidratos/aislamiento & purificación , Clonación Molecular , Perfilación de la Expresión Génica , Guanosina Difosfato Manosa/metabolismo , Phaeophyceae/enzimología , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Alineación de Secuencia
12.
Plant Cell ; 25(5): 1881-94, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23695979

RESUMEN

The Arabidopsis thaliana protein GOLGI-LOCALIZED NUCLEOTIDE SUGAR TRANSPORTER (GONST1) has been previously identified as a GDP-d-mannose transporter. It has been hypothesized that GONST1 provides precursors for the synthesis of cell wall polysaccharides, such as glucomannan. Here, we show that in vitro GONST1 can transport all four plant GDP-sugars. However, gonst1 mutants have no reduction in glucomannan quantity and show no detectable alterations in other cell wall polysaccharides. By contrast, we show that a class of glycosylated sphingolipids (glycosylinositol phosphoceramides [GIPCs]) contains Man and that this mannosylation is affected in gonst1. GONST1 therefore is a Golgi GDP-sugar transporter that specifically supplies GDP-Man to the Golgi lumen for GIPC synthesis. gonst1 plants have a dwarfed phenotype and a constitutive hypersensitive response with elevated salicylic acid levels. This suggests an unexpected role for GIPC sugar decorations in sphingolipid function and plant defense signaling. Additionally, we discuss these data in the context of substrate channeling within the Golgi.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Arabidopsis/metabolismo , Glicoesfingolípidos/metabolismo , Manosa/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Ácido Salicílico/metabolismo , Arabidopsis/genética , Proteínas de Arabidopsis/genética , Transporte Biológico/genética , Pared Celular/genética , Pared Celular/metabolismo , Glicosilación , Aparato de Golgi/metabolismo , Guanosina Difosfato Fucosa/metabolismo , Guanosina Difosfato Manosa/metabolismo , Azúcares de Guanosina Difosfato/metabolismo , Immunoblotting , Proteínas de Transporte de Membrana/genética , Microscopía Fluorescente , Mutación
13.
Microb Cell Fact ; 15(1): 142, 2016 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-27527078

RESUMEN

BACKGROUND: In this study we examine the integrity of the cell wall during scale up of a yeast fermentation process from laboratory scale (10 L) to industrial scale (10,000 L). In a previous study we observed a clear difference in the volume fraction occupied by yeast cells as revealed by wet cell weight (WCW) measurements between these scales. That study also included metabolite analysis which suggested hypoxia during scale up. Here we hypothesize that hypoxia weakens the yeast cell wall during the scale up, leading to changes in cell permeability, and/or cell mechanical resistance, which in turn may lead to the observed difference in WCW. We tested the cell wall integrity by probing the cell wall sensitivity to Zymolyase. Also exometabolomics data showed changes in supply of precursors for the glycosylation pathway. RESULTS: The results show a more sensitive cell wall later in the production process at industrial scale, while the sensitivity at early time points was similar at both scales. We also report exometabolomics data, in particular a link with the protein glycosylation pathway. Significantly lower levels of Man6P and progressively higher GDP-mannose indicated partially impaired incorporation of this sugar nucleotide during co- or post-translational protein glycosylation pathways at the 10,000 L compared to the 10 L scale. This impairment in glycosylation would be expected to affect cell wall integrity. Although cell viability from samples obtained at both scales were similar, cells harvested from 10 L bioreactors were able to re-initiate growth faster in fresh shake flask media than those harvested from the industrial scale. CONCLUSIONS: The results obtained help explain the WCW differences observed at both scales by hypoxia-triggered weakening of the yeast cell wall during the scale up.


Asunto(s)
Pared Celular/fisiología , Fermentación , Saccharomyces cerevisiae/crecimiento & desarrollo , Saccharomyces cerevisiae/metabolismo , Anaerobiosis , Técnicas de Cultivo Celular por Lotes , Reactores Biológicos , Glicosilación , Guanosina Difosfato Manosa/metabolismo , Hidrolasas/metabolismo , Microbiología Industrial , Manosafosfatos/metabolismo , Metaboloma , Procesamiento Proteico-Postraduccional , Saccharomyces cerevisiae/citología
14.
Oncology ; 88(5): 298-308, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25572677

RESUMEN

UNLABELLED: Objecitive: Fucosyltransferase 8 (FUT8), the only enzyme responsible for the core α1,6-fucosylation of asparagine-linked oligosaccharides of glycoproteins, is a vital enzyme in cancer development and progression. We examined FUT8 expression in non-small cell lung cancers (NSCLCs) to analyze its clinical significance. We also examined the expression of guanosine diphosphate-mannose-4,6-dehydratase (GMD), which is imperative for the synthesis of fucosylated oligosaccharides. METHODS: Using immunohistochemistry, we evaluated the expression of FUT8 and GMD in relation to patient survival and prognosis in potentially curatively resected NSCLCs. RESULTS: High expression of FUT8 was found in 67 of 129 NSCLCs (51.9%) and was significantly found in non-squamous cell carcinomas (p = 0.008). High expression of FUT8 was associated with poor survival (p = 0.03) and was also a significant and independent unfavorable prognostic factor in patients with potentially curatively resected NSCLCs (p = 0.047). High expression of GMD was significantly associated with high FUT8 expression (p = 0.04). CONCLUSIONS: High expression of FUT8 is associated with an unfavorable clinical outcome in patients with potentially curatively resected NSCLCs, suggesting that FUT8 can be a prognostic factor. The analysis of FUT8 expression and its core fucosylated products may provide new insights for the therapeutic targets of NSCLCs.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/enzimología , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Fucosiltransferasas/metabolismo , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/mortalidad , Adenocarcinoma/enzimología , Adenocarcinoma/mortalidad , Adenocarcinoma del Pulmón , Anciano , Western Blotting , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Femenino , Fucosiltransferasas/genética , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Guanosina Difosfato Manosa/metabolismo , Humanos , Hidroliasas/metabolismo , Inmunohistoquímica , Japón/epidemiología , Estimación de Kaplan-Meier , Pulmón/enzimología , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Valor Predictivo de las Pruebas , Pronóstico , Medición de Riesgo , Factores de Riesgo
15.
J Biol Chem ; 287(53): 44418-24, 2012 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-23139423

RESUMEN

Fungal cell walls frequently contain a polymer of mannose and galactose called galactomannan. In the pathogenic filamentous fungus Aspergillus fumigatus, this polysaccharide is made of a linear mannan backbone with side chains of galactofuran and is anchored to the plasma membrane via a glycosylphosphatidylinositol or is covalently linked to the cell wall. To date, the biosynthesis and significance of this polysaccharide are unknown. The present data demonstrate that deletion of the Golgi UDP-galactofuranose transporter GlfB or the GDP-mannose transporter GmtA leads to the absence of galactofuran or galactomannan, respectively. This indicates that the biosynthesis of galactomannan probably occurs in the lumen of the Golgi apparatus and thus contrasts with the biosynthesis of other fungal cell wall polysaccharides studied to date that takes place at the plasma membrane. Transglycosylation of galactomannan from the membrane to the cell wall is hypothesized because both the cell wall-bound and membrane-bound polysaccharide forms are affected in the generated mutants. Considering the severe growth defect of the A. fumigatus GmtA-deficient mutant, proving this paradigm might provide new targets for antifungal therapy.


Asunto(s)
Aspergillus fumigatus/metabolismo , Pared Celular/metabolismo , Guanosina Difosfato Manosa/metabolismo , Mananos/biosíntesis , Aspergillus fumigatus/química , Aspergillus fumigatus/genética , Proteínas Portadoras/genética , Pared Celular/química , Pared Celular/genética , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Galactosa/análogos & derivados , Mananos/química , Estructura Molecular
16.
Acta Biochim Biophys Sin (Shanghai) ; 45(9): 720-5, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23774504

RESUMEN

Human GDP-l-fucose synthase, also known as FX protein, synthesizes GDP-l-fucose from its substrate GDP-4-keto-6-deoxy-d-mannose. The reaction involves epimerization at both C-3 and C-5 followed by an NADPH-dependent reduction of the carbonyl at C-4. In this paper, the first crystal structure of human FX protein was determined at 2.37 Å resolution. The asymmetric unit of the crystal structure contains four molecules which form two homodimers. Each molecule consists of two domains, a Rossmann-fold NADPH-binding motif and a carboxyl terminal domain. Compared with the Escherichia coli GDP-l-fucose synthase, the overall structures of these two enzymes have four major differences. There are four loops in the structure of human FX protein corresponding to two α-helices and two ß-sheets in that of the E. coli enzyme. Besides, there are seven different amino acid residues binding with NAPDH comparing human FX protein with that from E. coli. The structure of human FX reveals the key catalytic residues and could be useful for the design of drugs for the treatment of inflammation, auto-immune diseases, and possibly certain types of cancer.


Asunto(s)
Carbohidrato Epimerasas/química , Cetona Oxidorreductasas/química , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Secuencia de Aminoácidos , Sitios de Unión , Biocatálisis , Carbohidrato Epimerasas/genética , Carbohidrato Epimerasas/metabolismo , Cristalografía por Rayos X , Escherichia coli/enzimología , Escherichia coli/genética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Guanosina Difosfato Manosa/análogos & derivados , Guanosina Difosfato Manosa/metabolismo , Azúcares de Guanosina Difosfato/metabolismo , Humanos , Cetona Oxidorreductasas/genética , Cetona Oxidorreductasas/metabolismo , Modelos Moleculares , NADP/química , NADP/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
17.
Parasite ; 19(1): 63-70, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22314241

RESUMEN

Leishmaniases are tropical and sub-tropical diseases for which classical drugs (i.e. antimonials) exhibit toxicity and drug resistance. Such a situation requires to find new chemical series with antileishmanial activity. This work consists in analyzing the structure of a validated target in Leishmania: the GDP-mannose pyrophosphorylase (GDP-MP), an enzyme involved in glycosylation and essential for amastigote survival. By comparing both human and L. infantum GDP-MP 3D homology models, we identified (i) a common motif of amino acids that binds to the mannose moiety of the substrate and, interestingly, (ii) a motif that is specific to the catalytic site of the parasite enzyme. This motif could then be used to design compounds that specifically inhibit the leishmanial GDP-MP, without any effect on the human homolog.


Asunto(s)
Antiprotozoarios/farmacología , Diseño de Fármacos , Leishmania infantum/enzimología , Nucleotidiltransferasas/química , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Antiprotozoarios/química , Antiprotozoarios/uso terapéutico , Secuencia de Consenso , Perros , Glicosilación , Guanosina Difosfato Manosa/química , Guanosina Difosfato Manosa/metabolismo , Interacciones Huésped-Parásitos , Humanos , Leishmania infantum/efectos de los fármacos , Leishmaniasis Visceral/tratamiento farmacológico , Modelos Moleculares , Conformación Molecular , Nucleotidiltransferasas/antagonistas & inhibidores , Nucleotidiltransferasas/metabolismo , Alineación de Secuencia , Especificidad de la Especie
18.
J Biol Chem ; 285(35): 27468-27476, 2010 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-20573954

RESUMEN

GMP catalyzes the formation of GDP-Man, a fundamental precursor for protein glycosylation and bacterial cell wall and capsular polysaccharide biosynthesis. Crystal structures of GMP from the thermophilic bacterium Thermotoga maritima in the apo form, in complex with the substrates mannose-1-phosphate or GTP and bound with the end product GDP-Man in the presence of the essential divalent cation Mg(2+), were solved in the 2.1-2.8 A resolution range. The T. maritima GMP molecule is organized in two separate domains: a N-terminal Rossman fold-like domain and a C-terminal left-handed beta-helix domain. Two molecules associate into a dimer through a tail-to-tail arrangement of the C-terminal domains. Comparative analysis of the structures along with characterization of enzymatic parameters reveals the bases of substrate specificity of this class of sugar nucleotidyltransferases. In particular, substrate and product binding are associated with significant changes in the conformation of loop regions lining the active center and in the relative orientation of the two domains. Involvement of both the N- and C-terminal domains, coupled to the catalytic role of a bivalent metal ion, highlights the catalytic features of bacterial GMPs compared with other members of the pyrophosphorylase superfamily.


