Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
J Biol Chem ; 292(3): 1005-1028, 2017 01 20.
Article in English | MEDLINE | ID: mdl-27909055

ABSTRACT

Proteomes of even well characterized organisms still contain a high percentage of proteins with unknown or uncertain molecular and/or biological function. A significant fraction of those proteins is predicted to have catalytic properties. Here we aimed at identifying the function of the Saccharomyces cerevisiae Ydr109c protein and its human homolog FGGY, both of which belong to the broadly conserved FGGY family of carbohydrate kinases. Functionally identified members of this family phosphorylate 3- to 7-carbon sugars or sugar derivatives, but the endogenous substrate of S. cerevisiae Ydr109c and human FGGY has remained unknown. Untargeted metabolomics analysis of an S. cerevisiae deletion mutant of YDR109C revealed ribulose as one of the metabolites with the most significantly changed intracellular concentration as compared with a wild-type strain. In human HEK293 cells, ribulose could only be detected when ribitol was added to the cultivation medium, and under this condition, FGGY silencing led to ribulose accumulation. Biochemical characterization of the recombinant purified Ydr109c and FGGY proteins showed a clear substrate preference of both kinases for d-ribulose over a range of other sugars and sugar derivatives tested, including l-ribulose. Detailed sequence and structural analyses of Ydr109c and FGGY as well as homologs thereof furthermore allowed the definition of a 5-residue d-ribulokinase signature motif (TCSLV). The physiological role of the herein identified eukaryotic d-ribulokinase remains unclear, but we speculate that S. cerevisiae Ydr109c and human FGGY could act as metabolite repair enzymes, serving to re-phosphorylate free d-ribulose generated by promiscuous phosphatases from d-ribulose 5-phosphate. In human cells, FGGY can additionally participate in ribitol metabolism.


Subject(s)
Pentoses/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Amino Acid Motifs , Gene Silencing , HEK293 Cells , Humans , Pentoses/genetics , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Phosphotransferases (Alcohol Group Acceptor)/genetics , Proteins/chemistry , Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics
2.
Metab Eng ; 49: 257-266, 2018 09.
Article in English | MEDLINE | ID: mdl-30172686

ABSTRACT

Methanol is a potentially attractive substrate for bioproduction of chemicals because of the abundance of natural gas and biogas-derived methane. To move towards utilizing methanol as a sole carbon source, here we engineer an Escherichia coli strain to couple methanol utilization with growth on five-carbon (C5) sugars. By deleting essential genes in the pentose phosphate pathway for pentose utilization and expressing heterologous enzymes from the ribulose-monophosphate (RuMP) pathway, we constructed a strain that cannot grow on xylose or ribose minimal media unless methanol is utilized, creating a phenotype termed "synthetic methanol auxotrophy". Our best strains were able to utilize methanol for growth at a rate of 0.17 ±â€¯0.006 (h-1) with methanol and xylose co-assimilation at a molar ratio of approximately 1:1. Genome sequencing and reversion of mutations indicated that mutations on genes encoding for adenylate cyclase (cyaA) and the formaldehyde detoxification operon (frmRAB) were necessary for the growth phenotype. The methanol auxotrophic strain was further engineered to produce ethanol or 1-butanol to final titers of 4.6 g/L and 2.0 g/L, respectively. 13C tracing showed that 43% and 71% of ethanol and 1-butanol produced had labeled carbon derived from methanol, respectively.


Subject(s)
1-Butanol/metabolism , Escherichia coli , Ethanol/metabolism , Methanol/metabolism , Pentoses/metabolism , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Genome, Bacterial , Mutation , Operon , Pentoses/genetics
3.
Biochim Biophys Acta Gen Subj ; 1861(2): 79-85, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27816753

ABSTRACT

BACKGROUND: Ribulose-1,5-bisphosphate carboxylase/oxygenase (Rubisco) is the chloroplast enzyme that fixes CO2 in photosynthesis, but the enzyme also fixes O2, which leads to the wasteful photorespiratory pathway. If we better understand the structure-function relationship of the enzyme, we might be able to engineer improvements. When the crystal structure of Chlamydomonas Rubisco was solved, four new posttranslational modifications were observed which are not present in other species. The modifications were 4-hydroxylation of the conserved Pro-104 and 151 residues, and S-methylation of the variable Cys-256 and 369 residues, which are Phe-256 and Val-369 in land plants. Because the modifications were only observed in Chlamydomonas Rubisco, they might account for the differences in kinetic properties between the algal and plant enzymes. METHODS: Site-directed mutagenesis and chloroplast transformation have been used to test the essentiality of these modifications by replacing each of the residues with alanine (Ala). Biochemical analyses were done to determine the specificity factors and kinetic constants. RESULTS: Replacing the modified-residues in Chlamydomonas Rubisco affected the enzyme's catalytic activity. Substituting hydroxy-Pro-104 and methyl-Cys-256 with alanine influenced Rubisco catalysis. CONCLUSION: This is the first study on these posttranslationally-modified residues in Rubisco by genetic engineering. As these forms of modifications/regulation are not available in plants, the modified residues could be a means to modulate Rubisco activity. GENERAL SIGNIFICANCE: With a better understanding of Rubisco structure-function, we can define targets for improving the enzyme.


Subject(s)
Chlamydomonas reinhardtii/genetics , Mutation/genetics , Oxygenases/genetics , Protein Processing, Post-Translational/genetics , Ribulosephosphates/genetics , Alanine/genetics , Catalysis , Chloroplasts/genetics , Genetic Engineering/methods , Kinetics , Mutagenesis, Site-Directed/methods , Pentoses/genetics , Photosynthesis/genetics , Ribulose-Bisphosphate Carboxylase/genetics
4.
Biomolecules ; 10(7)2020 07 10.
Article in English | MEDLINE | ID: mdl-32664266

ABSTRACT

A fluorometabolite, 5-fluoro-5-deoxy-D-ribulose (5-FDRul), from the culture broth of the soil bacterium Streptomyces sp. MA37, was identified through a combination of genetic manipulation, chemo-enzymatic synthesis and NMR comparison. Although 5-FDRul has been chemically synthesized before, it was not an intermediate or a shunt product in previous studies of fluorometalism in S. cattleya. Our study of MA37 demonstrates that 5-FDRul is a naturally occurring fluorometabolite, rendering it a new addition to this rare collection of natural products. The genetic inactivation of key biosynthetic genes involved in the fluorometabolisms in MA37 resulted in the increased accumulation of unidentified fluorometabolites as observed from 19F-NMR spectral comparison among the wild type (WT) of MA37 and the mutated variants, providing evidence of the presence of other new biosynthetic enzymes involved in the fluorometabolite pathway in MA37.


Subject(s)
Biosynthetic Pathways , Culture Media/chemistry , Mutation , Pentoses/analysis , Streptomyces/growth & development , Bacterial Proteins/genetics , Fluorine-19 Magnetic Resonance Imaging , Halogenation , Multigene Family , Pentoses/genetics , Sequence Analysis, DNA , Soil Microbiology , Streptomyces/chemistry , Streptomyces/genetics
5.
Enzyme Microb Technol ; 132: 109443, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31731969

ABSTRACT

l-Ribose is an important pharmaceutical intermediate that is used in the synthesis of numerous antiviral and anticancer drugs. However, it is a non-natural and expensive rare sugar. Recently, the enzymatic synthesis of l-ribose has attracted considerable attention owing to its considerable advantages over chemical approaches. In this work, a new strategy was developed for the production of l-ribose from the inexpensive starting material l-arabinose. The l-arabinose isomerase (l-AIase) gene from Alicyclobacillus hesperidum and the d-lyxose isomerase (d-LIase) gene from Thermoflavimicrobium dichotomicum were cloned and co-expressed in Escherichia coli, resulting in recombinant cells harboring the vector pCDFDuet-Alhe-LAI/Thdi-DLI. The co-expression system exhibited optimal activity at a temperature of 70 °C and pH 6.0, and the addition of Co2+ enhanced the catalytic activity by 27.8-fold. The system containing 50 g L-1 of recombinant cells were relatively stable up to 55 °C. The co-expression system (50 g L-1 of recombinant cells) afforded 20.9, 39.7, and 50.3 g L-1 of l-ribose from initial l-arabinose concentrations of 100, 300, and 500 g L-1, corresponding to conversion rate of 20.9%, 13.2%, and 10.0%, respectively. Overall, this study provides a viable approach for producing l-ribose from l-arabinose under slightly acidic conditions using a co-expression system harboring l-AIase and d-LIase genes.


Subject(s)
Aldose-Ketose Isomerases/metabolism , Arabinose/metabolism , Pentoses/metabolism , Ribose/biosynthesis , Aldose-Ketose Isomerases/genetics , Alicyclobacillus/enzymology , Alicyclobacillus/genetics , Bacillales/enzymology , Bacillales/genetics , Cloning, Molecular , Escherichia coli/genetics , Hydrogen-Ion Concentration , Pentoses/genetics , Temperature
6.
Int J Biol Macromol ; 79: 807-12, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26047894

ABSTRACT

Glucuronic acids in Arabidopsis thaliana xylans exist in 4-O-methylated (MeGlcA) and non-methylated (GlcA) forms at a ratio of about 3:2. The matrix-assisted laser desorption/ionization mass spectrometry analysis of the endoxylanase liberated acidic oligosaccharides from the Arabidopsis inflorescence stem showed that two peaks with GlcA (GlcA-Xyl4Ac1 and GlcA-Xyl5Ac2) had abnormally high intensities, as well as different tandem mass spectra, than their 4-O-methylated counterparts. These peaks were interestingly enriched in the xylan biosynthesis mutant irx7 and irx9-1. Multi-stages fragmentation analysis using negative ion electrospray-ion trap mass spectrometry indicated that this GlcA was further carrying a pentose residue in the glucuronoxylan-derived oligosaccharide from irx9-1. The structure was also identified in Arabidopsis wild type. The results prove evidence of a new pentose substitution on the GlcA residue of Arabidopsis GX, which is likely present in the primary walls.


Subject(s)
Arabidopsis Proteins/genetics , Arabidopsis/chemistry , Glucuronic Acid/chemistry , Pentoses/chemistry , Pentosyltransferases/genetics , Xylans/genetics , Arabidopsis/genetics , Arabidopsis Proteins/chemistry , Endo-1,4-beta Xylanases/genetics , Gene Expression Regulation, Plant , Glucuronic Acid/genetics , Oligosaccharides/chemistry , Pentoses/genetics , Pentosyltransferases/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Xylans/biosynthesis , Xylans/chemistry
7.
Biotechnol Adv ; 31(6): 827-37, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23510903

ABSTRACT

Interest in thermophilic bacteria as live-cell catalysts in biofuel and biochemical industry has surged in recent years, due to their tolerance of high temperature and wide spectrum of carbon-sources that include cellulose. However their direct employment as microbial cellular factories in the highly demanding industrial conditions has been hindered by uncompetitive biofuel productivity, relatively low tolerance to solvent and osmic stresses, and limitation in genome engineering tools. In this work we review recent advances in dissecting and engineering the metabolic and regulatory networks of thermophilic bacteria for improving the traits of key interest in biofuel industry: cellulose degradation, pentose-hexose co-utilization, and tolerance of thermal, osmotic, and solvent stresses. Moreover, new technologies enabling more efficient genetic engineering of thermophiles were discussed, such as improved electroporation, ultrasound-mediated DNA delivery, as well as thermo-stable plasmids and functional selection systems. Expanded applications of such technological advancements in thermophilic microbes promise to substantiate a synthetic biology perspective, where functional parts, module, chassis, cells and consortia were modularly designed and rationally assembled for the many missions at industry and nature that demand the extraordinary talents of these extremophiles.


Subject(s)
Bacteria/enzymology , Biofuels/microbiology , Gene Regulatory Networks , Metabolic Engineering , Bacteria/genetics , Carbohydrate Metabolism/genetics , Cellulose/chemistry , Cellulose/genetics , Genetic Engineering , Hexoses/chemistry , Hexoses/genetics , Humans , Pentoses/chemistry , Pentoses/genetics , Synthetic Biology
8.
PLoS One ; 8(8): e72902, 2013.
Article in English | MEDLINE | ID: mdl-24015281

ABSTRACT

D-Galactose-6-phosphate isomerase from Lactobacillus rhamnosus (LacAB; EC 5.3.1.26), which is encoded by the tagatose-6-phosphate pathway gene cluster (lacABCD), catalyzes the isomerization of D-galactose-6-phosphate to D-tagatose-6-phosphate during lactose catabolism and is used to produce rare sugars as low-calorie natural sweeteners. The crystal structures of LacAB and its complex with D-tagatose-6-phosphate revealed that LacAB is a homotetramer of LacA and LacB subunits, with a structure similar to that of ribose-5-phosphate isomerase (Rpi). Structurally, LacAB belongs to the RpiB/LacAB superfamily, having a Rossmann-like αßα sandwich fold as has been identified in pentose phosphate isomerase and hexose phosphate isomerase. In contrast to other family members, the LacB subunit also has a unique α7 helix in its C-terminus. One active site is distinctly located at the interface between LacA and LacB, whereas two active sites are present in RpiB. In the structure of the product complex, the phosphate group of D-tagatose-6-phosphate is bound to three arginine residues, including Arg-39, producing a different substrate orientation than that in RpiB, where the substrate binds at Asp-43. Due to the proximity of the Arg-134 residue and backbone Cα of the α6 helix in LacA to the last Asp-172 residue of LacB with a hydrogen bond, a six-carbon sugar-phosphate can bind in the larger pocket of LacAB, compared with RpiB. His-96 in the active site is important for ring opening and substrate orientation, and Cys-65 is essential for the isomerization activity of the enzyme. Two rare sugar substrates, D-psicose and D-ribulose, show optimal binding in the LacAB-substrate complex. These findings were supported by the results of LacA activity assays.


Subject(s)
Aldose-Ketose Isomerases/chemistry , Hexosephosphates/chemistry , Lacticaseibacillus rhamnosus/enzymology , Protein Folding , Aldose-Ketose Isomerases/genetics , Aldose-Ketose Isomerases/metabolism , Catalytic Domain/physiology , Crystallography, X-Ray , Fructose/chemistry , Fructose/genetics , Fructose/metabolism , Hexosephosphates/genetics , Hexosephosphates/metabolism , Lacticaseibacillus rhamnosus/genetics , Pentoses/chemistry , Pentoses/genetics , Pentoses/metabolism , Protein Structure, Quaternary , Protein Structure, Secondary , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL