Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Microorganisms ; 12(2)2024 Feb 19.
Article in English | MEDLINE | ID: mdl-38399816

ABSTRACT

Antimicrobial peptides (AMPs) are present in a wide range of plants, animals, and microorganisms. Since AMPs are characterized by their effectiveness against emergent antibiotic-resistant bacteria, they are attracting attention as next-generation antimicrobial compounds that could solve the problem of drug-resistant bacteria. Persulcatusin (IP), an antibacterial peptide derived from the hard tick Ixodes persulcatus, shows high antibacterial activity against various Gram- positive bacteria as well as multidrug-resistant bacteria. However, reports on the antibacterial action and resistance mechanisms of IP are scarce. In this study, we spontaneously generated mutants showing increased a minimum inhibitory concentration (MIC) of IP and analyzed their cross-resistance to other AMPs and antibiotics. We also used fluorescent probes to investigate the target of IP activity by evaluating IP-induced damage to the bacterial cytoplasmic membrane. Our findings suggest that the antimicrobial activity of IP on bacterial cytoplasmic membranes occurs via a mechanism of action different from that of known AMPs. Furthermore, we screened for mutants with high susceptibility to IP using a transposon mutant library and identified 16 genes involved in IP resistance. Our results indicate that IP, like other AMPs, depolarizes the bacterial cytoplasmic membrane, but it may also alter membrane structure and inhibit cell-wall synthesis.

2.
Int J Mol Sci ; 24(12)2023 Jun 16.
Article in English | MEDLINE | ID: mdl-37373388

ABSTRACT

d-amino acids have recently been found to be present in the extracellular milieu at millimolar levels and are therefore assumed to play a physiological function. However, the pathway (or potential pathways) by which these d-amino acids are secreted remains unknown. Recently, Escherichia coli has been found to possess one or more energy-dependent d-alanine export systems. To gain insight into these systems, we developed a novel screening system in which cells expressing a putative d-alanine exporter could support the growth of d-alanine auxotrophs in the presence of l-alanyl-l-alanine. In the initial screening, five d-alanine exporter candidates, AlaE, YmcD, YciC, YraM, and YidH, were identified. Transport assays of radiolabeled d-alanine in cells expressing these candidates indicated that YciC and AlaE resulted in lower intracellular levels of d-alanine. Further detailed transport assays of AlaE in intact cells showed that it exports d-alanine in an expression-dependent manner. In addition, the growth constraints on cells in the presence of 90 mM d-alanine were mitigated by the overexpression of AlaE, implying that AlaE could export free d-alanine in addition to l-alanine under conditions in which intracellular d/l-alanine levels are raised. This study also shows, for the first time, that YciC could function as a d-alanine exporter in intact cells.


Subject(s)
Amino Acid Transport Systems, Neutral , Escherichia coli Proteins , Escherichia coli , Alanine/metabolism , Escherichia coli Proteins/metabolism , Amino Acids/metabolism , Biological Transport , Amino Acid Transport Systems, Neutral/metabolism
3.
J Gen Appl Microbiol ; 69(3): 142-149, 2023 Dec 05.
Article in English | MEDLINE | ID: mdl-36567121

ABSTRACT

In the fermentative production of compounds by using microorganisms, control of the transporter activity responsible for substrate uptake and product efflux, in addition to intracellular metabolic modification, is important from a productivity perspective. However, there has been little progress in analyses of the functions of microbial membrane transporters, and because of the difficulty in finding transporters that transport target compounds, only a few transporters have been put to practical use. Here, we constructed a Corynebacterium glutamicum-derived transporter expression library (CgTP-Express library) with the fusion partner gene mstX and used a peptide-feeding method with the dipeptide L-Ala-L-Ala to search for alanine exporters in the library. Among 39 genes in the library, five candidate alanine exporters (NCgl2533, NCgl2683, NCgl0986, NCgl0453, and NCgl0929) were found; expression of NCgl2533 increased the alanine concentration in cell culture. The CgTP-Express library was thus effective for finding a new transporter candidate.


Subject(s)
Corynebacterium glutamicum , Membrane Transport Proteins , Fermentation , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Corynebacterium glutamicum/genetics , Corynebacterium glutamicum/metabolism , Alanine/genetics , Alanine/metabolism , Biological Transport , Metabolic Engineering/methods
4.
Microorganisms ; 10(11)2022 Oct 26.
Article in English | MEDLINE | ID: mdl-36363708

ABSTRACT

Staphylococcus aureus is one of the most important pathogens in humans as well as in livestock. Particularly, bovine mastitis caused by S. aureus is a serious issue in dairy farms due to disease recurrence. Here, cases of S. aureus-mediated intramammary infection occurring in the Miyagi Prefecture in Japan were monitored from May 2015 to August 2019; a total of 59 strains (49 from bovine milk and 10 from bulk milk) were obtained from 15 dairy farms and analyzed via sequence-based typing methods and antibiotic susceptibility tests. Two pairs of isolates were determined as recurrence cases from the same cows in distinct farms. The sequence type (ST), spa type, and coa type of each pair were the same: one pair showed ST705, t529, and VIb and the other showed ST352, t267, and VIc. In addition, the possession of toxin genes analyzed of each pair was exactly the same. Furthermore, seven oxacillin-sensitive clonal complex 398 isolates were obtained from a single farm. This is the first confirmed case of a Methicillin-Sensitive SA (MSSA) ST398 strain isolated from mastitis-containing cows in Japan. Our findings suggest that nationwide surveillance of the distribution of ST398 strains in dairy farms is important for managing human and animal health.

5.
Microbiology (Reading) ; 168(3)2022 03.
Article in English | MEDLINE | ID: mdl-35275050

ABSTRACT

AlaE is the smallest amino acid exporter identified in Escherichia coli. It exports l-alanine using the proton motive force and plays a pivotal role in maintaining intracellular l-alanine homeostasis by acting as a safety valve. However, our understanding of the molecular mechanisms of substrate export by AlaE is still limited because structural information is lacking. Due to its small size (149 amino acid residues), it has been speculated that AlaE functions by forming an oligomer. In this study, we performed chemical cross-linking and pull-down assays and showed that AlaE indeed generates homo-oligomers as a functional unit. Previous random mutagenesis experiments identified three loss-of-function AlaE point mutations in the predicted transmembrane helix 4 (TM4) region, two of which are present in the GxxxG motif. When alanine-scanning mutagenesis was applied to the TM4 region, the AlaE derivatives that had amino acid substitutions around the GxxxG motif showed low l-alanine export activities, indicating that the GxxxG motif in TM4 plays an important role in substrate export. However, these AlaE variants with low activity could still form oligomers. We therefore concluded that AlaE forms homo-oligomers and that the GxxxG motif in the TM4 region plays an essential role in AlaE activity but is not involved in AlaE oligomer formation.


Subject(s)
Amino Acid Transport Systems, Neutral , Escherichia coli Proteins , Alanine/metabolism , Amino Acid Motifs , Amino Acid Substitution , Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/metabolism , Biological Transport/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism
6.
Microorganisms ; 9(3)2021 Feb 25.
Article in English | MEDLINE | ID: mdl-33668720

ABSTRACT

In Escherichia coli, L-alanine is synthesized by three isozymes: YfbQ, YfdZ, and AvtA. When an E. coli L-alanine auxotrophic isogenic mutant lacking the three isozymes was grown on L-alanine-deficient minimal agar medium, L-alanine prototrophic mutants emerged considerably more frequently than by spontaneous mutation; the emergence frequency increased over time, and, in an L-alanine-supplemented minimal medium, correlated inversely with L-alanine concentration, indicating that the mutants were derived through stress-induced mutagenesis. Whole-genome analysis of 40 independent L-alanine prototrophic mutants identified 16 and 18 clones harboring point mutation(s) in pyruvate dehydrogenase complex and phosphotransacetylase-acetate kinase pathway, which respectively produce acetyl coenzyme A and acetate from pyruvate. When two point mutations identified in L-alanine prototrophic mutants, in pta (D656A) and aceE (G147D), were individually introduced into the original L-alanine auxotroph, the isogenic mutants exhibited almost identical growth recovery as the respective cognate mutants. Each original- and isogenic-clone pair carrying the pta or aceE mutation showed extremely low phosphotransacetylase or pyruvate dehydrogenase activity, respectively. Lastly, extracellularly-added pyruvate, which dose-dependently supported L-alanine auxotroph growth, relieved the L-alanine starvation stress, preventing the emergence of L-alanine prototrophic mutants. Thus, L-alanine starvation-provoked stress-induced mutagenesis in the L-alanine auxotroph could lead to intracellular pyruvate increase, which eventually induces L-alanine prototrophy.

7.
Int J Mol Sci ; 20(19)2019 Oct 07.
Article in English | MEDLINE | ID: mdl-31591285

ABSTRACT

The intracellular level of amino acids is determined by the balance between their anabolic and catabolic pathways. L-alanine is anabolized by three L-alanine synthesizing enzymes and catabolized by two racemases and D-amino acid dehydrogenase (DadA). In addition, its level is regulated by L-alanine movement across the inner membrane. We identified the novel gene alaE, encoding an L-alanine exporter. To elucidate the physiological function of L-Alanine exporter, AlaE, we determined the susceptibility of alaE-, dadA-, and alaE/dadA-deficient mutants, derived from the wild-type strain MG1655, to L-alanyl-L-alanine (Ala-Ala), which shows toxicity to the L-alanine-nonmetabolizing variant lacking alaE. The dadA-deficient mutant has a similar minimum inhibitory concentration (MIC) (>1.25 mg/mL) to that observed in MG1655. However, alaE- and alaE/dadA-deficient mutants had MICs of 0.04 and 0.0025 mg/mL, respectively. The results suggested that the efficacy of AlaE to relieve stress caused by toxic intracellular accumulation of L-alanine was higher than that of DadA. Consistent with this, the intracellular level of alanine in the alaE-mutant was much higher than that in MG1655 and the dadA-mutant. We, therefore, conclude that AlaE functions as a 'safety-valve' to prevent the toxic level accumulation of intracellular L-alanine under a peptide-rich environment, such as within the animal intestine.


Subject(s)
Alanine/metabolism , Amino Acid Transport Systems, Neutral/metabolism , D-Amino-Acid Oxidase/genetics , Escherichia coli Proteins/metabolism , Escherichia coli/growth & development , Alanine/toxicity , Amino Acid Transport Systems, Neutral/genetics , Biological Transport , D-Amino-Acid Oxidase/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Microbial Sensitivity Tests , Microbial Viability , Mutation , Stress, Physiological
8.
Vet Res ; 49(1): 22, 2018 02 26.
Article in English | MEDLINE | ID: mdl-29482613

ABSTRACT

Staphylococcus aureus is a major pathogen that causes subclinical mastitis associated with huge economic losses to the dairy industry. A few vaccines for bovine mastitis are available, and they are expected to induce the production of S. aureus-specific antibodies that prevent bacterial adherence to host cells or promote opsonization by phagocytes. However, the efficacy of such vaccines are still under debate; therefore, further research focusing on improving the current vaccines by seeking additional mechanisms of action is required to reduce economic losses due to mastitis in the dairy industry. Here, we generated S. aureus-specific bovine IgG antibodies (anti-S. aureus) that directly inhibited bacterial growth in vitro. Inhibition depended on specificity for anti-S. aureus, not the interaction between Protein A and the fragment crystallizable region of the IgG antibodies or bacterial agglutination. An in vitro culture study using S. aureus strain JE2 and its deletion mutant JE2ΔSrtA, which lacks the gene encoding sortase A, revealed that the effect of anti-S. aureus was sortase-A-independent. Sortase A is involved in the synthesis of cell-wall-associated proteins. Thus, other surface molecules, such as membrane proteins, cell surface polysaccharides, or both, may trigger the inhibition of bacterial growth by anti-S. aureus. Together, our findings contribute insights into developing new strategies to further improve the available mastitis vaccine by designing a novel antigen on the surface of S. aureus to induce inhibitory signals that prevent bacterial growth.


Subject(s)
Antibodies, Bacterial/metabolism , Cattle Diseases/immunology , Immunoglobulin G/metabolism , Staphylococcal Infections/veterinary , Staphylococcus aureus/immunology , Animals , Cattle , Male , Staphylococcal Infections/immunology , Staphylococcus aureus/growth & development
9.
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
10.
Arch Microbiol ; 199(1): 105-114, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27572251

ABSTRACT

The Escherichia coli alaE gene encodes the L-alanine exporter, AlaE, that catalyzes active export of L-alanine using proton electrochemical potential. The transporter comprises only 149 amino acid residues and four predicted transmembrane domains (TMs), which contain three charged amino acid residues. The AlaE-deficient L-alanine non-metabolizing cells (ΔalaE cells) appeared hypersusceptible to L-alanyl-L-alanine showing a minimum inhibitory concentration (MIC) of 2.5 µg/ml for the dipeptide due to a toxic accumulation of L-alanine. To elucidate the mechanism by which AlaE exports L-alanine, we replaced charged amino acid residues in the TMs, glutamic acid-30 (TM-I), arginine-45 (TM-II), and aspartic acid-84 (TM-III) with their respective charge-conserved amino acid or a net neutral cysteine. The ΔalaE cells producing R45K or R45C appeared hypersusceptible to the dipeptide, indicating that arginine-45 is essential for AlaE activity. MIC of the dipeptide in the ΔalaE cells expressing E30D and E30C was 156 µg/ml and >10,000 µg/ml, respectively, thereby suggesting that a negative charge at this position is not essential. The ΔalaE cells expressing D84E or D84C showed an MIC >10,000 and 78 µg/ml, respectively, implying that a negative charge is required at this position. These results were generally consistent with that of the L-alanine accumulation experiments in intact cells. We therefore concluded that charged amino acid residues (R45 and D84) in the AlaE transmembrane domain play a pivotal role in L-alanine export. Replacement of three cysteine residues at C22, C28 (both in TM-I), and C135 (C-terminal region) with alanine showed only a marginal effect on L-alanine export.


Subject(s)
Alanine/metabolism , Amino Acid Transport Systems, Neutral/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli/metabolism , Amino Acid Motifs , Amino Acid Substitution , Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/metabolism , Arginine/metabolism , Aspartic Acid/metabolism , Biological Transport , Cysteine/metabolism , Escherichia coli/chemistry , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mutagenesis, Site-Directed , Protein Domains
11.
Anim Sci J ; 88(7): 1027-1033, 2017 Jul.
Article in English | MEDLINE | ID: mdl-27878891

ABSTRACT

Streptococcus bovis, an etiologic agent of rumen acidosis in cattle, is a rumen bacterium that can grow in a chemically defined medium containing ammonia as a sole source of nitrogen. To understand its ability to assimilate inorganic ammonia, we focused on the function of glutamate dehydrogenase. In order to identify the gene encoding this enzyme, we first amplified an internal region of the gene by using degenerate primers corresponding to hexameric family I and NAD(P)+ binding motifs. Subsequently, inverse PCR was used to identify the whole gene, comprising an open reading frame of 1350 bp that encodes 449 amino acid residues that appear to have the substrate binding site of glutamate dehydrogenase observed in other organisms. Upon introduction of a recombinant plasmid harboring the gene into an Escherichia coli glutamate auxotroph lacking glutamate dehydrogenase and glutamate synthase, the transformants gained the ability to grow on minimal medium without glutamate supplementation. When cell extracts of the transformant were resolved by blue native polyacrylamide gel electrophoresis followed by activity staining, a single protein band appeared that corresponded to the size of S. bovis glutamate dehydrogenase. Based on these results, we concluded that the gene obtained encodes glutamate dehydrogenase in S. bovis.


Subject(s)
Cloning, Molecular , Glutamate Dehydrogenase/genetics , Rumen/microbiology , Streptococcus bovis/enzymology , Streptococcus bovis/genetics , Acidosis/microbiology , Acidosis/veterinary , Ammonia/metabolism , Animals , Base Sequence , Cattle , Cattle Diseases/microbiology , Glutamate Dehydrogenase/metabolism , Polymerase Chain Reaction , Sequence Analysis/methods , Stomach Diseases/microbiology , Stomach Diseases/veterinary , Streptococcus bovis/metabolism , Streptococcus bovis/pathogenicity
12.
J Vet Med Sci ; 78(9): 1439-1445, 2016 Oct 01.
Article in English | MEDLINE | ID: mdl-27246281

ABSTRACT

Epigallocatechin gallate (EGCG) is the major polyphenolic compound of green tea. Polyphenolic compounds were extracted from the leaf of Camellia sinensis (Japanese green tea), and the minimum inhibitory concentration against canine oral bacteria was measured. Subsequently, we investigated the inhibitory effects of polyphenolic compounds and EGCG on the growth of canine oral bacteria. EGCG showed antimicrobial activity against a model bacterium, Streptococcus mutans. Our results indicate that EGCG can inhibit the growth and biofilm formation of S. mutans and that EGCG does not interact with streptococcal lipoteichoic acid (LTA). Furthermore, our findings suggest that EGCG interacts with other component(s) of the bacterial membrane aside from streptococcal LTA to inhibit biofilm formation and damage biofilms.


Subject(s)
Anti-Bacterial Agents/pharmacology , Camellia sinensis/chemistry , Catechin/pharmacology , Microbiota/drug effects , Mouth/microbiology , Plant Leaves/chemistry , Tea , Animals , Biofilms/drug effects , Catechin/analogs & derivatives , Dogs , Dose-Response Relationship, Drug , Microbial Sensitivity Tests , Streptococcus mutans/drug effects , Tea/chemistry
13.
Microbiology (Reading) ; 162(7): 1243-1252, 2016 07.
Article in English | MEDLINE | ID: mdl-27166225

ABSTRACT

Escherichia coli has an l-alanine export system that protects the cells from toxic accumulation of intracellular l-alanine in the presence of l-alanyl-l-alanine (l-Ala-l-Ala). When a DadA-deficient strain was incubated with 6.0 mM l-Ala-l-Ala, we detected l-alanine and d-alanine using high-performance liquid chromatography (HPLC) analysis at a level of 7.0 mM and 3.0 mM, respectively, after 48 h incubation. Treatment of the culture supernatant with d-amino acid oxidase resulted in the disappearance of a signal corresponding to d-alanine. Additionally, the culture supernatant enabled a d-alanine auxotroph to grow without d-alanine supplementation, confirming that the signal detected by HPLC was authentic d-alanine. Upon introduction of an expression vector harbouring the alanine racemase genes, alr or dadX, the extracellular level of d-alanine increased to 11.5 mM and 8.5 mM, respectively, under similar conditions, suggesting that increased metabolic flow from l-alanine to d-alanine enhanced d-alanine secretion. When high-density DadA-deficient cells preloaded with l-Ala-l-Ala were treated with 20 µM carbonyl cyanide m-chlorophenyl hydrazone (CCCP), secretion of both l-alanine and d-alanine was enhanced ~twofold compared with that in cells without CCCP treatment. In contrast, the ATPase inhibitor dicyclohexylcarbodiimide did not exert such an effect on the l-alanine and d-alanine secretion. Furthermore, inverted membrane vesicles prepared from DadA-deficient cells lacking the l-alanine exporter AlaE accumulated [3H]D-alanine in an energy-dependent manner. This energy-dependent accumulation of [3H]D-alanine was strongly inhibited by CCCP. These results indicate that E. coli has a transport system(s) that exports d-alanine and that this function is most likely modulated by proton electrochemical potential.


Subject(s)
Alanine/metabolism , Biological Transport/physiology , Dipeptides/metabolism , Escherichia coli/metabolism , Adenosine Triphosphatases/antagonists & inhibitors , Alanine/chemistry , Alanine Racemase/genetics , Biological Transport/genetics , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Chromatography, High Pressure Liquid , D-Amino-Acid Oxidase/pharmacology , Dicyclohexylcarbodiimide/pharmacology , Escherichia coli/genetics
14.
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
15.
Molecules ; 20(5): 7790-806, 2015 Apr 29.
Article in English | MEDLINE | ID: mdl-25939068

ABSTRACT

The iron acquisition systems in Pseudomonas aeruginosa are inducible in response to low-iron conditions and important for growth of this organism under iron limitation. OprM is the essential outer membrane subunit of the MexAB-OprM xenobiotic efflux pump. We designed and constructed a new model antimicrobial screening system targeting both the iron-uptake system and xenobiotic efflux pumps. The oprM gene was placed immediately downstream of the ferri-pyoverdine receptor gene, fpvA, in the host lacking chromosomal oprM and the expression of oprM was monitored by an antibiotic susceptibility test under iron depleted and replete conditions. The recombinant cells showed wild-type susceptibility to pump substrate antibiotics, e.g., aztreonam, under iron limitation and became supersusceptible to them under iron repletion, suggesting that expression of oprM is under control of the iron acquisition system. Upon screening of a chemical library comprising 2952 compounds using this strain, a compound-ethyl 2-(1-acetylpiperidine-4-carboxamido)-4,5,6,7-tetrahydrobenzo[b]thiophene-3-carboxylate-was found to enhance the efficacy of aztreonam under iron limitation, suggesting that the compound inhibits either the iron acquisition system or the MexAB-OprM efflux pump. This compound was subsequently found to inhibit the growth of wild-type cells in the presence of sublethal amounts of aztreonam, regardless of the presence or absence of dipyridyl, an iron-chelator. The compound was eventually identified to block the function of the MexAB-OprM efflux pump, showing the validity of this new method.


Subject(s)
Bacterial Outer Membrane Proteins/genetics , Drug Resistance, Multiple, Bacterial/genetics , Iron Chelating Agents/pharmacology , Membrane Transport Proteins/genetics , Oligopeptides/metabolism , Pseudomonas aeruginosa/drug effects , Anti-Bacterial Agents/pharmacology , Aztreonam/pharmacology , Biological Transport/genetics , Chloramphenicol/pharmacology , Escherichia coli/drug effects , Gentamicins/pharmacology , Iron/metabolism , Membrane Transport Proteins/metabolism , Microbial Sensitivity Tests , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/metabolism
16.
Anim Sci J ; 85(3): 342-6, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23981031

ABSTRACT

The cathelicidin family is one of the several families of antimicrobial peptides (AMPs). A bovine myeloid antimicrobial peptide (BMAP-28) belongs to this family. Recently, the emergence of drug-resistant bacteria such as methicillin-resistant Staphylococcus aureus (MRSA) has become a big problem. AMPs are expected to be leading compounds of new antibiotics against drug-resistant bacteria. In this study, we focused on the activity of BMAP-28 against bacterial cell surfaces. First, we observed morphological change of MRSA caused by BMAP-28 using a scanning probe microscope. We also studied activities of BMAP-28 against adherence of S. aureus to fibronectin, collagen type I, collagen type IV. We confirmed whether BMAP-28 can bind to lipoteichoic acid (LTA) of S. aureus. BMAP-28 was indicated as damaging the cell surface of MRSA. In a particular range of concentrations, BMAP-28 promoted adherence of S. aureus against fibronectin and collagens. It was revealed that BMAP-28 and LTA of S. aureus bound with each other. Our study showed the potential of BMAP-28 which can damage MRSA and interact with LTA of S. aureus but promote its adherence in some concentrations. This study provides new points of which to take notice when we use AMPs as medicines.


Subject(s)
Bacterial Adhesion/drug effects , Methicillin-Resistant Staphylococcus aureus/drug effects , Proteins/pharmacology , Methicillin-Resistant Staphylococcus aureus/cytology , Microscopy, Electron, Scanning
17.
Anim Sci J ; 85(2): 174-9, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23905845

ABSTRACT

A bovine myeloid antimicrobial peptide antimicrobial peptide (BMAP-28) is a member of the cathelicidin family and acts as a component of innate immunity. There are few reports of susceptibility difference of methicillin-resistant Staphylococcus aureus (MRSA) and susceptible strains (MSSA) against BMAP-28. This study aims to clarify how a few amino acid substitutions of BMAP-28 are related to its antimicrobial activity using four analog peptides of BMAP-28. We also compared cellular fatty acid components of MSSA and MRSA using gas chromatography. We found that a few amino acid substitutions of BMAP-28 do not change antimicrobial activity. It was also revealed that the percentage of cis-11-eicosenoic acid in total detected fatty acids of MRSA was significantly higher than that of MSSA. In addition, the percentage of palmitic acid in total detected fatty acids of MRSA tended to be lower than that of MSSA. Our results will provide new information to deal with the question of differences in bacterial susceptibility against BMAP-28.


Subject(s)
Anti-Bacterial Agents/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Proteins/pharmacology , Staphylococcus aureus/drug effects , Amino Acid Substitution , Anti-Bacterial Agents/chemistry , Chromatography, Gas , Drug Resistance, Bacterial , Fatty Acids/metabolism , Fatty Acids, Monounsaturated/metabolism , Methicillin-Resistant Staphylococcus aureus/metabolism , Palmitic Acid/metabolism , Proteins/chemistry , Staphylococcus aureus/metabolism , Structure-Activity Relationship
18.
Curr Microbiol ; 66(4): 359-64, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23224413

ABSTRACT

Leptospires are a group of bacteria with a unique ultrastructure and a fascinating swimming behavior that cause a number of emerging and re-emerging diseases worldwide called leptospirosis. The unusual form of motility is thought to play a critical role in the infection process. However, the inhibition mechanism of antiserum on the motility of Leptospira to attenuate the infection efficiency is unknown. In this study, effect of antiserum on motility was quantitatively investigated by swimming speed. Relatively low concentration of antiserum was found to inhibit leptospiral motility, suggesting that the basic immunization can affect the infection efficiency. Recovery of motility a few hours later after the addition of antiserum was observed. This raises a hypothesis that Leptospira carries surface molecules bound with antibodies toward the cell end to escape and recovers the motility.


Subject(s)
Antibodies, Bacterial/immunology , Leptospira/immunology , Leptospira/physiology , Locomotion/drug effects , Time Factors
19.
Anim Sci J ; 83(6): 482-6, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22694332

ABSTRACT

A bovine myeloid antimicrobial peptide (BMAP-28) is a member of the cathelicidin family which is included in the innate immune system of mammals. Recently, there have been many studies about antimicrobial peptides. This study aims to clarify whether BMAP-28 has bactericidal activity against methicillin-resistant Staphylococcus aureus (MRSA) and compares its activity against methicillin-susceptible S. aureus (MSSA) and MRSA. We found that the peptide was effective in killing MRSA (minimal inhibitory concentration (MIC) range; 5-20 µg/mL). It was also revealed that MSSA (MIC range; 1.25-20 µg/mL) had two levels of susceptibility to BMAP-28. We also examined the effect of BMAP-28 on bacterial shape to visually show its activity. After exposure to the peptide, both MSSA and MRSA cells showed the morphological changes on their surfaces. Our results indicate that BMAP-28 is a promising candidate for medicine against drug-resistant bacteria.


Subject(s)
Methicillin Resistance/drug effects , Methicillin-Resistant Staphylococcus aureus/drug effects , Proteins/pharmacology , Staphylococcus aureus/drug effects , Animals , Cattle , Dose-Response Relationship, Drug , Immunity, Innate/genetics
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...