Asunto(s)
Proteínas Bacterianas/química , Cationes Bivalentes/química , Pared Celular/enzimología , Magnesio/química , Nucleotidiltransferasas/química , Thermotoga maritima/enzimología , Proteínas Bacterianas/metabolismo , Cationes Bivalentes/metabolismo , Cristalografía por Rayos X , Guanosina Difosfato Manosa/química , Guanosina Difosfato Manosa/metabolismo , Magnesio/metabolismo , Nucleotidiltransferasas/metabolismo , Polisacáridos Bacterianos/biosíntesis , Estructura Terciaria de Proteína , Especificidad por Sustrato
19.
Proteins ; 79(8): 2455-66, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21638333

RESUMEN

The Nudix hydrolase superfamily, characterized by the presence of the signature sequence GX(5)EX(7)REUXEEXGU (where U is I, L, or V), is a well-studied family in which relations have been established between primary sequence and substrate specificity for many members. For example, enzymes that hydrolyze the diphosphate linkage of ADP-ribose are characterized by having a proline 15 amino acids C-terminal of the Nudix signature sequence. GDPMK is a Nudix enzyme that conserves this characteristic proline but uses GDP-mannose as the preferred substrate. By investigating the structure of the GDPMK alone, bound to magnesium, and bound to substrate, the structural basis for this divergent substrate specificity and a new rule was identified by which ADP-ribose pyrophosphatases can be distinguished from purine-DP-mannose pyrophosphatases from primary sequence alone. Kinetic and mutagenesis studies showed that GDPMK hydrolysis does not rely on a single glutamate as the catalytic base. Instead, catalysis is dependent on residues that coordinate the magnesium ions and residues that position the substrate properly for catalysis. GDPMK was thought to play a role in biofilm formation because of its upregulation in response to RcsC signaling; however, GDPMK knockout strains show no defect in their capacity of forming biofilms.


Asunto(s)
Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Guanosina Difosfato Manosa/química , Guanosina Difosfato Manosa/metabolismo , Manosa/metabolismo , Pirofosfatasas/química , Pirofosfatasas/metabolismo , Secuencia de Aminoácidos , Datos de Secuencia Molecular , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Especificidad por Sustrato , Hidrolasas Nudix
20.
Glycobiology ; 21(7): 903-13, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21367879

RESUMEN

We describe the first biochemical characterization of the gumI gene product, an essential protein for xanthan polysaccharide synthesis. Cellular fractionation experiments reveal the presence of a protein associated with the membrane fraction, even in the absence of the other proteins responsible for the synthesis of glycolipid intermediates and the proteins involved in the polymerization and transport of the xanthan chains. By alkaline buffer extraction and detergent phase partitioning, GumI was categorized as a monotopic membrane protein. GumI was overexpressed in Escherichia coli, solubilized and purified in an active and stable form using a simple and reproducible two-step procedure. The purified recombinant GumI is a nonprocessive ß-mannosyltransferase that uses GDP-Man as a donor substrate and glucuronic acid-ß-1,2-mannose-α-1,3-glucose-ß-1,4-glucose-PP-polyisoprenyl as an acceptor. We also established the optimal biochemical conditions for GumI enzymatic activity. Sequence analysis revealed the presence of a conserved domain for glycosyltransferases (GTs) of the GT-B superfamily and homologous proteins in several prokaryote organisms. On the basis of this biochemical characterization, GumI may represent the founding member of a new GT family in the Carbohydrate-Active EnZymes classification.


Asunto(s)
Proteínas Bacterianas/aislamiento & purificación , Proteínas Bacterianas/metabolismo , Guanosina Difosfato Manosa/metabolismo , Manosiltransferasas/aislamiento & purificación , Manosiltransferasas/metabolismo , Polisacáridos Bacterianos/metabolismo , Xanthomonas campestris/enzimología , Proteínas Bacterianas/genética , Prueba de Complementación Genética , Manosiltransferasas/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Fracciones Subcelulares
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA