Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
J Microbiol Biotechnol ; 33(6): 724-735, 2023 Jun 28.
Article in English | MEDLINE | ID: mdl-37072678

ABSTRACT

NdgR, a global regulator in soil-dwelling and antibiotic-producing Streptomyces, is known to regulate branched-chain amino acid metabolism by binding to the upstream region of synthetic genes. However, its numerous and complex roles are not yet fully understood. To more fully reveal the function of NdgR, phospholipid fatty acid (PLFA) analysis with gas chromatography-mass spectrometry (GC-MS) was used to assess the effects of an ndgR deletion mutant of Streptomyces coelicolor. The deletion of ndgR was found to decrease the levels of isoleucine- and leucine-related fatty acids but increase those of valine-related fatty acids. Furthermore, the defects in leucine and isoleucine metabolism caused by the deletion impaired the growth of Streptomyces at low temperatures. Supplementation of leucine and isoleucine, however, could complement this defect under cold shock condition. NdgR was thus shown to be involved in the control of branched-chain amino acids and consequently affected the membrane fatty acid composition in Streptomyces. While isoleucine and valine could be synthesized by the same enzymes (IlvB/N, IlvC, IlvD, and IlvE), ndgR deletion did not affect them in the same way. This suggests that NdgR is involved in the upper isoleucine and valine pathways, or that its control over them differs in some respect.


Subject(s)
Streptomyces coelicolor , Streptomyces , Streptomyces coelicolor/genetics , Streptomyces coelicolor/metabolism , Isoleucine/metabolism , Valine , Leucine , Fatty Acids/metabolism , Amino Acids, Branched-Chain/genetics , Amino Acids, Branched-Chain/metabolism , Streptomyces/metabolism
2.
mBio ; 13(2): e0042522, 2022 04 26.
Article in English | MEDLINE | ID: mdl-35357210

ABSTRACT

The bacterial response to antibiotics eliciting resistance is one of the key challenges in global health. Despite many attempts to understand intrinsic antibiotic resistance, many of the underlying mechanisms still remain elusive. In this study, we found that iron supplementation promoted antibiotic resistance in Streptomyces coelicolor. Iron-promoted resistance occurred specifically against bactericidal antibiotics, irrespective of the primary target of antibiotics. Transcriptome profiling revealed that some genes in the central metabolism and respiration were upregulated under iron-replete conditions. Iron supported the growth of S. coelicolor even under anaerobic conditions. In the presence of potassium cyanide, which reduces aerobic respiration of cells, iron still promoted respiration and antibiotic resistance. This suggests the involvement of a KCN-insensitive type of respiration in the iron effect. This phenomenon was also observed in another actinobacterium, Mycobacterium smegmatis. Taken together, these findings provide insight into a bacterial resistance strategy that mitigates the activity of bactericidal antibiotics whose efficacy accompanies oxidative damage by switching the respiration mode. IMPORTANCE A widely investigated mode of antibiotic resistance occurs via mutations and/or by horizontal acquisition of resistance genes. In addition to this acquired resistance, most bacteria exhibit intrinsic resistance as an inducible and adaptive response to different classes of antibiotics. Increasing attention has been paid recently to intrinsic resistance mechanisms because this may provide novel therapeutic targets that help rejuvenate the efficacy of the current antibiotic regimen. In this study, we demonstrate that iron promotes the intrinsic resistance of aerobic actinomycetes Streptomyces coelicolor and Mycobacterium smegmatis against bactericidal antibiotics. A surprising role of iron to increase respiration, especially in a mode of using less oxygen, appears a fitting strategy to cope with bactericidal antibiotics known to kill bacteria through oxidative damage. This provides new insights into developing antimicrobial treatments based on the availability of iron and oxygen.


Subject(s)
Actinobacteria , Streptomyces coelicolor , Actinobacteria/metabolism , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Bacteria/genetics , Drug Resistance, Microbial , Iron/metabolism , Oxygen/metabolism , Respiration , Streptomyces coelicolor/genetics , Streptomyces coelicolor/metabolism
3.
PLoS One ; 13(11): e0207278, 2018.
Article in English | MEDLINE | ID: mdl-30440014

ABSTRACT

Flavonols are a flavonoid subfamily widely distributed in plants, including several ones of great importance in human and animal diet (apple, tomato, broccoli, onion, beans, tea). These polyphenolic nutraceuticals exert potent antimicrobial (membrane potential disruptors), antioxidant (free-radical scavengers), pharmacokinetic (CYP450 modulators), anti-inflammatory (lipoxygenase inhibitors), antiangiogenic (VEGF inhibitors) and antitumor (cyclin inhibitors) activities. Biotechnological production of these nutraceuticals, for example via heterologous biosynthesis in industrial actinomycetes, is favored since in plants these polyphenols appear as inactive glycosylated derivatives, in low concentrations or as part of complex mixtures with other polyphenolic compounds. In this work, we describe the de novo biosynthesis of three important flavonols, myricetin, kaempferol and quercetin, in the industrially relevant actinomycetes Streptomyces coelicolor and S. albus. De novo biosynthesis of kaempferol, myricetin and quercetin in actinomycetes has not been described before.


Subject(s)
Dietary Supplements , Flavonoids , Kaempferols , Microorganisms, Genetically-Modified , Quercetin , Streptomyces coelicolor , Flavonoids/biosynthesis , Flavonoids/genetics , Kaempferols/biosynthesis , Kaempferols/genetics , Microorganisms, Genetically-Modified/genetics , Microorganisms, Genetically-Modified/metabolism , Quercetin/biosynthesis , Quercetin/genetics , Streptomyces coelicolor/genetics , Streptomyces coelicolor/metabolism
4.
J Bacteriol ; 199(18)2017 09 15.
Article in English | MEDLINE | ID: mdl-28461451

ABSTRACT

Two-component systems (TCSs), the predominant signal transduction pathways employed by bacteria, play important roles in physiological metabolism in Streptomyces Here, a novel TCS, GluR-GluK (encoded by SCO5778-SCO5779), which is located divergently from the gluABCD operon encoding a glutamate uptake system, was identified as being involved in glutamate sensing and uptake as well as antibiotic biosynthesis in Streptomyces coelicolor Under the condition of minimal medium (MM) supplemented with different concentrations of glutamate, deletion of the gluR-gluK operon (gluR-K) resulted in enhanced actinorhodin (ACT) but reduced undecylprodigiosin (RED) and yellow type I polyketide (yCPK) production, suggesting that GluR-GluK plays a differential role in antibiotic biosynthesis. Furthermore, we found that the response regulator GluR directly promotes the expression of gluABCD under the culture condition of MM with a high concentration of glutamate (75 mM). Using the biolayer interferometry assay, we demonstrated that glutamate acts as the direct signal of the histidine kinase GluK. It was therefore suggested that upon sensing high concentrations of glutamate, GluR-GluK would be activated and thereby facilitate glutamate uptake by increasing gluABCD expression. Finally, we demonstrated that the role of GluR-GluK in antibiotic biosynthesis is independent of its function in glutamate uptake. Considering the wide distribution of the glutamate-sensing (GluR-GluK) and uptake (GluABCD) module in actinobacteria, it could be concluded that the GluR-GluK signal transduction pathway involved in secondary metabolism and glutamate uptake should be highly conserved in this bacterial phylum.IMPORTANCE In this study, a novel two-component system (TCS), GluR-GluK, was identified to be involved in glutamate sensing and uptake as well as antibiotic biosynthesis in Streptomyces coelicolor A possible GluR-GluK working model was proposed. Upon sensing high glutamate concentrations (such as 75 mM), activated GluR-GluK could regulate both glutamate uptake and antibiotic biosynthesis. However, under a culture condition of MM supplemented with low concentrations of glutamate (such as 10 mM), although GluR-GluK is activated, its activity is sufficient only for the regulation of antibiotic biosynthesis. To the best of our knowledge, this is the first report describing a TCS signal transduction pathway for glutamate sensing and uptake in actinobacteria.


Subject(s)
Glutamic Acid/metabolism , Histidine Kinase/metabolism , Signal Transduction , Streptomyces coelicolor/metabolism , Transcription Factors/metabolism , Biological Transport , Culture Media/chemistry , Gene Deletion , Gene Expression Regulation , Histidine Kinase/genetics , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Operon , Streptomyces coelicolor/genetics , Transcription Factors/genetics
5.
Appl Microbiol Biotechnol ; 99(23): 10177-89, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26428242

ABSTRACT

The molecular mechanisms regulating tryptophan biosynthesis in actinomycetes are poorly understood; similarly, the possible roles of tryptophan in the differentiation program of microorganism life-cycle are still underexplored. To unveil the possible regulatory effect of this amino acid on gene expression, an integrated study based on quantitative teverse transcription-PCR (qRT-PCR) and proteomic approaches was performed on the actinomycete model Streptomyces coelicolor. Comparative analyses on the microorganism growth in a minimal medium with or without tryptophan supplementation showed that biosynthetic trp gene expression in S. coelicolor is not subjected to a negative regulation by the presence of the end product. Conversely, tryptophan specifically induces the transcription of trp genes present in the biosynthetic gene cluster of the calcium-dependent antibiotic (CDA), a lipopeptide containing D- and L-tryptophan residues. In addition, tryptophan stimulates the transcription of the CDA gene cluster regulator cdaR and, coherently, CDA production. Surprisingly, tryptophan also promotes the production of actinorhodin, another antibiotic that does not contain this amino acid in its structure. Combined 2D-DIGE and nano liquid chromatography electrospray linear ion trap tandem mass spectrometry (LC-ESI-LIT-MS/MS) analyses revealed that tryptophan exerts a growth-stage-dependent global effect on S. coelicolor proteome, stimulating anabolic pathways and promoting the accumulation of key factors associated with morphological and physiological differentiation at the late growth stages. Phenotypic observations by scanning electron microscopy and spore production assays demonstrated an increased sporulation in the presence of tryptophan. Transcriptional analysis of catabolic genes kynA and kynB suggested that the actinomycete also uses tryptophan as a carbon and nitrogen source. In conclusion, this study originally provides the molecular basis underlying the stimulatory effect of tryptophan on the production of antibiotics and morphological development program of this actinomycete.


Subject(s)
Gene Expression Regulation, Bacterial/drug effects , Streptomyces coelicolor/cytology , Streptomyces coelicolor/physiology , Tryptophan/metabolism , Carbon/metabolism , Chromatography, Liquid , Culture Media/chemistry , Electrophoresis, Gel, Two-Dimensional , Energy Metabolism , Gene Expression Profiling , Microscopy, Electron, Scanning , Nitrogen/metabolism , Proteome/analysis , Spectrometry, Mass, Electrospray Ionization , Spores, Bacterial/growth & development , Spores, Bacterial/ultrastructure , Streptomyces coelicolor/genetics , Streptomyces coelicolor/metabolism
6.
Appl Microbiol Biotechnol ; 99(5): 2329-37, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25547841

ABSTRACT

Cobalamin (Cbl) (synonym, vitamin B12) is the cobalt-containing cofactor produced only by some prokaryotes. Streptomyces is an effective Cbl producer. To study the role of Cbl production in Streptomyces, a knockout mutant for Cbl biosynthesis (cob) was generated in Streptomyces coelicolor A3 (2). The growth of the mutant was similar to that of the wild type in a rich medium, but inhibited in minimal medium, suggesting the involvement of Cbl in some step of primary metabolism. Methionine synthesis catalyzed by MetH, the Cbl-dependent methionine synthase, is a candidate. However, supplementing the minimal medium with methionine did not rescue the growth of the cob mutant, indicating that the availability of Cbl affects another primary function. Transcriptional analysis confirmed that the mutant induced metE encoding an alternative Cbl-independent methionine synthase, probably due to the Cbl-dependent riboswitch mechanism. The cob mutant produced low levels of pigment antibiotics and formed fewer aerial mycelium and spores in a rich medium, suggesting that a Cbl-dependent mechanism controls development. A similar developmental defect was observed for a knockout mutant for SCO4800, encoding the putative Cbl-dependent isobutyryl-CoA mutase (Icm) small subunit. Since the knockout of the Icm large subunit (SCO5415) did not affect the developmental phenotype, SCO4800 likely regulates development independently from SCO5415. Effective Cbl production is fundamental to the diverse functions underlying the complex developmental life cycle of S. coelicolor A3 (2).


Subject(s)
Gene Expression Regulation, Fungal , Streptomyces coelicolor/growth & development , Streptomyces coelicolor/metabolism , Vitamin B 12/biosynthesis , Culture Media/chemistry , Gene Expression Profiling , Gene Knockout Techniques , Streptomyces coelicolor/genetics
7.
Nucleic Acids Res ; 42(3): 2003-14, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24234448

ABSTRACT

Superoxide dismutases (SODs) are widely distributed enzymes that convert superoxides to hydrogen peroxide and molecular oxygen, using various metals as cofactors. Many actinobacteria contain genes for both Ni-containing (sodN) and Fe-containing (sodF) SODs. In Streptomyces coelicolor, expression of the sodF and sodN genes is inversely regulated by nickel-specific Nur, a Fur-family regulator. With sufficient nickel, Nur directly represses sodF transcription, while inducing sodN indirectly. Bioinformatic search revealed that a conserved 19-nt stretch upstream of sodN matches perfectly with the sodF downstream sequence. We found that the sodF gene produced a stable small-sized RNA species (s-SodF) of ∼ 90 nt that harbors the anti-sodN sequence complementary to sodN mRNA from the 5'-end up to the ribosome binding site. Absence of nearby promoters and sensitivity to 5'-phosphate-specific exonuclease indicated that the s-SodF RNA is a likely processed product of sodF mRNA. The s-SodF RNA caused a significant decrease in the half-life of the sodN mRNA. Therefore, Nur activates sodN expression through inhibiting the synthesis of sodF mRNA, from which inhibitory s-SodF RNA is generated. This reveals a novel mechanism by which antagonistic regulation of one gene is achieved by small RNA processed from the 3'UTR of another gene's mRNA.


Subject(s)
Gene Expression Regulation, Bacterial , RNA Processing, Post-Transcriptional , RNA, Small Untranslated/metabolism , Streptomyces coelicolor/genetics , Superoxide Dismutase/genetics , Transcription Factors/metabolism , 3' Untranslated Regions , Mutation , RNA Stability , RNA, Messenger/metabolism , Streptomyces coelicolor/growth & development , Streptomyces coelicolor/metabolism , Superoxide Dismutase/metabolism
8.
Mol Microbiol ; 87(1): 30-48, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23106203

ABSTRACT

The two-component system AfsQ1/Q2 of Streptomyces coelicolor was identified in our previous work as a pleiotropic regulator for antibiotic biosynthesis and morphological differentiation under the condition of a minimal medium supplemented with 75 mM glutamate. In this work, we report the dissection of the mechanism underlying the function of AfsQ1/Q2 on antibiotic production and also the identification of the AfsQ1/Q2 regulon. The results showed that AfsQ1/Q2 stimulated antibiotic ACT, RED and CDA production directly through the pathway-specific activator genes actII-ORF4, redZ and cdaR respectively. In addition, expression of sigQ that encodes a sigma factor and is divergently transcribed from afsQ1 was also subject to direct regulation by AfsQ1/Q2. The precise AfsQ1 binding sites in the upstream regions of these target genes were determined by DNase I footprinting assays coupled with site-directed DNA mutagenesis. By computational prediction and functional analysis, at least 17 new AfsQ1 targets were identified, including pstS gene encoding a high-affinity phosphate-binding protein and two developmental genes whiD, bldM. For the AfsQ1/Q2 regulon, an AfsQ1 binding motif comprising the sequence GTnAC-n(6) -GTnAC has been defined. Interestingly, we found from electrophoretic mobility shift assays and transcriptional analysis that AfsQ1/Q2 can also function as a repressor for nitrogen assimilation, and AfsQ1 can compete with GlnR for the promoter regions of glnA and nirB, suggesting the cross-regulation between AfsQ1/Q2 and GlnR in nitrogen metabolism. These findings suggested that AfsQ1/Q2 is important not only for antibiotic biosynthesis but also in maintaining the metabolic homeostasis of nutrient utilization under the stress of high concentration of glutamate in S. coelicolor.


