Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Biosci Bioeng ; 123(4): 444-450, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28057466

ABSTRACT

The alaE gene in Escherichia coli encodes an l-alanine exporter that catalyzes the active export of l-alanine using proton electrochemical potential. In our previous study, alaE expression was shown to increase in the presence of l-alanyl-l-alanine (Ala-Ala). In this study, the global regulator leucine-responsive regulatory protein (Lrp) was identified as an activator of the alaE gene. A promoter less ß-galactosidase gene was fused to an alaE upstream region (240 nucleotides). Cells that were lacZ-deficient and harbored this reporter plasmid showed significant induction of ß-galactosidase activity (approximately 17-fold) in the presence of 6 mM l-alanine, l-leucine, and Ala-Ala. However, a reporter plasmid possessing a smaller alaE upstream region (180 nucleotides) yielded transformants with strikingly low enzyme activity under the same conditions. In contrast, lrp-deficient cells showed almost no ß-galactosidase induction, indicating that Lrp positively regulates alaE expression. We next performed an electrophoretic mobility shift assay (EMSA) and a DNase I footprinting assay using purified hexahistidine-tagged Lrp (Lrp-His). Consequently, we found that Lrp-His binds to the alaE upstream region spanning nucleotide -161 to -83 with a physiologically relevant affinity (apparent KD, 288.7 ± 83.8 nM). Furthermore, the binding affinity of Lrp-His toward its cis-element was increased by l-alanine and l-leucine, but not by Ala-Ala and d-alanine. Based on these results, we concluded that the gene expression of the alaE is regulated by Lrp in response to intracellular levels of l-alanine, which eventually leads to intracellular homeostasis of l-alanine concentrations.


Subject(s)
Alanine/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial , Leucine-Responsive Regulatory Protein/metabolism , Alanine/pharmacology , Amino Acid Transport Systems, Neutral/biosynthesis , Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/metabolism , Base Sequence , DNA Footprinting , Deoxyribonuclease I/metabolism , Dipeptides/metabolism , Dipeptides/pharmacology , Electrophoretic Mobility Shift Assay , Escherichia coli/drug effects , Escherichia coli Proteins/biosynthesis , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial/drug effects , Genes, Reporter/genetics , Leucine/metabolism , Leucine/pharmacology , Leucine-Responsive Regulatory Protein/deficiency , Operon/drug effects , Protein Binding/drug effects , Regulatory Sequences, Nucleic Acid/genetics , Up-Regulation/drug effects , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
2.
Microbiologyopen ; 4(4): 632-43, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26073055

ABSTRACT

We previously reported that the alaE gene of Escherichia coli encodes the l-alanine exporter AlaE. The objective of this study was to elucidate the mechanism of the AlaE exporter. The minimum inhibitory concentration of l-alanine and l-alanyl-l-alanine in alaE-deficient l-alanine-nonmetabolizing cells MLA301ΔalaE was 4- and >4000-fold lower, respectively, than in the alaE-positive parent cells MLA301, suggesting that AlaE functions as an efflux pump to avoid a toxic-level accumulation of intracellular l-alanine and its derivatives. Furthermore, the growth of the alaE-deficient mutant derived from the l-alanine-metabolizing strain was strongly inhibited in the presence of a physiological level of l-alanyl-l-alanine. Intact MLA301ΔalaE and MLA301ΔalaE/pAlaE cells producing plasmid-borne AlaE, accumulated approximately 200% and 50%, respectively, of the [(3) H]l-alanine detected in MLA301 cells, suggesting that AlaE exports l-alanine. When 200 mmol/L l-alanine-loaded inverted membrane vesicles prepared from MLA301ΔalaE/pAlaE were placed in a solution containing 200 mmol/L or 0.34 µmol/L l-alanine, energy-dependent [(3) H]l-alanine accumulation occurred under either condition. This energy-dependent uphill accumulation of [(3) H]l-alanine was strongly inhibited in the presence of carbonyl cyanide m-chlorophenylhydrazone but not by dicyclohexylcarbodiimide, suggesting that the AlaE-mediated l-alanine extrusion was driven by proton motive force. Based on these results, physiological roles of the l-alanine exporter are discussed.


