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










Publication year range
1.
Nucleic Acids Res ; 51(19): e98, 2023 10 27.
Article in English | MEDLINE | ID: mdl-37718823

ABSTRACT

Modular cloning has become a benchmark technology in synthetic biology. However, a notable disparity exists between its remarkable development and the need for standardization to facilitate seamless interoperability among systems. The field is thus impeded by an overwhelming proliferation of organism-specific systems that frequently lack compatibility. To overcome these issues, we present Golden Standard (GS), a Type IIS assembly method underpinned by the Standard European Vector Architecture. GS unlocks modular cloning applications for most bacteria, and delivers combinatorial multi-part assembly to create genetic circuits of up to twenty transcription units (TUs). Reliance on MoClo syntax renders GS fully compatible with many existing tools and it sets the path towards efficient reusability of available part libraries and assembled TUs. GS was validated in terms of DNA assembly, portability, interoperability and phenotype engineering in α-, ß-, γ- and δ-proteobacteria. Furthermore, we provide a computational pipeline for parts characterization that was used to assess the performance of GS parts. To promote community-driven development of GS, we provide a dedicated web-portal including a repository of parts, vectors, and Wizard and Setup tools that guide users in designing constructs. Overall, GS establishes an open, standardized framework propelling the progress of synthetic biology as a whole.


Subject(s)
Genetic Engineering , Proteobacteria , Cloning, Molecular , Genetic Engineering/methods , Genetic Vectors , Proteobacteria/genetics , Synthetic Biology/methods , DNA, Bacterial/genetics
2.
Front Bioeng Biotechnol ; 11: 1176445, 2023.
Article in English | MEDLINE | ID: mdl-37152640

ABSTRACT

Replacing traditional substrates in industrial bioprocesses to advance the sustainable production of chemicals is an urgent need in the context of the circular economy. However, since the limited degradability of non-conventional carbon sources often returns lower yields, effective exploitation of such substrates requires a multi-layer optimization which includes not only the provision of a suitable feedstock but the use of highly robust and metabolically versatile microbial biocatalysts. We tackled this challenge by means of systems metabolic engineering and validated Escherichia coli W as a promising cell factory for the production of the key building block chemical 2-ketoisovalerate (2-KIV) using whey as carbon source, a widely available and low-cost agro-industrial waste. First, we assessed the growth performance of Escherichia coli W on mono and disaccharides and demonstrated that using whey as carbon source enhances it significantly. Second, we searched the available literature and used metabolic modeling approaches to scrutinize the metabolic space of E. coli and explore its potential for overproduction of 2-KIV identifying as basic strategies the block of pyruvate depletion and the modulation of NAD/NADP ratio. We then used our model predictions to construct a suitable microbial chassis capable of overproducing 2-KIV with minimal genetic perturbations, i.e., deleting the pyruvate dehydrogenase and malate dehydrogenase. Finally, we used modular cloning to construct a synthetic 2-KIV pathway that was not sensitive to negative feedback, which effectively resulted in a rerouting of pyruvate towards 2-KIV. The resulting strain shows titers of up to 3.22 ± 0.07 g/L of 2-KIV and 1.40 ± 0.04 g/L of L-valine in 24 h using whey in batch cultures. Additionally, we obtained yields of up to 0.81 g 2-KIV/g substrate. The optimal microbial chassis we present here has minimal genetic modifications and is free of nutritional autotrophies to deliver high 2-KIV production rates using whey as a non-conventional substrate.

3.
Int J Mol Sci ; 24(8)2023 Apr 11.
Article in English | MEDLINE | ID: mdl-37108252

ABSTRACT

Bacillus subtilis is an effective workhorse for the production of many industrial products. The high interest aroused by B. subtilis has guided a large metabolic modeling effort of this species. Genome-scale metabolic models (GEMs) are powerful tools for predicting the metabolic capabilities of a given organism. However, high-quality GEMs are required in order to provide accurate predictions. In this work, we construct a high-quality, mostly manually curated genome-scale model for B. subtilis (iBB1018). The model was validated by means of growth performance and carbon flux distribution and provided significantly more accurate predictions than previous models. iBB1018 was able to predict carbon source utilization with great accuracy while identifying up to 28 metabolites as potential novel carbon sources. The constructed model was further used as a tool for the construction of the panphenome of B. subtilis as a species, by means of multistrain genome-scale reconstruction. The panphenome space was defined in the context of 183 GEMs representative of 183 B. subtilis strains and the array of carbon sources sustaining growth. Our analysis highlights the large metabolic versatility of the species and the important role of the accessory metabolism as a driver of the panphenome, at a species level.


Subject(s)
Bacillus subtilis , Metabolic Networks and Pathways , Bacillus subtilis/genetics , Bacillus subtilis/metabolism , Metabolic Networks and Pathways/genetics , Genome , Carbon/metabolism
4.
Nucleic Acids Res ; 51(D1): D1558-D1567, 2023 01 06.
Article in English | MEDLINE | ID: mdl-36420904

ABSTRACT

The SEVA platform (https://seva-plasmids.com) was launched one decade ago, both as a database (DB) and as a physical repository of plasmid vectors for genetic analysis and engineering of Gram-negative bacteria with a structure and nomenclature that follows a strict, fixed architecture of functional DNA segments. While the current update keeps the basic features of earlier versions, the platform has been upgraded not only with many more ready-to-use plasmids but also with features that expand the range of target species, harmonize DNA assembly methods and enable new applications. In particular, SEVA 4.0 includes (i) a sub-collection of plasmids for easing the composition of multiple DNA segments with MoClo/Golden Gate technology, (ii) vectors for Gram-positive bacteria and yeast and [iii] off-the-shelf constructs with built-in functionalities. A growing collection of plasmids that capture part of the standard-but not its entirety-has been compiled also into the DB and repository as a separate corpus (SEVAsib) because of its value as a resource for constructing and deploying phenotypes of interest. Maintenance and curation of the DB were accompanied by dedicated diffusion and communication channels that make the SEVA platform a popular resource for genetic analyses, genome editing and bioengineering of a large number of microorganisms.


Subject(s)
Bacteria , Databases, Factual , Bacteria/genetics , Cloning, Molecular , DNA , Genetic Vectors , Phenotype , Plasmids/genetics
5.
Front Microbiol ; 9: 506, 2018.
Article in English | MEDLINE | ID: mdl-29623071

ABSTRACT

Alkylbenzenes, such as toluene and m-xylene, are an important class of contaminant hydrocarbons that are widespread and tend to accumulate in subsurface anoxic environments. The peripheral pathway for the anaerobic oxidation of toluene in bacteria consists of an initial activation catalyzed by a benzylsuccinate synthase (encoded by bss genes), and a subsequent modified ß-oxidation of benzylsuccinate to benzoyl-CoA and succinyl-CoA (encoded by bbs genes). We have shown here that the bss and bbs genes, which are located within an integrative and conjugative element, are essential for anaerobic degradation of toluene but also for m-xylene oxidation in the denitrifying beta-proteobacterium Azoarcus sp. CIB. New insights into the transcriptional organization and regulation of a complete gene cluster for anaerobic catabolism of toluene/m-xylene in a single bacterial strain are presented. The bss and bbs genes are transcriptionally coupled into two large convergent catabolic operons driven by the PbssD and PbbsA promoters, respectively, whose expression is inducible when cells grow anaerobically in toluene or m-xylene. An adjacent tdiSR operon driven by the PtdiS promoter encodes a putative two-component regulatory system. TdiR behaves as a transcriptional activator of the PbssD, PbbsA, and PtdiS promoters, being benzylsuccinate/(3-methyl)benzylsuccinate, rather than toluene/m-xylene, the inducers that may trigger the TdiS-mediated activation of TdiR. In addition to the TdiSR-based specific control, the expression of the bss and bbs genes in Azoarcus sp. CIB is under an overimposed regulation that depends on certain environmental factors, such as the presence/absence of oxygen or the availability of preferred carbon sources (catabolite repression). This work paves the way for future strategies toward the reliable assessment of microbial activity in toluene/m-xylene contaminated environments.

6.
Proc Natl Acad Sci U S A ; 113(46): 13174-13179, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27799551

ABSTRACT

Cyclic diguanosine monophosphate (c-di-GMP) is a second messenger that controls diverse functions in bacteria, including transitions from planktonic to biofilm lifestyles, virulence, motility, and cell cycle. Here we describe TolR, a hybrid two-component system (HTCS), from the ß-proteobacterium Azoarcus sp. strain CIB that degrades c-di-GMP in response to aromatic hydrocarbons, including toluene. This response protects cells from toluene toxicity during anaerobic growth. Whereas wild-type cells tolerated a sudden exposure to a toxic concentration of toluene, a tolR mutant strain or a strain overexpressing a diguanylate cyclase gene lost viability upon toluene shock. TolR comprises an N-terminal aromatic hydrocarbon-sensing Per-Arnt-Sim (PAS) domain, followed by an autokinase domain, a response regulator domain, and a C-terminal c-di-GMP phosphodiesterase (PDE) domain. Autophosphorylation of TolR in response to toluene exposure initiated an intramolecular phosphotransfer to the response regulator domain that resulted in c-di-GMP degradation. The TolR protein was engineered as a functional sensor histidine kinase (TolRSK) and an independent response regulator (TolRRR). This classic two-component system (CTCS) operated less efficiently than TolR, suggesting that TolR was evolved as a HTCS to optimize signal transduction. Our results suggest that TolR enables Azoarcus sp. CIB to adapt to toxic aromatic hydrocarbons under anaerobic conditions by modulating cellular levels of c-di-GMP. This is an additional role for c-di-GMP in bacterial physiology.


Subject(s)
Azoarcus/metabolism , Bacterial Proteins/metabolism , Cyclic GMP/analogs & derivatives , Membrane Proteins/metabolism , Toluene/toxicity , Azoarcus/drug effects , Bacterial Proteins/genetics , Cyclic GMP/metabolism , Membrane Proteins/genetics
8.
Front Microbiol ; 7: 1156, 2016.
Article in English | MEDLINE | ID: mdl-27516758

ABSTRACT

The emergence and spread of antibiotic-resistant bacteria is pushing the need of alternative treatments. In this context, phage therapy is already a reality to successfully fight certain multiresistant bacteria. Among different phage gene products, murein hydrolases responsible of phage progeny liberation (also called lysins or endolysins) are weapons that target specific peptidoglycan bonds, leading to lysis and death of susceptible bacteria when added from the outside. In the pneumococcal system, all but one phage murein hydrolases reported to date share a choline-binding domain that recognizes cell walls containing choline residues in the (lipo)teichoic acids. Some purified pneumococcal or phage murein hydrolases, as well as several chimeric proteins combining natural catalytic and cell wall-binding domains (CBDs) have been used as effective antimicrobials. In this work we have constructed a novel chimeric N-acetylmuramoyl-L-alanine amidase (PL3) by fusing the catalytic domain of the Pal amidase (a phage-coded endolysin) to the CBD of the LytA amidase, the major pneumococcal autolysin. The physicochemical properties of PL3 and the bacteriolytic effect against several pneumococci (including 48 multiresistant representative strain) and related species, like Streptococcus pseudopneumoniae, Streptococcus mitis, and Streptococcus oralis, have been studied. Results have shown that low doses of PL3, in the range of 0.5-5 µg/ml, are enough to practically sterilize all choline-containing strains tested. Moreover, a single 20-µg dose of PL3 fully protected zebrafish embryos from infection by S. pneumoniae D39 strain. Importantly, PL3 keeps 95% enzymatic activity after 4 weeks at 37°C and can be lyophilized without losing activity, demonstrating a remarkable robustness. Such stability, together with a prominent efficacy against a narrow spectrum of human pathogens, confers to PL3 the characteristic to be an effective therapeutic. In addition, our results demonstrate that the structure/function-based domain shuffling approach is a successful method to construct tailor-made endolysins with higher bactericidal activities than their parental enzymes.

9.
Angew Chem Int Ed Engl ; 55(24): 6882-6, 2016 06 06.
Article in English | MEDLINE | ID: mdl-27111486

ABSTRACT

Muropeptides are a group of bacterial natural products generated from the cell wall in the course of its turnover. These compounds are cell-wall recycling intermediates and are also involved in signaling within the bacterium. However, the identity of these signaling molecules remains elusive. The identification and characterization of 20 muropeptides from Pseudomonas aeruginosa is described. The least abundant of these metabolites is present at 100 and the most abundant at 55,000 molecules per bacterium. Analysis of these muropeptides under conditions of induction of resistance to a ß-lactam antibiotic identified two signaling muropeptides (N-acetylglucosamine-1,6-anhydro-N-acetylmuramyl pentapeptide and 1,6-anhydro-N-acetylmuramyl pentapeptide). Authentic synthetic samples of these metabolites were shown to activate expression of ß-lactamase in the absence of any ß-lactam antibiotic, thus indicating that they serve as chemical signals in this complex biochemical pathway.


Subject(s)
Anti-Bacterial Agents/pharmacology , Peptides/metabolism , Pseudomonas aeruginosa/chemistry , beta-Lactam Resistance/drug effects , beta-Lactams/pharmacology , Anti-Bacterial Agents/chemistry , Molecular Conformation , Peptides/chemistry , Pseudomonas aeruginosa/metabolism , beta-Lactamases/metabolism , beta-Lactams/chemistry
10.
J Med Chem ; 58(10): 4194-203, 2015 May 28.
Article in English | MEDLINE | ID: mdl-25923368

ABSTRACT

Rapid emergence of antibiotic resistance is one of the most challenging global public health concerns. In particular, vancomycin-resistant Enterococcus faecium infections have been increasing in frequency, representing 25% of enterococci infections in intensive care units. A novel class of 1,2,4-triazolo[1,5-a]pyrimidines active against E. faecium is reported herein. We used a three-component Biginelli-like heterocyclization reaction for the synthesis of a series of these derivatives based on reactions of aldehydes, ß-dicarbonyl compounds, and 3-alkylthio-5-amino-1,2,4-triazoles. The resulting compounds were assayed for antimicrobial activity against the ESKAPE panel of bacteria, followed by investigation of their in vitro activities. These analyses identified a subset of 1,2,4-triazolo[1,5-a]pyrimidines that had good narrow-spectrum antibacterial activity against E. faecium and exhibited metabolic stability with low intrinsic clearance. Macromolecular synthesis assays revealed cell-wall biosynthesis as the target of these antibiotics.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Enterococcus faecium/drug effects , Pyrimidines/chemistry , Anti-Bacterial Agents/chemical synthesis , Bacterial Proteins/metabolism , Cell Wall/drug effects , Cell Wall/metabolism , Chemistry Techniques, Synthetic , Drug Evaluation, Preclinical/methods , Drug Stability , Humans , Microbial Sensitivity Tests , Penicillin-Binding Proteins/metabolism , Structure-Activity Relationship , Triazoles/chemistry
11.
J Am Chem Soc ; 137(1): 190-200, 2015 Jan 14.
Article in English | MEDLINE | ID: mdl-25495032

ABSTRACT

Pseudomonas aeruginosa is an opportunistic Gram-negative bacterial pathogen. A primary contributor to its ability to resist ß-lactam antibiotics is the expression, following detection of the ß-lactam, of the AmpC ß-lactamase. As AmpC expression is directly linked to the recycling of the peptidoglycan of the bacterial cell wall, an important question is the identity of the signaling molecule(s) in this relationship. One mechanism used by clinical strains to elevate AmpC expression is loss of function of penicillin-binding protein 4 (PBP4). As the mechanism of the ß-lactams is PBP inactivation, this result implies that the loss of the catalytic function of PBP4 ultimately leads to induction of antibiotic resistance. PBP4 is a bifunctional enzyme having both dd-carboxypeptidase and endopeptidase activities. Substrates for both the dd-carboxypeptidase and the 4,3-endopeptidase activities were prepared by multistep synthesis, and their turnover competence with respect to PBP4 was evaluated. The endopeptidase activity is specific to hydrolysis of 4,3-cross-linked peptidoglycan. PBP4 catalyzes both reactions equally well. When P. aeruginosa is grown in the presence of a strong inducer of AmpC, the quantities of both the stem pentapeptide (the substrate for the dd-carboxypeptidase activity) and the 4,3-cross-linked peptidoglycan (the substrate for the 4,3-endopeptidase activity) increase. In the presence of ß-lactam antibiotics these altered cell-wall segments enter into the muropeptide recycling pathway, the conduit connecting the sensing event in the periplasm and the unleashing of resistance mechanisms in the cytoplasm.


Subject(s)
Penicillin-Binding Proteins/metabolism , Pseudomonas aeruginosa/chemistry , Anti-Bacterial Agents/pharmacology , Biocatalysis , Molecular Conformation , Penicillin-Binding Proteins/chemistry , Penicillin-Binding Proteins/genetics , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/metabolism , beta-Lactam Resistance/drug effects
12.
ACS Chem Biol ; 9(9): 2058-66, 2014 Sep 19.
Article in English | MEDLINE | ID: mdl-24988330

ABSTRACT

The lytic transglycosylases are essential bacterial enzymes that catalyze the nonhydrolytic cleavage of the glycan strands of the bacterial cell wall. We describe here the structural and catalytic properties of MltC, one of the seven lytic transglycosylases found in the genome of the Gram-negative bacterium Escherichia coli. The 2.3 Å resolution X-ray structure of a soluble construct of MltC shows a unique, compared to known lytic transglycosylase structures, two-domain structure characterized by an expansive active site of 53 Å length extending through an interface between the domains. The structures of three complexes of MltC with cell wall analogues suggest the positioning of the peptidoglycan in the active site both as a substrate and as a product. One complex is suggested to correspond to an intermediate in the course of sequential and exolytic cleavage of the peptidoglycan. Moreover, MltC partitioned its reactive oxocarbenium-like intermediate between trapping by the C6-hydroxyl of the muramyl moiety (lytic transglycosylase activity, the major path) and by water (muramidase activity). Genomic analysis identifies the presence of an MltC homologue in no less than 791 bacterial genomes. While the role of MltC in cell wall assembly and maturation remains uncertain, we propose a functional role for this enzyme as befits the uniqueness of its two-domain structure.


Subject(s)
Escherichia coli Proteins/metabolism , Glycosyltransferases/chemistry , Glycosyltransferases/metabolism , Catalytic Domain , Cell Wall/metabolism , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/cytology , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Models, Molecular , Mutation , Peptidoglycan/chemistry , Protein Conformation , Protein Structure, Tertiary
13.
Biochemistry ; 53(10): 1548-50, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24564530

ABSTRACT

ß-Lactam antibiotics have faced obsolescence with the emergence of methicillin-resistant Staphylococcus aureus (MRSA). A complex set of events ensues upon exposure of MRSA to these antibiotics, which culminates in proteolysis of BlaI or MecI, two gene repressors, and results in the induction of resistance. We report studies on the mechanism of binding of these gene repressors to the operator regions by fluorescence anisotropy. Within the range of in vivo concentrations for BlaI and MecI, these proteins interact with their regulatory elements in a reversible manner, as both a monomer and a dimer.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Methicillin-Resistant Staphylococcus aureus/metabolism , Repressor Proteins/metabolism , beta-Lactams/pharmacology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Drug Resistance, Bacterial , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/genetics , Operator Regions, Genetic , Operon , Promoter Regions, Genetic , Repressor Proteins/chemistry , Repressor Proteins/genetics
14.
ACS Med Chem Lett ; 3(3): 238-242, 2012 Mar 08.
Article in English | MEDLINE | ID: mdl-22844551

ABSTRACT

Gram-negative bacteria have evolved an elaborate process for the recycling of their cell wall, which is initiated in the periplasmic space by the action of lytic transglycosylases. The product of this reaction, ß-D-N-acetylglucosamine-(1→4)-1,6-anhydro-ß-D-N-acetylmuramyl-L-Ala-γ-D-Glu-meso-DAP-D-Ala-D-Ala (compound 1), is internalized to begin the recycling events within the cytoplasm. The first step in the cytoplasmic recycling is catalyzed by the NagZ glycosylase, which cleaves in a hydrolytic reaction the N-acetylglucosamine glycosidic bond of metabolite 1. The reactions catalyzed by both the lytic glycosylases and NagZ are believed to involve oxocarbenium transition species. We describe herein the synthesis and evaluation of four iminosaccharides as possible mimetics of the oxocarbenium species, and disclose one as a potent (compound 3, K(i) = 300 ± 15 nM) competitive inhibitor of NagZ.

15.
J Biol Chem ; 287(13): 10494-10508, 2012 Mar 23.
Article in English | MEDLINE | ID: mdl-22303008

ABSTRACT

We have studied for the first time the transcriptional regulatory circuit that controls the expression of the box genes encoding the aerobic hybrid pathway used to assimilate benzoate via coenzyme A (CoA) derivatives in bacteria. The promoters responsible for the expression of the box cluster in the ß-proteobacterium Azoarcus sp., their cognate transcriptional repressor, the BoxR protein, and the inducer molecule (benzoyl-CoA) have been characterized. The BoxR protein shows a significant sequence identity to the BzdR transcriptional repressor that controls the bzd genes involved in the anaerobic degradation of benzoate. Because the boxR gene is present in all box clusters so far identified in bacteria, the BoxR/benzoyl-CoA regulatory system appears to be a widespread strategy to control this aerobic hybrid pathway. Interestingly, the paralogous BoxR and BzdR regulators act synergistically to control the expression of the box and bzd genes. This cross-regulation between anaerobic and aerobic pathways for the catabolism of aromatic compounds has never been shown before, and it may reflect a biological strategy to increase the cell fitness in organisms that survive in environments subject to changing oxygen concentrations.


Subject(s)
Azoarcus/metabolism , Bacterial Proteins/metabolism , Benzoates/metabolism , Repressor Proteins/metabolism , Aerobiosis/drug effects , Aerobiosis/physiology , Anaerobiosis/drug effects , Anaerobiosis/physiology , Anti-Infective Agents/metabolism , Anti-Infective Agents/pharmacology , Azoarcus/genetics , Bacterial Proteins/genetics , Base Sequence , Benzoates/pharmacology , Coenzyme A/genetics , Coenzyme A/metabolism , Molecular Sequence Data , Repressor Proteins/genetics
16.
Appl Environ Microbiol ; 76(1): 383-5, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19915033

ABSTRACT

Regulation of aromatic degradation in obligate anaerobes was studied in the Fe(III)-respiring model organism Geobacter metallireducens GS-15. A two-component system and a sigma54-dependent promoter were identified that are both involved in the regulation of the gene coding for benzoate-coenzyme A ligase, catalyzing the initial step of benzoate degradation.


Subject(s)
Gene Expression Regulation, Bacterial , Geobacter/metabolism , Hydrocarbons, Aromatic/metabolism , Signal Transduction , Adaptation, Physiological , Artificial Gene Fusion , Bacterial Proteins/biosynthesis , Base Sequence , Binding Sites , Coenzyme A Ligases/biosynthesis , Genes, Reporter , Geobacter/genetics , Molecular Sequence Data , Promoter Regions, Genetic , RNA Polymerase Sigma 54/metabolism , Transcription Initiation Site , beta-Galactosidase/biosynthesis , beta-Galactosidase/genetics
17.
Microbiol Mol Biol Rev ; 73(1): 71-133, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19258534

ABSTRACT

Aromatic compounds belong to one of the most widely distributed classes of organic compounds in nature, and a significant number of xenobiotics belong to this family of compounds. Since many habitats containing large amounts of aromatic compounds are often anoxic, the anaerobic catabolism of aromatic compounds by microorganisms becomes crucial in biogeochemical cycles and in the sustainable development of the biosphere. The mineralization of aromatic compounds by facultative or obligate anaerobic bacteria can be coupled to anaerobic respiration with a variety of electron acceptors as well as to fermentation and anoxygenic photosynthesis. Since the redox potential of the electron-accepting system dictates the degradative strategy, there is wide biochemical diversity among anaerobic aromatic degraders. However, the genetic determinants of all these processes and the mechanisms involved in their regulation are much less studied. This review focuses on the recent findings that standard molecular biology approaches together with new high-throughput technologies (e.g., genome sequencing, transcriptomics, proteomics, and metagenomics) have provided regarding the genetics, regulation, ecophysiology, and evolution of anaerobic aromatic degradation pathways. These studies revealed that the anaerobic catabolism of aromatic compounds is more diverse and widespread than previously thought, and the complex metabolic and stress programs associated with the use of aromatic compounds under anaerobic conditions are starting to be unraveled. Anaerobic biotransformation processes based on unprecedented enzymes and pathways with novel metabolic capabilities, as well as the design of novel regulatory circuits and catabolic networks of great biotechnological potential in synthetic biology, are now feasible to approach.


Subject(s)
Amino Acids, Aromatic/metabolism , Bacteria, Anaerobic/genetics , Environmental Pollutants/metabolism , Genomics , Hydrocarbons, Aromatic/metabolism , Multigene Family , Anaerobiosis , Bacteria, Anaerobic/metabolism , Biodegradation, Environmental , Ecosystem , Fermentation/genetics , Photophosphorylation/genetics
18.
Environ Microbiol ; 10(2): 474-82, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18177371

ABSTRACT

In this work, the gcdH gene from the denitrifying beta-proteobacterium Azoarcus sp. CIB was shown to encode a glutaryl-CoA dehydrogenase, which is essential for the anaerobic catabolism of many aromatic compounds and some alicyclic and dicarboxylic acids. The primary structure of the GcdH protein is highly conserved in many organisms. The divergently transcribed gcdR gene, encoding a LysR-type transcriptional regulator, accounts for the glutaconate/glutarate-specific activation of the Pg promoter driving expression of gcdH. The Azoarcus sp. CIBdgcdH mutant strain harbouring a disrupted gcdH gene was used as host to identify heterologous gcdH genes, such as that from Pseudomonas putida KT2440. Moreover, the expression of gcdH from P. putida can be efficiently controlled by the GcdR activator in Azoarcus sp. CIB, demonstrating the existence of cross-talk between GcdR regulators and gcdH promoters from members of different phylogenetic subgroups of proteobacteria.


Subject(s)
Azoarcus/enzymology , Gene Expression Regulation, Bacterial , Genes, Regulator , Glutaryl-CoA Dehydrogenase/genetics , Transcription, Genetic , Anaerobiosis , Azoarcus/classification , Azoarcus/genetics , Azoarcus/growth & development , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Base Sequence , Cloning, Molecular , Evolution, Molecular , Glutaryl-CoA Dehydrogenase/chemistry , Glutaryl-CoA Dehydrogenase/metabolism , Hydrocarbons, Aromatic/metabolism , Molecular Sequence Data , Phylogeny , Promoter Regions, Genetic , Sequence Analysis, DNA
19.
Appl Environ Microbiol ; 73(16): 5118-24, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17574992

ABSTRACT

Pseudomonas pseudoalcaligenes CECT5344 grows in minimal medium containing cyanide as the sole nitrogen source. Under these conditions, an O2-dependent respiration highly resistant to cyanide was detected in cell extracts. The structural genes for the cyanide-resistant terminal oxidase, cioA and cioB, are clustered and encode the integral membrane proteins that correspond to subunits I and II of classical cytochrome bd, although the presence of heme d in the membrane could not be detected by difference spectra. The cio operon from P. pseudoalcaligenes presents a singular organization, starting upstream of cioAB by the coding sequence of a putative ferredoxin-dependent sulfite or nitrite reductase and spanning downstream two additional open reading frames that encode uncharacterized gene products. PCR amplifications of RNA (reverse transcription-PCR) indicated the cyanide-dependent up-regulation and cotranscription along the operon. The targeted disruption of cioA eliminates both the expression of the cyanide-stimulated respiratory activity and the growth with cyanide as the nitrogen source, which suggests a critical role of this cytochrome bd-related oxidase in the metabolism of cyanide by P. pseudoalcaligenes CECT5344.


Subject(s)
Bacterial Proteins/metabolism , Cyanides/pharmacology , Drug Resistance, Bacterial , Oxidoreductases/metabolism , Pseudomonas pseudoalcaligenes/drug effects , Bacterial Proteins/genetics , Bacterial Proteins/physiology , Chromosome Mapping , Cloning, Molecular , Gene Expression Regulation, Bacterial/drug effects , Gene Order , Models, Genetic , Molecular Sequence Data , Mutagenesis , Operon , Oxidoreductases/genetics , Oxidoreductases/physiology , Pseudomonas pseudoalcaligenes/enzymology , Pseudomonas pseudoalcaligenes/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA
20.
J Biol Chem ; 280(11): 10683-94, 2005 Mar 18.
Article in English | MEDLINE | ID: mdl-15634675

ABSTRACT

In this work, we have studied the transcriptional regulation of the bzd operon involved in the anaerobic catabolism of benzoate in the denitrifying Azoarcus sp. strain CIB. The transcription start site of the P(N) promoter running the expression of the bzd catabolic genes was identified. Gel retardation assays and P(N)::lacZ translational fusion experiments performed both in Azoarcus sp. CIB and Escherichia coli cells have shown that bzdR encodes a specific repressor that controls the inducible expression of the adjacent bzd catabolic operon, being the first intermediate of the catabolic pathway (i.e. benzoyl-CoA, the actual inducer molecule). This is the first report of a transcriptional repressor and a CoA-derived aromatic inducer controlling gene expression in the anaerobic catabolism of aromatic compounds. DNase I footprinting experiments revealed that BzdR protected three regions (operators) at the P(N) promoter. The three operators contain direct repetitions of a TGCA sequence that forms part of longer palindromic structures. In agreement with the repressor role of BzdR, operator region I spans the transcription initiation site as well as the -10 sequence for recognition of the RNA polymerase. Primary sequence analyses of BzdR showed an unusual modular organization with an N-terminal region homologous to members of the HTH-XRE family of transcriptional regulators and a C-terminal region similar to shikimate kinases. A three-dimensional model of the N-terminal and C-terminal regions of BzdR, generated by comparison with the crystal structures of the SinR regulator from Bacillus subtilis and the shikimate kinase I protein from E. coli, strongly suggests that they contain the helix-turn-helix DNA-binding motif and the benzoyl-CoA binding groove, respectively. The BzdR protein constitutes, therefore, the prototype of a new subfamily of transcriptional regulators.


Subject(s)
Azoarcus/metabolism , Transcription Factors/chemistry , Transcription Factors/physiology , Transcription, Genetic , Amino Acid Motifs , Amino Acid Sequence , Bacillus subtilis/metabolism , Base Sequence , Chromosomes/metabolism , Chromosomes/ultrastructure , Crystallography, X-Ray , DNA-Directed RNA Polymerases/metabolism , Deoxyribonuclease I/chemistry , Deoxyribonuclease I/metabolism , Escherichia coli/metabolism , Genotype , Lac Operon , Models, Genetic , Models, Molecular , Molecular Sequence Data , Multigene Family , Plasmids/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Biosynthesis , Protein Conformation , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...