Subject(s)
Bacterial Proteins/metabolism , Regulon/genetics , Streptomyces coelicolor/genetics , Streptomyces coelicolor/metabolism , Trans-Activators/metabolism , Anti-Bacterial Agents/biosynthesis , Bacterial Proteins/genetics , DNA Footprinting , DNA, Bacterial/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Deoxyribonuclease I/metabolism , Electrophoretic Mobility Shift Assay , Gene Expression Regulation, Bacterial , Glutamic Acid/metabolism , Mutagenesis, Site-Directed , Mutation , Nitrogen/metabolism , Promoter Regions, Genetic , Trans-Activators/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation
9.
J Bacteriol ; 194(24): 6837-46, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23065973

ABSTRACT

We show here that NdgR, a known transcriptional activator of isopropylmalate dehydratase in actinomycetes, may have other targets in the cell. An in-frame deletion mutant of ndgR showed unexpectedly poor growth in defined minimal medium even in the presence of leucine. To our surprise, it was supplementation of cysteine and methionine that corrected the growth. Based on this, we propose that NdgR induces cysteine-methionine biosynthesis. Direct involvement of NdgR in the very last steps of methionine synthesis with methionine synthase (metH) and 5,10-methylenetetrahydrofolate reductase (metF) was examined. From a pulldown assay, it was seen that NdgR was enriched from crude cell lysates with a strong affinity to metH and metF upstream sequences. Direct physical interaction of NdgR with these targets was further examined with a gel mobility shift assay. ndgR, leuC, metH, and metF were inducible in M145 cells upon nutrient downshift from rich to minimal medium but were not induced in the ndgR knockout mutant. Taking these observations together, NdgR-dependent metH-metF expression would account for the abnormal growth phenotype of the ndgR mutant although there may be additional NdgR-dependent genes in the Cys-Met metabolic pathways. As the first transcriptional factor reported for regulating Cys-Met metabolism in Streptomyces, NdgR links two disparate amino acid families, branched-chain amino acids (BCAAs) and sulfur amino acids, at the transcriptional level. Considering that Cys-Met metabolism is connected to mycothiol and one-carbon metabolism, NdgR may have broad physiological impacts.


Subject(s)
Leucine/biosynthesis , Methionine/biosynthesis , Streptomyces coelicolor/genetics , Streptomyces coelicolor/metabolism , Transcription Factors/metabolism , Transcriptional Activation , 5,10-Methylenetetrahydrofolate Reductase (FADH2)/metabolism , 5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/metabolism , Cysteine/metabolism , Gene Expression Regulation, Bacterial , Genes, Bacterial , Isomerases/genetics , Isomerases/metabolism , Streptomyces coelicolor/growth & development , Transcription Factors/genetics , Transcription, Genetic
10.
Future Microbiol ; 7(6): 705-17, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22702525

ABSTRACT

Ribosome-inactivating proteins (RIPs) are toxic due to their N-glycosidase activity catalyzing depurination at the universally conserved α-sarcin loop of the 60S ribosomal subunit. In addition, RIPs have been shown to also have other enzymatic activities, including polynucleotide:adenosine glycosidase activity. RIPs are mainly produced by different plant species, but are additionally found in a number of bacteria, fungi, algae and some mammalian tissues. This review describes the occurrence of RIPs, with special emphasis on bacterial RIPs, including the Shiga toxin and RIP in Streptomyces coelicolor recently identified in S. coelicolor. The properties of RIPs, such as enzymatic activity and targeting specificity, and how their unique biological activity could be potentially turned into medical or agricultural tools to combat tumors, viruses and fungi, are highlighted.


Subject(s)
Bacterial Proteins/toxicity , Bacterial Proteins/therapeutic use , Ribosome Inactivating Proteins/toxicity , Ribosome Inactivating Proteins/therapeutic use , Algal Proteins/toxicity , Fungal Proteins/toxicity , Humans , Plant Proteins/toxicity , Shigella/metabolism , Streptomyces coelicolor/metabolism
11.
Res Microbiol ; 163(5): 340-8, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22659143

ABSTRACT

The model actinobacterium Streptomyces coelicolor A3(2) uses nitrate and sulfate as nitrogen and sulfur sources, respectively. The final step prior to assimilation into amino acids is the 6-electron reduction of the nitrite and sulfite anions, catalyzed by siroheme-dependent nitrite (NirBD) and sulfite (SirA) reductases. There are two predicted nitrite/sulfite reductases annotated in the genome of S. coelicolor, but it is unclear which is responsible for nitrite and which for sulfite reduction. Here we demonstrate that a knock-out in the genes SCO2487 and SCO2488 encoding NirBD prevents use of nitrite as a nitrogen source, while a knock-out in SCO6102 encoding SirA prevents sulfate assimilation. Both mutations could be phenotypically complemented by supplementation of the growth medium with ammonium or casamino acids in the case of the nirBD mutants or sulfur-containing amino acids in the case of the sirA mutants. No functional redundancy between the genes was observed and we demonstrate that NirBD is exclusively required for assimilatory nitrite (it does not detoxify nitrite) and SirA exclusively for assimilatory sulfite reduction.


Subject(s)
Nitrite Reductases/genetics , Nitrites/metabolism , Oxidoreductases Acting on Sulfur Group Donors/genetics , Streptomyces coelicolor/enzymology , Streptomyces coelicolor/genetics , Sulfates/metabolism , Culture Media/chemistry , Electron Transport , Gene Knockout Techniques , Nitrite Reductases/metabolism , Oxidation-Reduction , Oxidoreductases Acting on Sulfur Group Donors/metabolism , Streptomyces coelicolor/metabolism
12.
PLoS One ; 7(3): e32697, 2012.
Article in English | MEDLINE | ID: mdl-22403700

ABSTRACT

ArgR is the regulator of arginine biosynthesis genes in Streptomyces species. Transcriptomic comparison by microarrays has been made between Streptomyces coelicolor M145 and its mutant S. coelicolor ΔargR under control, unsupplemented conditions, and in the presence of arginine. Expression of 459 genes was different in transcriptomic assays, but only 27 genes were affected by arginine supplementation. Arginine and pyrimidine biosynthesis genes were derepressed by the lack of ArgR, while no strong effect on expression resulted on arginine supplementation. Several nitrogen metabolism genes expression as glnK, glnA and glnII, were downregulated in S. coelicolor ΔargR. In addition, downregulation of genes for the yellow type I polyketide CPK antibiotic and for the antibiotic regulatory genes afsS and scbR was observed. The transcriptomic data were validated by either reverse transcription-PCR, expression of the gene-promoter coupled to the luciferase gene, proteomic or by electrophoresis mobility shift assay (EMSA) using pure Strep-tagged ArgR. Two ARG-boxes in the arginine operon genes suggest that these genes are more tightly controlled. Other genes, including genes encoding regulatory proteins, possess a DNA sequence formed by a single ARG-box which responds to ArgR, as validated by EMSA.


Subject(s)
Bacterial Proteins/metabolism , Repressor Proteins/metabolism , Streptomyces coelicolor/genetics , Streptomyces coelicolor/metabolism , Arginine/biosynthesis , Arginine/metabolism , Arginine/pharmacology , Bacterial Proteins/genetics , Base Sequence , Binding Sites , Biological Transport/genetics , DNA, Bacterial/biosynthesis , Mutation , Nitrogen/metabolism , Nucleotides/biosynthesis , Proteomics , RNA, Bacterial/biosynthesis , Repressor Proteins/genetics , Streptomyces coelicolor/drug effects , Streptomyces coelicolor/growth & development , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Transcriptome/drug effects
13.
Nucleic Acids Res ; 40(12): 5227-39, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22406834

ABSTRACT

Determining transcriptional regulator activities is a major focus of systems biology, providing key insight into regulatory mechanisms and co-regulators. For organisms such as Escherichia coli, transcriptional regulator binding site data can be integrated with expression data to infer transcriptional regulator activities. However, for most organisms there is only sparse data on their transcriptional regulators, while their associated binding motifs are largely unknown. Here, we address the challenge of inferring activities of unknown regulators by generating de novo (binding) motifs and integrating with expression data. We identify a number of key regulators active in the metabolic switch, including PhoP with its associated directed repeat PHO box, candidate motifs for two SARPs, a CRP family regulator, an iron response regulator and that for LexA. Experimental validation for some of our predictions was obtained using gel-shift assays. Our analysis is applicable to any organism for which there is a reasonable amount of complementary expression data and for which motifs (either over represented or evolutionary conserved) can be identified in the genome.


Subject(s)
Streptomyces coelicolor/genetics , Transcription Factors/metabolism , Transcriptome , Bacterial Proteins/metabolism , Binding Sites , Gene Expression Profiling , Gene Regulatory Networks , Genomics/methods , Glutamic Acid/metabolism , Nucleotide Motifs , Phosphates/metabolism , Streptomyces coelicolor/metabolism
14.
Electron. j. biotechnol ; Electron. j. biotechnol;15(1): 8-8, Jan. 2012. ilus, tab
Article in English | LILACS | ID: lil-640534

ABSTRACT

Background: The growing problem of environmental pollution caused by synthetic plastics has led to the search for alternative materials such as biodegradable plastics. Of the biopolymers presently under development, starch/natural rubber is one promising alternative. Several species of bacteria and fungi are capable of degrading natural rubber and many can degrade starch. Results: Streptomyces coelicolor CH13 was isolated from soil according to its ability to produce translucent halos on a mineral salts medium, MSM, supplemented with natural rubber and to degrade starch. Scanning electron microscope studies showed that it colonized the surfaces of strips of a new starch/natural rubber biopolymer and rubber gloves and caused degradation by forming holes, and surface degradation. Starch was completely removed and polyisoprene chains were broken down to produce aldehyde and/or carbonyl groups. After 6 weeks of cultivation with strips of the polymers in MSM, S. coelicolor CH13 reduced the weight of the starch/NR biopolymer by 92 percent and that of the rubber gloves by 14.3 percent. Conclusions: This study indicated that this bacterium causes the biodegradation of the new biopolymer and natural rubber and confirms that this new biopolymer can be degraded in the environment and would be suitable as a ‘green plastic’ derived from natural sources.


Subject(s)
Starch/metabolism , Biopolymers/metabolism , Rubber/metabolism , Streptomyces coelicolor/metabolism , Streptomyces coelicolor/chemistry , Biodegradation, Environmental , Biopolymers/chemistry , Rubber/chemistry
15.
J Bacteriol ; 190(16): 5616-23, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18556792

ABSTRACT

Secondary transporters of citrate in complex with metal ions belong to the bacterial CitMHS family, about which little is known. The transport of metal-citrate complexes in Streptomyces coelicolor has been investigated. The best cofactor for citrate uptake in Streptomyces coelicolor is Fe(3+), but uptake was also noted for Ca(2+), Pb(2+), Ba(2+), and Mn(2+). Uptake was not observed with the Mg(2+), Ni(2+), or Co(2+) cofactor. The transportation of iron- and calcium-citrate makes these systems unique among the CitMHS family members reported to date. No complementary uptake akin to that observed for the CitH (Ca(2+), Ba(2+), Sr(2+)) and CitM (Mg(2+), Ni(2+), Mn(2+), Co(2+), Zn(2+)) systems of Bacillus subtilis was noted. Competitive experiments using EGTA confirmed that metal-citrate complex formation promoted citrate uptake. Uptake of free citrate was not observed. The open reading frame postulated as being responsible for the metal-citrate transport observed in Streptomyces coelicolor was cloned and overexpressed in Escherichia coli strains with the primary Fe(3+)-citrate transport system (fecABCDE) removed. Functional expression was successful, with uptake of Ca(2+)-citrate, Fe(3+)-citrate, and Pb(2+)-citrate observed. No free-citrate transport was observed in IPTG (isopropyl-beta-d-thiogalactopyranoside)-induced or -uninduced E. coli. Metabolism of the Fe(3+)-citrate and Ca(2+)-citrate complexes, but not the Pb(2+)-citrate complex, was observed. Rationalization is based on the difference in metal-complex coordination upon binding of the metal by citrate.


Subject(s)
Carrier Proteins/metabolism , Cations, Divalent/metabolism , Citric Acid/metabolism , Metals/metabolism , Streptomyces coelicolor/metabolism , Bacillus subtilis/metabolism , Cloning, Molecular , Egtazic Acid/metabolism , Escherichia coli/genetics , Gene Expression , Hydrogen-Ion Concentration , Substrate Specificity , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL