Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 19.659
Filter
1.
Proc Natl Acad Sci U S A ; 121(28): e2402543121, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38959031

ABSTRACT

The outer membrane (OM) of gram-negative bacteria serves as a vital organelle that is densely populated with OM proteins (OMPs) and plays pivotal roles in cellular functions and virulence. The assembly and insertion of these OMPs into the OM represent a fundamental process requiring specialized molecular chaperones. One example is the translocation and assembly module (TAM), which functions as a transenvelope chaperone promoting the folding of specific autotransporters, adhesins, and secretion systems. The catalytic unit of TAM, TamA, comprises a catalytic ß-barrel domain anchored within the OM and three periplasmic polypeptide-transport-associated (POTRA) domains that recruit the TamB subunit. The latter acts as a periplasmic ladder that facilitates the transport of unfolded OMPs across the periplasm. In addition to their role in recruiting the auxiliary protein TamB, our data demonstrate that the POTRA domains mediate interactions with the inner surface of the OM, ultimately modulating the membrane properties. Through the integration of X-ray crystallography, molecular dynamic simulations, and biomolecular interaction methodologies, we located the membrane-binding site on the first and second POTRA domains. Our data highlight a binding preference for phosphatidylglycerol, a minor lipid constituent present in the OM, which has been previously reported to facilitate OMP assembly. In the context of the densely OMP-populated membrane, this association may serve as a mechanism to secure lipid accessibility for nascent OMPs through steric interactions with existing OMPs, in addition to creating favorable conditions for OMP biogenesis.


Subject(s)
Bacterial Outer Membrane Proteins , Escherichia coli Proteins , Bacterial Outer Membrane Proteins/metabolism , Bacterial Outer Membrane Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Protein Domains , Bacterial Outer Membrane/metabolism , Escherichia coli/metabolism , Escherichia coli/genetics , Molecular Chaperones/metabolism , Molecular Chaperones/chemistry , Protein Folding , Periplasm/metabolism , Models, Molecular
2.
Subcell Biochem ; 104: 17-31, 2024.
Article in English | MEDLINE | ID: mdl-38963481

ABSTRACT

The copper efflux regulator (CueR) is a classical member of the MerR family of metalloregulators and is common in gram-negative bacteria. Through its C-terminal effector-binding domain, CueR senses cytoplasmic copper ions to regulate the transcription of genes contributing to copper homeostasis, an essential process for survival of all cells. In this chapter, we review the regulatory roles of CueR in the model organism Escherichia coli and the mechanisms for CueR in copper binding, DNA recognition, and interplay with RNA polymerase in regulating transcription. In light of biochemical and structural analyses, we provide molecular details for how CueR represses transcription in the absence of copper ions, how copper ions mediate CueR conformational change to form holo CueR, and how CueR bends and twists promoter DNA to activate transcription. We also characterize the functional domains and key residues involved in these processes. Since CueR is a representative member of the MerR family, elucidating its regulatory mechanisms could help to understand the CueR-like regulators in other organisms and facilitate the understanding of other metalloregulators in the same family.


Subject(s)
Copper , Escherichia coli Proteins , Escherichia coli , Gene Expression Regulation, Bacterial , Copper/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Transcription, Genetic , Promoter Regions, Genetic , Trans-Activators
3.
Subcell Biochem ; 104: 49-71, 2024.
Article in English | MEDLINE | ID: mdl-38963483

ABSTRACT

Across living organisms, division is necessary for cell survival and passing heritable information to the next generation. For this reason, cell division is highly conserved among eukaryotes and prokaryotes. Among the most highly conserved cell division proteins in eukaryotes are tubulin and actin. Tubulin polymerizes to form microtubules, which assemble into cytoskeletal structures in eukaryotes, such as the mitotic spindle that pulls chromatids apart during mitosis. Actin polymerizes to form a morphological framework for the eukaryotic cell, or cytoskeleton, that undergoes reorganization during mitosis. In prokaryotes, two of the most highly conserved cell division proteins are the tubulin homolog FtsZ and the actin homolog FtsA. In this chapter, the functions of the essential bacterial cell division proteins FtsZ and FtsA and their roles in assembly of the divisome at the septum, the site of cell division, will be discussed. In most bacteria, including Escherichia coli, the tubulin homolog FtsZ polymerizes at midcell, and this step is crucial for recruitment of many other proteins to the division site. For this reason, both FtsZ abundance and polymerization are tightly regulated by a variety of proteins. The actin-like FtsA protein polymerizes and tethers FtsZ polymers to the cytoplasmic membrane. Additionally, FtsA interacts with later stage cell division proteins, which are essential for division and for building the new cell wall at the septum. Recent studies have investigated how actin-like polymerization of FtsA on the lipid membrane may impact division, and we will discuss this and other ways that division in bacteria is regulated through FtsZ and FtsA.


Subject(s)
Bacterial Proteins , Cell Division , Cytoskeletal Proteins , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Cytoskeletal Proteins/metabolism , Cytoskeletal Proteins/genetics , Escherichia coli/metabolism , Escherichia coli/genetics , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , Bacteria/metabolism , Bacteria/genetics
4.
Arh Hig Rada Toksikol ; 75(2): 147-154, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38963138

ABSTRACT

Mistakes in translation are mostly associated with toxic effects in the cell due to the production of functionally aberrant and misfolded proteins. However, under certain circumstances mistranslation can have beneficial effects and enable cells to preadapt to other stress conditions. Mistranslation may be caused by mistakes made by aminoacyl-tRNA synthetases, essential enzymes that link amino acids to cognate tRNAs. There is an Escherichia coli strain expressing isoleucyl-tRNA synthetase mutant variant with inactivated editing domain which produces mistranslated proteomes where valine (Val) and norvaline (Nva) are misincorporated into proteins instead of isoleucine. We compared this strain with the wild-type to determine the effects of such mistranslation on bacterial growth in oxidative stress conditions. When the cells were pre-incubated with 0.75 mmol/L Nva or 1.5 mmol/L Val or Nva and exposed to hydrogen peroxide, no beneficial effect of mistranslation was observed. However, when the editing-deficient strain was cultivated in medium supplemented with 0.75 mmol/L Val up to the early or mid-exponential phase of growth and then exposed to oxidative stress, it slightly outgrew the wild-type grown in the same conditions. Our results therefore show a modest adaptive effect of isoleucine mistranslation on bacterial growth in oxidative stress, but only in specific conditions. This points to a delicate balance between deleterious and beneficial effects of mistranslation.


Subject(s)
Escherichia coli , Oxidative Stress , Oxidative Stress/drug effects , Escherichia coli/drug effects , Escherichia coli/genetics , Protein Biosynthesis/drug effects , Escherichia coli Proteins/genetics , Hydrogen Peroxide
5.
Elife ; 132024 Jul 03.
Article in English | MEDLINE | ID: mdl-38959062

ABSTRACT

Bacterial exonuclease III (ExoIII), widely acknowledged for specifically targeting double-stranded DNA (dsDNA), has been documented as a DNA repair-associated nuclease with apurinic/apyrimidinic (AP)-endonuclease and 3'→5' exonuclease activities. Due to these enzymatic properties, ExoIII has been broadly applied in molecular biosensors. Here, we demonstrate that ExoIII (Escherichia coli) possesses highly active enzymatic activities on ssDNA. By using a range of ssDNA fluorescence-quenching reporters and fluorophore-labeled probes coupled with mass spectrometry analysis, we found ExoIII cleaved the ssDNA at 5'-bond of phosphodiester from 3' to 5' end by both exonuclease and endonuclease activities. Additional point mutation analysis identified the critical residues for the ssDNase action of ExoIII and suggested the activity shared the same active center with the dsDNA-targeted activities of ExoIII. Notably, ExoIII could also digest the dsDNA structures containing 3'-end ssDNA. Considering most ExoIII-assisted molecular biosensors require the involvement of single-stranded DNA (ssDNA) or nucleic acid aptamer containing ssDNA, the activity will lead to low efficiency or false positive outcome. Our study revealed the multi-enzymatic activity and the underlying molecular mechanism of ExoIII on ssDNA, illuminating novel insights for understanding its biological roles in DNA repair and the rational design of ExoIII-ssDNA involved diagnostics.


Subject(s)
DNA, Single-Stranded , Escherichia coli , Exodeoxyribonucleases , Exodeoxyribonucleases/metabolism , Exodeoxyribonucleases/genetics , DNA, Single-Stranded/metabolism , DNA, Single-Stranded/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli/enzymology , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics
6.
RNA Biol ; 21(1): 31-41, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38952121

ABSTRACT

Large ribosomal RNAs (rRNAs) are modified heavily post-transcriptionally in functionally important regions but, paradoxically, individual knockouts (KOs) of the modification enzymes have minimal impact on Escherichia coli growth. Furthermore, we recently constructed a strain with combined KOs of five modification enzymes (RluC, RlmKL, RlmN, RlmM and RluE) of the 'critical region' of the peptidyl transferase centre (PTC) in 23S rRNA that exhibited only a minor growth defect at 37°C (although major at 20°C). However, our combined KO of modification enzymes RluC and RlmE (not RluE) resulted in conditional lethality (at 20°C). Although the growth rates for both multiple-KO strains were characterized, the molecular explanations for such deficits remain unclear. Here, we pinpoint biochemical defects in these strains. In vitro fast kinetics at 20°C and 37°C with ribosomes purified from both strains revealed, counterintuitively, the slowing of translocation, not peptide bond formation or peptidyl release. Elongation rates of protein synthesis in vivo, as judged by the kinetics of ß-galactosidase induction, were also slowed. For the five-KO strain, the biggest deficit at 37°C was in 70S ribosome assembly, as judged by a dominant 50S peak in ribosome sucrose gradient profiles at 5 mM Mg2+. Reconstitution of this 50S subunit from purified five-KO rRNA and ribosomal proteins supported a direct role in ribosome biogenesis of the PTC region modifications per se, rather than of the modification enzymes. These results clarify the importance and roles of the enigmatic rRNA modifications.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Peptidyl Transferases , Protein Biosynthesis , RNA, Ribosomal , Ribosomes , Peptidyl Transferases/metabolism , Peptidyl Transferases/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Ribosomes/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , RNA, Ribosomal/genetics , RNA, Ribosomal/metabolism , RNA, Ribosomal, 23S/metabolism , RNA, Ribosomal, 23S/genetics , Kinetics
7.
Nat Commun ; 15(1): 5683, 2024 Jul 06.
Article in English | MEDLINE | ID: mdl-38971825

ABSTRACT

Bacteria within mature biofilms are highly resistant to antibiotics than planktonic cells. Oxygen limitation contributes to antibiotic resistance in mature biofilms. Nitric oxide (NO) induces biofilm dispersal; however, low NO levels stimulate biofilm formation, an underexplored process. Here, we introduce a mechanism of anaerobic biofilm formation by investigating the antibiofilm activity of tyrosol, a component in wine. Tyrosol inhibits E. coli and Pseudomonas aeruginosa biofilm formation by enhancing NO production. YbfA is identified as a target of tyrosol and its downstream targets are sequentially determined. YbfA activates YfeR, which then suppresses the anaerobic regulator FNR. This suppression leads to decreased NO production, elevated bis-(3'-5')-cyclic dimeric GMP levels, and finally stimulates anaerobic biofilm formation in the mature stage. Blocking YbfA with tyrosol treatment renders biofilm cells as susceptible to antibiotics as planktonic cells. Thus, this study presents YbfA as a promising antibiofilm target to address antibiotic resistance posed by biofilm-forming bacteria, with tyrosol acting as an inhibitor.


Subject(s)
Anti-Bacterial Agents , Biofilms , Escherichia coli , Nitric Oxide , Phenylethyl Alcohol , Pseudomonas aeruginosa , Biofilms/drug effects , Biofilms/growth & development , Phenylethyl Alcohol/analogs & derivatives , Phenylethyl Alcohol/pharmacology , Anti-Bacterial Agents/pharmacology , Escherichia coli/drug effects , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/physiology , Nitric Oxide/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/antagonists & inhibitors , Anaerobiosis/drug effects , Microbial Sensitivity Tests , Gene Expression Regulation, Bacterial/drug effects , Cyclic GMP/metabolism , Cyclic GMP/analogs & derivatives , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/antagonists & inhibitors
8.
BMC Microbiol ; 24(1): 248, 2024 Jul 06.
Article in English | MEDLINE | ID: mdl-38971718

ABSTRACT

BACKGROUND: The usage of fluoroquinolones in Norwegian livestock production is very low, including in broiler production. Historically, quinolone-resistant Escherichia coli (QREC) isolated from Norwegian production animals rarely occur. However, with the introduction of a selective screening method for QREC in the Norwegian monitoring programme for antimicrobial resistance in the veterinary sector in 2014; 89.5% of broiler caecal samples and 70.7% of broiler meat samples were positive. This triggered the concern if there could be possible links between broiler and human reservoirs of QREC. We are addressing this by characterizing genomes of QREC from humans (healthy carriers and patients) and broiler isolates (meat and caecum). RESULTS: The most frequent mechanism for quinolone resistance in both broiler and human E. coli isolates were mutations in the chromosomally located gyrA and parC genes, although plasmid mediated quinolone resistance (PMQR) was also identified. There was some relatedness of the isolates within human and broiler groups, but little between these two groups. Further, some overlap was seen for isolates with the same sequence type isolated from broiler and humans, but overall, the SNP distance was high. CONCLUSION: Based on data from this study, QREC from broiler makes a limited contribution to the incidence of QREC in humans in Norway.


Subject(s)
Anti-Bacterial Agents , Chickens , Drug Resistance, Bacterial , Escherichia coli Infections , Escherichia coli , Quinolones , Animals , Chickens/microbiology , Escherichia coli/genetics , Escherichia coli/drug effects , Escherichia coli/isolation & purification , Humans , Norway , Escherichia coli Infections/veterinary , Escherichia coli Infections/microbiology , Drug Resistance, Bacterial/genetics , Quinolones/pharmacology , Anti-Bacterial Agents/pharmacology , Genomics , Plasmids/genetics , Poultry Diseases/microbiology , Microbial Sensitivity Tests , Genome, Bacterial/genetics , DNA Gyrase/genetics , DNA Topoisomerase IV/genetics , Meat/microbiology , Mutation , Escherichia coli Proteins/genetics , Cecum/microbiology
9.
Proc Natl Acad Sci U S A ; 121(26): e2401154121, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38889150

ABSTRACT

Almost all elongator tRNAs (Transfer RNAs) harbor 5-methyluridine 54 and pseudouridine 55 in the T arm, generated by the enzymes TrmA and TruB, respectively, in Escherichia coli. TrmA and TruB both act as tRNA chaperones, and strains lacking trmA or truB are outcompeted by wild type. Here, we investigate how TrmA and TruB contribute to cellular fitness. Deletion of trmA and truB in E. coli causes a global decrease in aminoacylation and alters other tRNA modifications such as acp3U47. While overall protein synthesis is not affected in ΔtrmA and ΔtruB strains, the translation of a subset of codons is significantly impaired. As a consequence, we observe translationally reduced expression of many specific proteins, that are either encoded with a high frequency of these codons or that are large proteins. The resulting proteome changes are not related to a specific growth phenotype, but overall cellular fitness is impaired upon deleting trmA and truB in accordance with a general protein synthesis impact. In conclusion, we demonstrate that universal modifications of the tRNA T arm are critical for global tRNA function by enhancing tRNA maturation, tRNA aminoacylation, and translation, thereby improving cellular fitness irrespective of the growth conditions which explains the conservation of trmA and truB.


Subject(s)
Escherichia coli , RNA, Transfer , RNA, Transfer/metabolism , RNA, Transfer/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Protein Biosynthesis , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , tRNA Methyltransferases/metabolism , tRNA Methyltransferases/genetics , RNA Processing, Post-Transcriptional
10.
PLoS One ; 19(6): e0305823, 2024.
Article in English | MEDLINE | ID: mdl-38917203

ABSTRACT

SlyD is a widely-occurring prokaryotic FKBP-family prolyl isomerase with an additional chaperone domain. Often, such as in Escherichia coli, a third domain is found at its C-terminus that binds nickel and provides it for nickel-enzyme biogenesis. SlyD has been found to bind signal peptides of proteins that are translocated by the Tat pathway, a system for the transport of folded proteins across membranes. Using peptide arrays to analyze these signal peptide interactions, we found that SlyD interacted only with positively charged peptides, with a preference for arginines over lysines, and large hydrophobic residues enhanced binding. Especially a twin-arginine motif was recognized, a pair of highly conserved arginines adjacent to a stretch of hydrophobic residues. Using isothermal titration calorimetry (ITC) with purified SlyD and a signal peptide-containing model Tat substrate, we could show that the wild type twin-arginine signal peptide was bound with higher affinity than an RR>KK mutated variant, confirming that positive charges are recognized by SlyD, with a preference of arginines over lysines. The specific role of negative charges of the chaperone domain surface and of hydrophobic residues in the chaperone active site was further analyzed by ITC of mutated SlyD variants. Our data show that the supposed key hydrophobic residues of the active site are indeed crucial for binding, and that binding is influenced by negative charges on the chaperone domain. Recognition of positive charges is likely achieved by a large negatively charged surface region of the chaperone domain, which is highly conserved although individual positions are variable.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Molecular Chaperones , Peptidylprolyl Isomerase , Protein Binding , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/chemistry , Peptidylprolyl Isomerase/metabolism , Peptidylprolyl Isomerase/genetics , Escherichia coli/metabolism , Escherichia coli/genetics , Molecular Chaperones/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/chemistry , Protein Sorting Signals , Hydrophobic and Hydrophilic Interactions , Calorimetry , Arginine/metabolism , Amino Acid Sequence
11.
Microbiol Res ; 285: 127774, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38833829

ABSTRACT

Extended-spectrumß-lactam producing Escherichia coli (ESBL-EC) readily colonizes live poultry and serves as a major source of contamination in retail chicken meat, posing significant threats to public health. This study aims to investigate the impact of inappropriate antibiotic use on the dissemination and exacerbation of antibiotic resistance in ESBL-EC and explore the underlying molecular mechanisms. Through experimental analysis, we propose a hypothesis that inappropriate antibiotic use may exacerbate resistance by affecting vesicle formation and protein secretion. Experimental results demonstrate that under the influence of amoxicillin, the concentration of proteins secreted in outer membrane vehicles (OMVs) by ESBL-EC significantly increases, along with a significant upregulation in the expression of the CTX-M-55-type Extended-spectrum beta-lactamase (CTX-M-55). Proteomic analysis and differential gene knockout experiments identified the key protein YdcZ, associated with OMVs formation and protein transportation in ESBL-EC under amoxicillin treatment. Further investigations reveal direct interactions between YdcZ and other proteins (YdiH and BssR). Upon ydcz gene knockout, a significant decrease in protein concentration within OMVs is observed, accompanied by a noticeable reduction in protection against sensitive bacteria. These findings suggest a critical role of YdcZ in regulating the process of protein transportation to OMVs in ESBL-EC under the influence of amoxicillin. In summary, our research uncovers the significant role of inappropriate antibiotic use in promoting the secretion of OMVs by ESBL-EC, aiding the survival of antibiotic-sensitive bacteria in the vicinity of infection sites. These findings provide new insights into the mechanisms underlying antibiotic-induced bacterial resistance dissemination and offer novel avenues for exploring prevention and control strategies against bacterial resistance propagation.


Subject(s)
Amoxicillin , Anti-Bacterial Agents , Escherichia coli Proteins , Escherichia coli , Protein Transport , beta-Lactamases , Anti-Bacterial Agents/pharmacology , Escherichia coli/genetics , Escherichia coli/drug effects , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , beta-Lactamases/metabolism , beta-Lactamases/genetics , Amoxicillin/pharmacology , Animals , Microbial Sensitivity Tests , Proteomics , Bacterial Outer Membrane Proteins/metabolism , Bacterial Outer Membrane Proteins/genetics , Chickens/microbiology , Drug Resistance, Bacterial , Bacterial Outer Membrane/drug effects , Bacterial Outer Membrane/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Infections/drug therapy
12.
J Agric Food Chem ; 72(25): 14216-14228, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38860925

ABSTRACT

Two-component systems (TCSs) sensing and responding to various stimuli outside and inside cells are valuable resources for developing biosensors with synthetic biology applications. However, the use of TCS-based biosensors suffers from a limited effector spectrum, hypersensitivity, low dynamic range, and unwanted signal crosstalk. Here, we developed a tailor-made Escherichia coli whole-cell γ-aminobutyric acid (GABA) biosensor by engineering a chimeric GABA chemoreceptor PctC and TCS. By testing different TCSs, the chimeric PctC/PhoQ showed the response to GABA. Chimera-directed evolution and introduction of the insulated chimeric pair PctC/PhoQ*PhoP* produced biosensors with up to 3.50-fold dynamic range and good orthogonality. To further enhance the dynamic range and lower the basal leakage, three strategies, engineering of PhoP DNA binding sites, fine-tuning reporter expression by optimizing transcription/translation components, and a tobacco etch virus protease-controlled protein degradation, were integrated. This chimeric biosensor displayed a low basal leakage, a large dynamic range (15.8-fold), and a high threshold level (22.7 g L-1). Finally, the optimized biosensor was successfully applied in the high-throughput microdroplet screening of GABA-overproducing Corynebacterium glutamicum, demonstrating its desired properties for extracellular signal biosensing.


Subject(s)
Biosensing Techniques , Escherichia coli Proteins , Escherichia coli , gamma-Aminobutyric Acid , Biosensing Techniques/methods , Biosensing Techniques/instrumentation , gamma-Aminobutyric Acid/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism
13.
Microb Cell Fact ; 23(1): 173, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38867236

ABSTRACT

BACKGROUND: The microbial chiral product (R)-3-hydroxybutyrate (3-HB) is a gateway to several industrial and medical compounds. Acetyl-CoA is the key precursor for 3-HB, and several native pathways compete with 3-HB production. The principal competing pathway in wild-type Escherichia coli for acetyl-CoA is mediated by citrate synthase (coded by gltA), which directs over 60% of the acetyl-CoA into the tricarboxylic acid cycle. Eliminating citrate synthase activity (deletion of gltA) prevents growth on glucose as the sole carbon source. In this study, an alternative approach is used to generate an increased yield of 3-HB: citrate synthase activity is reduced but not eliminated by targeted substitutions in the chromosomally expressed enzyme. RESULTS: Five E. coli GltA variants were examined for 3-HB production via heterologous overexpression of a thiolase (phaA) and NADPH-dependent acetoacetyl-CoA reductase (phaB) from Cupriavidus necator. In shake flask studies, four variants showed nearly 5-fold greater 3-HB yield compared to the wild-type, although pyruvate accumulated. Overexpression of either native thioesterases TesB or YciA eliminated pyruvate formation, but diverted acetyl-CoA towards acetate formation. Overexpression of pantothenate kinase similarly decreased pyruvate formation but did not improve 3-HB yield. Controlled batch studies at the 1.25 L scale demonstrated that the GltA[A267T] variant produced the greatest 3-HB titer of 4.9 g/L with a yield of 0.17 g/g. In a phosphate-starved repeated batch process, E. coli ldhA poxB pta-ackA gltA::gltA[A267T] generated 15.9 g/L 3-HB (effective concentration of 21.3 g/L with dilution) with yield of 0.16 g/g from glucose as the sole carbon source. CONCLUSIONS: This study demonstrates that GltA variants offer a means to affect the generation of acetyl-CoA derived products. This approach should benefit a wide range of acetyl-CoA derived biochemical products in E. coli and other microbes. Enhancing substrate affinity of the introduced pathway genes like thiolase towards acetyl-CoA will likely further increase the flux towards 3-HB while reducing pyruvate and acetate accumulation.


Subject(s)
3-Hydroxybutyric Acid , Acetyl Coenzyme A , Citrate (si)-Synthase , Escherichia coli , Escherichia coli/genetics , Escherichia coli/metabolism , Acetyl Coenzyme A/metabolism , Citrate (si)-Synthase/metabolism , Citrate (si)-Synthase/genetics , 3-Hydroxybutyric Acid/metabolism , 3-Hydroxybutyric Acid/biosynthesis , Metabolic Engineering/methods , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , Ketone Oxidoreductases/metabolism , Ketone Oxidoreductases/genetics , Alcohol Oxidoreductases
14.
J Agric Food Chem ; 72(25): 14274-14283, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38867465

ABSTRACT

ß-Alanine, a valuable ß-type amino acid, is experiencing increased demand due to its multifaceted applications in food flavoring, nutritional supplements, pharmaceuticals, and the chemical industry. Nevertheless, the sustainable biosynthesis of ß-alanine currently faces challenges due to the scarcity of robust strains, attributed to the complexities of modulating multiple genes and the inherent physiological constraints. Here, systems metabolic engineering was implemented in Escherichia coli to overcome these limitations. First, an efficient l-aspartate-α-decarboxylase (ADC) was recruited for ß-alanine biosynthesis. To conserve phosphoenolpyruvate flux, we subsequently modified the endogenous glucose assimilation system by inactivating the phosphotransferase system (PTS) and introducing an alternative non-PTS system, which increased ß-alanine production to 1.70 g/L. The supply of key precursors, oxaloacetate and l-aspartate, was synergistically improved through comprehensive modulation, including strengthening main flux and blocking bypass metabolism, which significantly increased the ß-alanine titer to 3.43 g/L. Next, the expression of ADC was optimized by promoter and untranslated region (UTR) engineering. Further transport engineering, which involved disrupting ß-alanine importer CycA and heterologously expressing ß-alanine exporter NCgI0580, improved ß-alanine production to 8.48 g/L. Additionally, corn steep liquor was used to develop a cost-effective medium. The final strain produced 74.03 g/L ß-alanine with a yield of 0.57 mol/mol glucose during fed-batch fermentation.


Subject(s)
Escherichia coli , Fermentation , Glucose , Metabolic Engineering , beta-Alanine , beta-Alanine/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Glucose/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism
15.
Microbiol Spectr ; 12(7): e0070424, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38860818

ABSTRACT

The impact of chromosomally encoded wild-type or extended-spectrum (ESAC) AmpC ß-lactamases of Escherichia coli on susceptibility to ceftazidime, cefepime, and cefiderocol was evaluated in different genetic backgrounds, including wild-type, PBP3-modified, and porin-deficient E. coli strains. Recombinant E. coli strains possessing the different backgrounds and producing variable ESACs were evaluated. Although ESAC enzymes conferred resistance to ceftazidime and decreased susceptibility to cefepime as expected, we showed here that cefiderocol was also a substrate of ESAC enzymes. IMPORTANCE: We showed here that chromosomally encoded intrinsic extended-spectrum cephalosporinases of Escherichia coli may impact susceptibility not only to ceftazidime and cefepime but also to cefiderocol.


Subject(s)
Anti-Bacterial Agents , Bacterial Proteins , Cefiderocol , Cephalosporins , Escherichia coli , Microbial Sensitivity Tests , beta-Lactamases , Escherichia coli/genetics , Escherichia coli/drug effects , Escherichia coli/enzymology , beta-Lactamases/genetics , beta-Lactamases/metabolism , Cephalosporins/pharmacology , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Ceftazidime/pharmacology , Chromosomes, Bacterial/genetics , Cefepime/pharmacology
16.
Antimicrob Resist Infect Control ; 13(1): 66, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38886812

ABSTRACT

BACKGROUND: Carbapenem-resistant E. coli (CREco) pose a significant public health threat due to their multidrug resistance. Colistin is often a last-resort treatment against CREco; however, the emergence of colistin resistance gene mcr-1 complicates treatment options. METHODS: Two E. coli strains (ECO20 and ECO21), recovered from hospitalized patients in distinct wards, exhibited resistance to carbapenems and colistin. Whole-genome sequencing and phenotypic characterization were employed to study resistance patterns, plasmid profiles, transferability of resistance and virulence genes, and siderophore production capabilities. Comparative genome analysis was used to investigate the genetic environment of mcr-1, blaNDM-7, and virulence clusters. RESULTS: Both E. coli strains exhibited thr presence of both mcr-1 and blaNDM-7 genes, showing high resistance to multiple antibiotics. Genomic analysis revealed the clonal transmission of these strains, possessing identical plasmid profiles (pMCR, pNDM, and pVir) associated with colistin resistance, carbapenem resistance, and virulence factors. Conjugation experiments confirmed the transferability of these plasmids, indicating their potential to disseminate resistance and virulence traits to other strains. Comparative genomic analyses unveiled the distribution of mcr-1 (IncX4-type) and blaNDM (IncX3-type) plasmids across diverse bacterial species, emphasizing their adaptability and threat. The novelty of pVir indicates its potential role in driving the evolution of highly adaptable and pathogenic strains. CONCLUSIONS: Our findings underscore the co-occurrence of mcr-1, blaNDM-7, and siderophore-producing plasmids in E. coli, which poses a significant concern for global health. This research is crucial to unravel the complex mechanisms governing plasmid transfer and recombination and to devise robust strategies to control their spread in healthcare settings.


Subject(s)
Anti-Bacterial Agents , Drug Resistance, Multiple, Bacterial , Escherichia coli Infections , Escherichia coli Proteins , Escherichia coli , Plasmids , Siderophores , Plasmids/genetics , Escherichia coli/genetics , Escherichia coli/drug effects , Escherichia coli Proteins/genetics , Humans , Escherichia coli Infections/microbiology , Anti-Bacterial Agents/pharmacology , China , Drug Resistance, Multiple, Bacterial/genetics , Whole Genome Sequencing , Colistin/pharmacology , Microbial Sensitivity Tests , beta-Lactamases/genetics , Hospitals , Carbapenems/pharmacology , Virulence Factors/genetics
17.
J Agric Food Chem ; 72(26): 14821-14829, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38897918

ABSTRACT

d-Allulose, a C-3 epimer of d-fructose, has great market potential in food, healthcare, and medicine due to its excellent biochemical and physiological properties. Microbial fermentation for d-allulose production is being developed, which contributes to cost savings and environmental protection. A novel metabolic pathway for the biosynthesis of d-allulose from a d-xylose-methanol mixture has shown potential for industrial application. In this study, an artificial antisense RNA (asRNA) was introduced into engineered Escherichia coli to diminish the flow of pentose phosphate (PP) pathway, while the UDP-glucose-4-epimerase (GalE) was knocked out to prevent the synthesis of byproducts. As a result, the d-allulose yield on d-xylose was increased by 35.1%. Then, we designed a d-xylose-sensitive translation control system to regulate the expression of the formaldehyde detoxification operon (FrmRAB), achieving self-inductive detoxification by cells. Finally, fed-batch fermentation was carried out to improve the productivity of the cell factory. The d-allulose titer reached 98.6 mM, with a yield of 0.615 mM/mM on d-xylose and a productivity of 0.969 mM/h.


Subject(s)
Escherichia coli , Fermentation , Methanol , RNA, Antisense , Xylose , Escherichia coli/genetics , Escherichia coli/metabolism , Xylose/metabolism , RNA, Antisense/genetics , RNA, Antisense/metabolism , Methanol/metabolism , Metabolic Engineering , Fructose/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism
18.
J Agric Food Chem ; 72(26): 14809-14820, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38899780

ABSTRACT

Vanillin is one of the world's most extensively used flavoring agents with high application value. However, the yield of vanillin biosynthesis remains limited due to the low efficiency of substrate uptake and the inhibitory effect on cell growth caused by vanillin. Here, we screened high-efficiency ferulic acid importer TodX and vanillin exporters PP_0178 and PP_0179 by overexpressing genes encoding candidate transporters in a vanillin-producing engineered Escherichia coli strain VA and further constructed an autoregulatory bidirectional transport system by coexpressing TodX and PP_0178/PP_0179 with a vanillin self-inducible promoter ADH7. Compared with strain VA, strain VA-TodX-PP_0179 can efficiently transport ferulic acid across the cell membrane and convert it to vanillin, which significantly increases the substrate utilization rate efficiency (14.86%) and vanillin titer (51.07%). This study demonstrated that the autoregulatory bidirectional transport system significantly enhances the substrate uptake efficiency while alleviating the vanillin toxicity issue, providing a promising viable route for vanillin biosynthesis.


Subject(s)
Benzaldehydes , Escherichia coli , Metabolic Engineering , Benzaldehydes/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli/drug effects , Biological Transport , Coumaric Acids/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism
19.
Bull Exp Biol Med ; 176(6): 791-795, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38890214

ABSTRACT

E. coli exposure to ciprofloxacin disturbs cysteine homeostasis; an increase in the intracellular concentration of cysteine is dangerous due to its ability to enhance ROS generation. Unlike wild-type bacteria, in which the cysteine content did not exceed the control level, cells of the gshA mutant lacking glutathione are characterized by increased concentration of intracellular cysteine in proportion to the concentrations of the antibiotic, despite the intensive export of cysteine into the medium. At low concentrations of ciprofloxacin, the mutant strain formed half as many colonies as the parent strain in the survival test. These findings attest to the important role of the incorporation of excess cysteine into glutathione as one of the mechanisms of cysteine homeostasis during the stress response to antibiotic.


Subject(s)
Anti-Bacterial Agents , Ciprofloxacin , Cysteine , Escherichia coli , Homeostasis , Ciprofloxacin/pharmacology , Cysteine/metabolism , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli/metabolism , Homeostasis/drug effects , Anti-Bacterial Agents/pharmacology , Glutathione/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Reactive Oxygen Species/metabolism , Microbial Sensitivity Tests , Mutation
20.
PLoS Genet ; 20(6): e1011335, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38913742

ABSTRACT

The outer membrane of gram-negative bacteria is a barrier to chemical and physical stress. Phospholipid transport between the inner and outer membranes has been an area of intense investigation and, in E. coli K-12, it has recently been shown to be mediated by YhdP, TamB, and YdbH, which are suggested to provide hydrophobic channels for phospholipid diffusion, with YhdP and TamB playing the major roles. However, YhdP and TamB have different phenotypes suggesting distinct functions. It remains unclear whether these functions are related to phospholipid metabolism. We investigated a synthetic cold sensitivity caused by deletion of fadR, a transcriptional regulator controlling fatty acid degradation and unsaturated fatty acid production, and yhdP, but not by ΔtamB ΔfadR or ΔydbH ΔfadR. Deletion of tamB recuses the ΔyhdP ΔfadR cold sensitivity further demonstrating the phenotype is related to functional diversification between these genes. The ΔyhdP ΔfadR strain shows a greater increase in cardiolipin upon transfer to the non-permissive temperature and genetically lowering cardiolipin levels can suppress cold sensitivity. These data also reveal a qualitative difference between cardiolipin synthases in E. coli, as deletion of clsA and clsC suppresses cold sensitivity but deletion of clsB does not. Moreover, increased fatty acid saturation is necessary for cold sensitivity and lowering this level genetically or through supplementation of oleic acid suppresses the cold sensitivity of the ΔyhdP ΔfadR strain. Together, our data clearly demonstrate that the diversification of function between YhdP and TamB is related to phospholipid metabolism. Although indirect regulatory effects are possible, we favor the parsimonious hypothesis that YhdP and TamB have differential phospholipid-substrate transport preferences. Thus, our data provide a potential mechanism for independent control of the phospholipid composition of the inner and outer membranes in response to changing conditions based on regulation of abundance or activity of YhdP and TamB.


Subject(s)
Escherichia coli Proteins , Phospholipids , Phospholipids/metabolism , Phospholipids/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Biological Transport/genetics , Cardiolipins/metabolism , Cardiolipins/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Cold Temperature , Escherichia coli K12/genetics , Escherichia coli K12/metabolism , Gene Expression Regulation, Bacterial , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Fatty Acids/metabolism , Phospholipid Transfer Proteins/genetics , Phospholipid Transfer Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...