Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Appl Environ Microbiol ; 66(5): 1974-9, 2000 May.
Article in English | MEDLINE | ID: mdl-10788369

ABSTRACT

Diglycerol phosphate accumulates under salt stress in the archaeon Archaeoglobus fulgidus (L. O. Martins, R. Huber, H. Huber, K. O. Stetter, M. S. da Costa, and H. Santos, Appl. Environ. Microbiol. 63:896-902, 1997). This solute was purified after extraction from the cell biomass. In addition, the optically active and the optically inactive (racemic) forms of the compound were synthesized, and the ability of the solute to act as a protecting agent against heating was tested on several proteins derived from mesophilic or hyperthermophilic sources. Diglycerol phosphate exerted a considerable stabilizing effect against heat inactivation of rabbit muscle lactate dehydrogenase, baker's yeast alcohol dehydrogenase, and Thermococcus litoralis glutamate dehydrogenase. Highly homologous and structurally well-characterized rubredoxins from Desulfovibrio gigas, Desulfovibrio desulfuricans (ATCC 27774), and Clostridium pasteurianum were also examined for their thermal stabilities in the presence or absence of diglycerol phosphate, glycerol, and inorganic phosphate. These proteins showed different intrinsic thermostabilities, with half-lives in the range of 30 to 100 min. Diglycerol phosphate exerted a strong protecting effect, with approximately a fourfold increase in the half-lives for the loss of the visible spectra of D. gigas and C. pasteurianum rubredoxins. In contrast, the stability of D. desulfuricans rubredoxin was not affected. These different behaviors are discussed in the light of the known structural features of rubredoxins. The data show that diglycerol phosphate is a potentially useful protein stabilizer in biotechnological applications.


Subject(s)
Archaeoglobus fulgidus/chemistry , Enzymes/chemistry , Glycerophosphates/pharmacology , Rubredoxins/chemistry , Alcohol Dehydrogenase/chemistry , Alcohol Dehydrogenase/drug effects , Animals , Archaeoglobus fulgidus/physiology , Biomass , Cloning, Molecular , Clostridium/metabolism , Desulfovibrio/metabolism , Drug Stability , Enzyme Stability , Enzymes/drug effects , Glutamate Dehydrogenase/chemistry , Glutamate Dehydrogenase/drug effects , Glycerol/pharmacology , Glycerophosphates/chemical synthesis , Glycerophosphates/isolation & purification , Hot Temperature , L-Lactate Dehydrogenase/chemistry , L-Lactate Dehydrogenase/drug effects , Muscle, Skeletal/enzymology , Phosphates/pharmacology , Rabbits , Recombinant Proteins/chemistry , Recombinant Proteins/drug effects , Rubredoxins/drug effects , Saccharomyces cerevisiae/enzymology , Thermococcus/enzymology
2.
EMBO J ; 19(5): 831-42, 2000 Mar 01.
Article in English | MEDLINE | ID: mdl-10698925

ABSTRACT

MalY represents a bifunctional pyridoxal 5'-phosphate-dependent enzyme acting as a beta-cystathionase and as a repressor of the maltose regulon. Here we present the crystal structures of wild-type and A221V mutant protein. Each subunit of the MalY dimer is composed of a large pyridoxal 5'-phosphate-binding domain and a small domain similar to aminotransferases. The structural alignment with related enzymes identifies residues that are generally responsible for beta-lyase activity and depicts a unique binding mode of the pyridoxal 5'-phosphate correlated with a larger, more flexible substrate-binding pocket. In a screen for MalY mutants with reduced mal repressor properties, mutations occurred in three clusters: I, 83-84; II, 181-189 and III, 215-221, which constitute a clearly distinguished region in the MalY crystal structure far away from the cofactor. The tertiary structure of one of these mutants (A221V) demonstrates that positional rearrangements are indeed restricted to regions I, II and III. Therefore, we propose that a direct protein-protein interaction with MalT, the central transcriptional activator of the maltose system, underlies MalY-dependent repression of the maltose system.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cystathionine gamma-Lyase/chemistry , Cystathionine gamma-Lyase/metabolism , Escherichia coli Proteins , Escherichia coli/enzymology , Maltose/metabolism , Protein Conformation , Repressor Proteins , Crystallography, X-Ray , Escherichia coli/chemistry , Escherichia coli/genetics , Gene Expression Regulation, Bacterial , Maltose/genetics , Structure-Activity Relationship
3.
J Bacteriol ; 181(11): 3358-67, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10348846

ABSTRACT

Maltose metabolism was investigated in the hyperthermophilic archaeon Thermococcus litoralis. Maltose was degraded by the concerted action of 4-alpha-glucanotransferase and maltodextrin phosphorylase (MalP). The first enzyme produced glucose and a series of maltodextrins that could be acted upon by MalP when the chain length of glucose residues was equal or higher than four, to produce glucose-1-phosphate. Phosphoglucomutase activity was also detected in T. litoralis cell extracts. Glucose derived from the action of 4-alpha-glucanotransferase was subsequently metabolized via an Embden-Meyerhof pathway. The closely related organism Pyrococcus furiosus used a different metabolic strategy in which maltose was cleaved primarily by the action of an alpha-glucosidase, a p-nitrophenyl-alpha-D-glucopyranoside (PNPG)-hydrolyzing enzyme, producing glucose from maltose. A PNPG-hydrolyzing activity was also detected in T. litoralis, but maltose was not a substrate for this enzyme. The two key enzymes in the pathway for maltose catabolism in T. litoralis were purified to homogeneity and characterized; they were constitutively synthesized, although phosphorylase expression was twofold induced by maltodextrins or maltose. The gene encoding MalP was obtained by complementation in Escherichia coli and sequenced (calculated molecular mass, 96,622 Da). The enzyme purified from the organism had a specific activity for maltoheptaose, at the temperature for maximal activity (98 degrees C), of 66 U/mg. A Km of 0.46 mM was determined with heptaose as the substrate at 60 degrees C. The deduced amino acid sequence had a high degree of identity with that of the putative enzyme from the hyperthermophilic archaeon Pyrococcus horikoshii OT3 (66%) and with sequences of the enzymes from the hyperthermophilic bacterium Thermotoga maritima (60%) and Mycobacterium tuberculosis (31%) but not with that of the enzyme from E. coli (13%). The consensus binding site for pyridoxal 5'-phosphate is conserved in the T. litoralis enzyme.


Subject(s)
Glucosyltransferases/metabolism , Glycogen Debranching Enzyme System/metabolism , Maltose/metabolism , Thermococcus/enzymology , Amino Acid Sequence , Binding Sites , Cloning, Molecular , Enzyme Induction , Glucose/metabolism , Glucosephosphates/metabolism , Glucosyltransferases/chemistry , Glucosyltransferases/genetics , Glucosyltransferases/isolation & purification , Glycogen Debranching Enzyme System/isolation & purification , Maltose/pharmacology , Molecular Sequence Data , Molecular Weight , Nitrophenylgalactosides/metabolism , Oligosaccharides/metabolism , Oligosaccharides/pharmacology , Phosphoglucomutase/metabolism , Polysaccharides/metabolism , Polysaccharides/pharmacology , Pyridoxal Phosphate/metabolism , Sequence Alignment , Temperature , Thermococcus/drug effects , Thermococcus/growth & development , Thermococcus/metabolism
4.
J Bacteriol ; 180(10): 2630-5, 1998 May.
Article in English | MEDLINE | ID: mdl-9573146

ABSTRACT

Klebsiella oxytoca M5a1 has the capacity to transport and to metabolize alpha-, beta- and gamma-cyclodextrins. Cyclodextrin transport is mediated by the products of the cymE, cymF, cymG, cymD, and cymA genes, which are functionally homologous to the malE, malF, malG, malK, and lamB gene products of Escherichia coli. CymE, which is the periplasmic binding protein, has been overproduced and purified. By substrate-induced fluorescence quenching, the binding of ligands was analyzed. CymE bound alpha-cyclodextrin, beta-cyclodextrin, and gamma-cyclodextrin, with dissociation constants (Kd) of 0.02, 0.14 and 0.30 microM, respectively, and linear maltoheptaose, with a Kd of 70 microM. In transport experiments, alpha-cyclodextrin was taken up by the cym system of K. oxytoca three to five times less efficiently than maltohexaose by the E. coli maltose system. Besides alpha-cyclodextrin, maltohexaose was also taken up by the K. oxytoca cym system, but because of the inability of maltodextrins to induce the cym system, growth of E. coli mal mutants on linear maltodextrin was not observed when the cells harbored only the cym uptake system. Strains which gained this capacity by mutation could easily be selected, however.


Subject(s)
Bacterial Proteins/metabolism , Carrier Proteins/isolation & purification , Cyclodextrins/metabolism , Genes, Bacterial/physiology , Klebsiella/metabolism , Polysaccharides/metabolism , Biological Transport , Carrier Proteins/metabolism
5.
J Bacteriol ; 179(24): 7679-86, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9401025

ABSTRACT

malQ mutants of Escherichia coli lacking amylomaltase cannot grow on maltose. They express the maltose system constitutively and are sensitive to maltose when grown on another carbon source. In an attempt to isolate a multicopy suppressor that would result in growth on maltose, we transformed a malQ mutant with a gene bank of E. coli DNA which had been digested with Sau3a and cloned in pBR322. We screened the transformants on MacConkey maltose plates. A colony was isolated that appeared to be resistant to maltose and was pink on these plates, but it was still unable to grow on minimal medium with maltose as the carbon source. The plasmid was isolated, and the gene causing this phenotype was characterized. The deduced amino acid sequence of the encoded protein shows homology to that of lipases and esterases. We termed the gene aes, for acetyl esterase. Extracts of cells harboring plasmid-encoded aes under its own promoter exhibit a fivefold higher capacity to hydrolyze p-nitrophenyl acetate than do extracts of cells of plasmid-free strains. Similarly, strains harboring plasmid-encoded aes are able to grow on triacetyl glycerol (triacetin) whereas the plasmid-free strains are not. The expression of plasmid-encoded aes resulted in strong repression of the maltose transport genes in malT+ strains (10-fold reduction), but not in a malT(Con) strain which is independent of the inducer. Also, overproduction of MalT counteracted the Aes-dependent repression, indicating a direct interaction between MalT and Aes.


Subject(s)
Acetylesterase/genetics , Bacterial Proteins/genetics , Escherichia coli/genetics , Genes, Bacterial , Amino Acid Sequence , Base Sequence , Biological Transport , Cytoplasm/enzymology , Escherichia coli/enzymology , Gene Expression Regulation, Bacterial , Maltose/metabolism , Molecular Sequence Data , Plasmids/genetics , Sequence Homology, Amino Acid
6.
J Bacteriol ; 179(4): 1298-306, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9023215

ABSTRACT

glk, the structural gene for glucokinase of Escherichia coli, was cloned and sequenced. Overexpression of glk resulted in the synthesis of a cytoplasmic protein with a molecular weight of 35,000. The enzyme was purified, and its kinetic parameters were determined. Its Km values for glucose and ATP were 0.78 and 3.76 mM, respectively. Its Vmax was 158 U/mg of protein. A chromosomal glk-lacZ fusion was constructed and used to monitor glk expression. Under all conditions tested, only growth on glucose reduced the expression of glk by about 50%. A fruR mutation slightly increased the expression of glk-lacZ, whereas the overexpression of plasmid-encoded fruR+ weakly decreased expression. A FruR consensus binding motif was found 123 bp upstream of the potential transcriptional start site of glk. Overexpression of glk interfered with the expression of the maltose system. Repression was strongest in strains that exhibited constitutive mal gene expression due to endogenous induction and, in the absence of a functional MalK protein, the ATP-hydrolyzing subunit of the maltose transport system. It was least effective in wild-type strains growing on maltose or in strains constitutive for the maltose system due to a mutation in malT rendering the mal gene expression independent of inducer. This demonstrates that free internal glucose plays an essential role in the formation of the endogenous inducer of the maltose system.


Subject(s)
Escherichia coli Proteins , Escherichia coli/enzymology , Glucokinase/genetics , Glucokinase/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Base Sequence , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/growth & development , Gene Expression Regulation, Bacterial , Genes, Bacterial , Glucokinase/chemistry , Glucokinase/isolation & purification , Glucose/metabolism , Kinetics , Maltose/metabolism , Molecular Sequence Data , Mutagenesis, Insertional , Mutation , Recombinant Fusion Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism
7.
Appl Environ Microbiol ; 62(10): 3861-3, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8837441

ABSTRACT

We report an improvement of a published procedure using Escherichia coli to synthesize 14C-labeled trehalose from [14C]glucose (B. Brand and W. Boos, Appl. Environ. Microbiol. 55:2414-2415, 1989). Instead of inducing the expression of the trehalose-synthesizing enzymes encoded by the chromosomal genes otsAB by high osmolarity, we now induce their expression from a plasmid under normal growth conditions by the addition of IPTG (isopropyl-beta-D-thiogalactopyranoside). Instead of using a pgi zwf double mutant to prevent glucose utilization, we use a pgi::Tn10 insertion only. In addition to being defective in treA, which encodes a periplasmic trehalase, the strain is now also defective in treF, which encodes a newly discovered cytoplasmic trehalase. This strain is genetically stable; it has no growth defects; and after induction with IPTG, it will transform [14C]glucose to [14C]trehalose in minimal medium without any carbon source under aerobic conditions at a rate of 3 nmol/min/10(9) cells. With the improved method, the overall yield of trehalose from glucose is about 80% and the process takes place without dilution of the specific radioactivity of the glucose residues. The accumulated trehalose is extracted from the bacteria by 70% hot ethanol and can easily be purified radiochemically by chromatographic techniques.


Subject(s)
Escherichia coli/metabolism , Trehalose/biosynthesis , DNA Transposable Elements/genetics , Disaccharidases/genetics , Escherichia coli/genetics , Gene Expression Regulation, Bacterial/drug effects , Genetic Vectors/genetics , Glucose/metabolism , Glucose-6-Phosphate Isomerase/genetics , Glucosyltransferases/genetics , Isopropyl Thiogalactoside/pharmacology , Kinetics , Phosphoric Monoester Hydrolases/genetics , Trehalase/genetics , Trehalose/metabolism
8.
J Bacteriol ; 178(16): 4773-7, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8759837

ABSTRACT

The hyperthermophilic marine archaeon Thermococcus litoralis exhibits high-affinity transport activity for maltose and trehalose at 85 degrees C. The K(m) for maltose transport was 22 nM, and that for trehalose was 17 nM. In cells that had been grown on peptone plus yeast extract, the Vmax for maltose uptake ranged from 3.2 to 7.5 nmol/min/mg of protein in different cell cultures. Cells grown in peptone without yeast extract did not show significant maltose or trehalose uptake. We found that the compound in yeast extract responsible for the induction of the maltose and trehalose transport system was trehalose. [14C]maltose uptake at 100 nM was not significantly inhibited by glucose, sucrose, or maltotriose at a 100 microM concentration but was completely inhibited by trehalose and maltose. The inhibitor constant, Ki, of trehalose for inhibiting maltose uptake was 21 nM. In contrast, the ability of maltose to inhibit the uptake of trehalose was not equally strong. With 20 nM [14C]trehalose as the substrate, a 10-fold excess of maltose was necessary to inhibit uptake to 50%. However, full inhibition was observed at 2 microM maltose. The detergent-solubilized membranes of trehalose-induced cells contained a high-affinity binding protein for maltose and trehalose, with an M(r) of 48,000, that exhibited the same substrate specificity as the transport system found in whole cells. We conclude that maltose and trehalose are transported by the same high-affinity membrane-associated system. This represents the first report on sugar transport in any hyperthermophilic archaeon.


Subject(s)
Archaea/metabolism , Maltose/metabolism , Membrane Proteins/metabolism , Trehalose/metabolism , Archaea/growth & development , Biological Transport/drug effects , Carbohydrates/pharmacology , Carbon Radioisotopes , Carrier Proteins/isolation & purification , Carrier Proteins/metabolism , Cell Membrane/metabolism , Culture Media , Hot Temperature , Kinetics , Membrane Proteins/isolation & purification , Peptones , Radioisotope Dilution Technique , Saccharomyces cerevisiae
9.
J Biol Chem ; 271(18): 10681-9, 1996 May 03.
Article in English | MEDLINE | ID: mdl-8631875

ABSTRACT

malZ is a member of the mal regulon. It is controlled by MatT, the transcriptional activator of the maltose system. MalZ has been purified and identified as an enzyme hydrolyzing maltotriose and longer maltodextrins to glucose and maltose. MalZ is dispensable for growth on maltose or maltodextrins. Mutants lacking amylomaltase (encoded by malQ), the major maltose utilizing enzyme, cannot grow on maltose, maltotriose, or maltotetraose, despite the fact that they contain an effective transport system and MalZ. From such a malQ mutant a pseudorevertant was isolated that was able to grow on maltose. The suppressor mutation was mapped in malZ. The mutant gene was cloned. It contained a Trp to Cys exchange at position 292 of the deduced protein sequence. Surprisingly, the purified mutant enzyme was still unable to hydrolyze maltose as was the wild type enzyme, while both were able to release glucose from maltodextrins. However, the mutant enzyme had gained the ability to transfer dextrinyl moieties to glucose, maltose, and other maltodextrins. Thus, it had gained an activity associated with amylomaltase. It was the MalZ292-associated transferase reaction that allowed the utilization of maltose. In addition, we discovered that mutant and wild type enzyme alike were highly active as gamma-cyclodextrinases.