Subject(s)
Alanine/metabolism , Alanine/toxicity , Amino Acid Transport Systems, Neutral/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Amino Acid Transport Systems, Neutral/deficiency , Biological Transport , Dipeptides/metabolism , Dipeptides/toxicity , Escherichia coli/genetics , Escherichia coli/growth & development , Gene Deletion , Genetic Complementation Test , Microbial Sensitivity Tests , Plasmids , Proton-Motive Force
3.
Biosci Biotechnol Biochem ; 75(5): 930-8, 2011.
Article in English | MEDLINE | ID: mdl-21597182

ABSTRACT

For Escherichia coli, it has been assumed that L-alanine is synthesized by alanine-valine transaminase (AvtA) in conjunction with an unknown alanine aminotransferase(s). We isolated alanine auxotrophs from a prototrophic double mutant deficient in AvtA and YfbQ, a novel alanine aminotransferase, by chemical mutagenesis. A shotgun cloning experiment identified two genes, uncharacterized yfdZ and serC, that complemented the alanine auxotrophy. When the yfdZ- or serC-mutation was introduced into the double mutant, one triple mutant (avtA yfbQ yfdZ) showed alanine auxotrophy, and another (avtA yfbQ serC), prototrophy. In addition, we found that four independent alanine auxotrophs possessed a point mutation in yfdZ but not in serC. We also found that yfdZ expression was induced in minimal medium. Furthermore, yfbQ-bearing plasmid conferred the ability to excrete alanine on the mutant lacking D-amino acid dehydrogenase-encoding gene, dadA. From these results, we concluded that E. coli synthesizes L-alanine by means of three aminotransferases, YfbQ, YfdZ, and AvtA.


Subject(s)
Alanine Transaminase/metabolism , Alanine/biosynthesis , Escherichia coli/genetics , Escherichia coli/metabolism , Mutation , Alanine Transaminase/genetics , Culture Media/metabolism , Escherichia coli/enzymology , Escherichia coli/isolation & purification , Gene Expression Regulation, Bacterial , Mutagenesis , Phenotype
4.
Appl Environ Microbiol ; 77(12): 4027-34, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21531828

ABSTRACT

We previously isolated a mutant hypersensitive to L-alanyl-L-alanine from a non-L-alanine-metabolizing Escherichia coli strain and found that it lacked an inducible l-alanine export system. Consequently, this mutant showed a significant accumulation of intracellular L-alanine and a reduction in the L-alanine export rate compared to the parent strain. When the mutant was used as a host to clone a gene(s) that complements the dipeptide-hypersensitive phenotype, two uncharacterized genes, ygaW and ytfF, and two characterized genes, yddG and yeaS, were identified. Overexpression of each gene in the mutant resulted in a decrease in the intracellular l-alanine level and enhancement of the L-alanine export rate in the presence of the dipeptide, suggesting that their products function as exporters of L-alanine. Since ygaW exhibited the most striking impact on both the intra- and the extracellular L-alanine levels among the four genes identified, we disrupted the ygaW gene in the non-L-alanine-metabolizing strain. The resulting isogenic mutant showed the same intra- and extracellular L-alanine levels as observed in the dipeptide-hypersensitive mutant obtained by chemical mutagenesis. When each gene was overexpressed in the wild-type strain, which does not intrinsically excrete alanine, only the ygaW gene conferred on the cells the ability to excrete alanine. In addition, expression of the ygaW gene was induced in the presence of the dipeptide. On the basis of these results, we concluded that YgaW is likely to be the physiologically most relevant exporter for L-alanine in E. coli and proposed that the gene be redesignated alaE for alanine export.


Subject(s)
Alanine/metabolism , Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Gene Deletion , Genes, Bacterial , Genetic Complementation Test
5.
FEMS Microbiol Lett ; 316(2): 83-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21208269

ABSTRACT

An Escherichia coli strain that exhibits a double auxotrophy for L-alanine and D-alanine was constructed. During growth in the presence of the dipeptide L-alanyl-L-alanine (Ala-Ala), this was fully consumed with concomitant extracellular accumulation of l-alanine in a twofold molar concentration compared with the dipeptide. This finding indicates that the strain not only can hardly degrade L-alanine but has an export system(s) for L-alanine. To obtain access to the system, we chemically mutagenized the L-alanine-nonmetabolizing strain and isolated mutants with increased Ala-Ala sensitivity. Two such mutants accumulated L-alanine up to 150-190 mM in the cytoplasm with a reduced rate of L-alanine export relative to the parent strain in the presence of Ala-Ala. Furthermore, when chloramphenicol was added together with Ala-Ala, the parent strain accumulated L-alanine in the cytoplasm to a level similar to that observed in the mutants in the absence of chloramphenicol. In contrast, the intracellular l-alanine level in the mutants did not change irrespective of chloramphenicol treatment. From these results, we conclude that E. coli has an inducible l-alanine export carrier, together with a second, as yet unidentified, mechanism of alanine export.


Subject(s)
Alanine/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Mutation , Biological Transport , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism
6.
Exp Appl Acarol ; 53(1): 71-7, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20596886

ABSTRACT

Defensins are small cysteine-rich cationic proteins found in both vertebrates and invertebrates constituting the front line of host innate immunity. To examine the importance of the tertiary structure of tick defensin in its antimicrobial activity, we synthesized two types of the peptides with tertiary structure or primary one on basis of the information of the sequence in the defensin originated from the taiga tick, Ixodes persulcatus. Chemically synthesized peptides were used to investigate the activity spectrum against Staphylococcus aureus, Borrelia garinii and flora-associated bacteria. Both synthetic peptides showed antimicrobial activity against S. aureus in short-time killing within 1 h, but they do not show the activity against B. garinii, Stenotrophomonas maltophila and Bacillus spp., which were frequently isolated from the midgut of I. persulcatus. The teriary structure brought more potent activity to S. aureus than primary one in short-time killing. We also examined its antimicrobial activity by evaluation of growth inhibition in the presence of the synthetic peptides. Minimum inhibitory concentration (MIC) was ranged from 1.2 to 5.0 µg/ml in tertiary peptide and from 10 to 40 µg/ml in primary peptide, when 10 strains of S. aureus were used. From the curve of cumulative inhibition rates, MIC50 (MIC which half of the strains showed) to S. aureus is about 1.2 µg/ml in the peptide with tertiary structure and about 10 µg/ml in the linear one. Corynebacterium renale is 10 times or more sensitive to tertiary peptide than primary one. In conclusion, the presence of 3 disulfide bridges, which stabilize the molecule and maintain the tertiary structure, is considered to have an effect on their antimicrobial activities against Gram-positive bacteria such as S. aureus.


Subject(s)
Defensins/chemistry , Ixodes/immunology , Animals , Ixodes/chemistry , Ixodes/microbiology , Microbial Sensitivity Tests , Protein Structure, Tertiary , Structure-Activity Relationship
7.
Antimicrob Agents Chemother ; 54(4): 1492-7, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20100880

ABSTRACT

Pseudomonas aeruginosa is equipped with the Sec and Tat protein secretion systems, which translocate the xenobiotic transporter MexAB-OprM and the pathogenic factor phospholipase C (PlcH), respectively. When the signal sequence of MexA was replaced with that of PlcH, the hybrid protein was successfully expressed and recovered from the periplasmic fraction, suggesting that the hybrid protein had been translocated across the inner membrane. MexA-deficient cells harboring the plasmid carrying the plcH-mexA fusion gene showed antibiotic resistance comparable to that of the wild-type cells. This result suggested that MexA secreted via the Tat machinery was properly assembled and functioned as a subunit of the MexAB-OprM efflux pump. A mutation was introduced into the chromosomal tatC gene encoding an inner membrane component of the Tat protein secretion machinery in mexA-deficient cells, and they were transformed with the plasmid carrying the plcH-mexA fusion gene. The transformants showed antibiotic susceptibility comparable to that of mexA-deficient cells, indicating that the hybrid protein was not transported to the periplasm. Whole-cell lysate of the mexA-tatC double mutant harboring the plcH-mexA plasmid produced mainly unprocessed PlcH-MexA. The periplasmic fraction showed no detectable anti-MexA antibody-reactive material. On the basis of these results, we concluded that MexA could be translocated across the inner membrane through the Tat pathway and assembled with its cognate partners, MexB and OprM, and that this complex machinery was fully functional. This hybrid protein translocation system has the potential to be a powerful screening tool for antimicrobial agents targeting the Tat system, which is not present in mammalian cells.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/metabolism , Membrane Transport Proteins/metabolism , Pseudomonas aeruginosa/metabolism , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/genetics , Base Sequence , DNA Primers/genetics , DNA, Bacterial/genetics , Genes, Bacterial , Membrane Transport Proteins/genetics , Microbial Sensitivity Tests , Mutation , Plasmids/genetics , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transferases (Other Substituted Phosphate Groups)/genetics , Transferases (Other Substituted Phosphate Groups)/metabolism , Xenobiotics/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...