Subject(s)
Bacterial Proteins/metabolism , DNA-Binding Proteins , Escherichia coli Proteins , Escherichia coli/enzymology , Glucosyltransferases/biosynthesis , Glycoside Hydrolases/metabolism , Mutation , Transcription Factors , Bacterial Proteins/genetics , Base Sequence , Escherichia coli/genetics , Genes, Bacterial , Glucosyltransferases/metabolism , Glycogen Debranching Enzyme System/metabolism , Glycoside Hydrolases/genetics , Hydrolysis , Molecular Sequence Data , Polysaccharides/metabolism
10.
J Bacteriol ; 177(17): 5035-9, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7665481

ABSTRACT

The Escherichia coli maltose system consists of a number of genes whose products are involved in the uptake and metabolism of maltose and maltodextrins. MalT is the central positive gene activator of the regulon and is, together with the cyclic AMP-catabolite gene activator protein system, necessary for the expression of the maltose genes. Expression of malY, a MalT-independent gene, leads to the repression of all MalT-dependent genes. We have purified MalY to homogeneity and found it to be a pyridoxal-5-phosphate-containing enzyme with the enzymatic activity of a beta C-S lyase (cystathionase). MalY is a monomeric protein of 42,000 to 44,000 Da. Strains expressing MalY constitutively abolish the methionine requirement of metC mutants. The enzymatic activity of MetC, the cleavage of cystathionine to homocysteine, ammonia, and pyruvate, can be catalyzed by MalY. However, the cystathionase activity is not required for the function of MalY in repressing the maltose system. By site-directed mutagenesis, we changed the conserved lysine residue at the pyridoxal phosphate binding site (position 233) of MalY to isoleucine. This abolished beta C-S lyase activity but not the ability of the protein to repress the maltose system. Also, the overexpression of plasmid-encoded metC did not affect mal gene expression, nor did the deduced amino acid sequence of MetC show homology to that of MalY.


Subject(s)
Bacterial Proteins/isolation & purification , Cystathionine gamma-Lyase/isolation & purification , Escherichia coli Proteins , Escherichia coli/enzymology , Repressor Proteins/isolation & purification , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Base Sequence , Cystathionine gamma-Lyase/biosynthesis , Cystathionine gamma-Lyase/genetics , Escherichia coli/genetics , Maltose/metabolism , Molecular Sequence Data , Mutagenesis , Recombinant Proteins/biosynthesis , Regulon/genetics , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Sequence Homology, Amino Acid
11.
J Bacteriol ; 175(17): 5655-65, 1993 Sep.
Article in English | MEDLINE | ID: mdl-8366051

ABSTRACT

The maltose system in Escherichia coli consists of cell envelope-associated proteins and enzymes that catalyze the uptake and utilization of maltose and alpha,1-4-linked maltodextrins. The presence of these sugars in the growth medium induces the maltose system (exogenous induction), even though only maltotriose has been identified in vitro as an inducer (O. Raibaud and E. Richet, J. Bacteriol., 169:3059-3061, 1987). Induction is dependent on MalT, the positive regulator protein of the system. In the presence of exogenous glucose, the maltose system is normally repressed because of catabolite repression and inducer exclusion brought about by the phosphotransferase-mediated vectorial phosphorylation of glucose. In contrast, the increase of free, unphosphorylated glucose in the cell induces the maltose system. A ptsG ptsM glk mutant which cannot grow on glucose can accumulate [14C]glucose via galactose permeases. In this strain, internal glucose is polymerized to maltose, maltotriose, and maltodextrins in which only the reducing glucose residue is labeled. This polymerization does not require maltose enzymes, since it still occurs in malT mutants. Formation of maltodextrins from external glucose as well as induction of the maltose system is absent in a mutant lacking phosphoglucomutase, and induction by external glucose could be regained by the addition of glucose-1-phosphate entering the cells via a constitutive glucose phosphate transport system. malQ mutants, which lack amylomaltase, are constitutive for the expression of the maltose genes. This constitutive nature is due to the formation of maltose and maltodextrins from the degradation of glycogen.


Subject(s)
Escherichia coli/metabolism , Glucose/metabolism , Glucosephosphates/metabolism , Maltose/biosynthesis , Trisaccharides/biosynthesis , Adenosine Diphosphate/metabolism , Biological Transport , Enzyme Induction , Escherichia coli/enzymology , Galactose/metabolism , Mutation , Phenotype , Phosphoglucomutase/genetics , Uridine Diphosphate/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...