Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Molecules ; 28(11)2023 May 23.
Article in English | MEDLINE | ID: mdl-37298737

ABSTRACT

The spread of antibiotic resistance is an urgent threat to global health that requires new therapeutic approaches. Treatments for pathogenic Gram-negative bacteria are particularly challenging to identify due to the robust OM permeability barrier in these organisms. One strategy is to use antibiotic adjuvants, a class of drugs that have no significant antibacterial activity on their own but can act synergistically with certain antibiotics. Previous studies described the discovery and development of polyaminoisoprenyl molecules as antibiotic adjuvants with an OM effect. In particular, the compound NV716 has been shown to sensitize Pseudomonas aeruginosa to tetracycline antibiotics such as doxycycline. Here, we sought to explore the disruption of OM to sensitize P. aeruginosa to otherwise inactive antimicrobials using a series of tetracycline derivatives in the presence of NV716. We found that OM disruption expands the hydrophobicity threshold consistent with antibacterial activity to include hydrophobic molecules, thereby altering permeation rules in Gram-negative bacteria.


Subject(s)
Anti-Bacterial Agents , Pseudomonas aeruginosa , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Tetracycline/pharmacology , Drug Resistance, Microbial , Microbial Sensitivity Tests , Gram-Negative Bacteria
2.
Commun Biol ; 5(1): 1059, 2022 10 05.
Article in English | MEDLINE | ID: mdl-36198902

ABSTRACT

Gram-negative porins are the main entry for small hydrophilic molecules. We studied translocation of structurally related cephalosporins, ceftazidime (CAZ), cefotaxime (CTX) and cefepime (FEP). CAZ is highly active on E. coli producing OmpF (Outer membrane protein F) but less efficient on cells expressing OmpC (Outer membrane protein C), whereas FEP and CTX kill bacteria regardless of the porin expressed. This matches with the different capacity of CAZ and FEP to accumulate into bacterial cells as quantified by LC-MS/MS (Liquid Chromatography Tandem Mass Spectrometry). Furthermore, porin reconstitution into planar lipid bilayer and zero current assays suggest permeation of ≈1,000 molecules of CAZ per sec and per channel through OmpF versus ≈500 through OmpC. Here, the instant killing is directly correlated to internal drug concentration. We propose that the net negative charge of CAZ represents a key advantage for permeation through OmpF porins that are less cation-selective than OmpC. These data could explain the decreased susceptibility to some cephalosporins of enterobacteria that exclusively express OmpC porins.


Subject(s)
Cephalosporins , Enterobacteriaceae , Cefepime/metabolism , Cefotaxime/metabolism , Ceftazidime , Cephalosporins/pharmacology , Chromatography, Liquid , Escherichia coli/metabolism , Lipid Bilayers/metabolism , Monobactams/metabolism , Porins/chemistry , Porins/metabolism , Tandem Mass Spectrometry
3.
Microbiology (Reading) ; 166(8): 759-776, 2020 08.
Article in English | MEDLINE | ID: mdl-32490790

ABSTRACT

Bacterial lipoproteins are secreted proteins that are post-translationally lipidated. Following synthesis, preprolipoproteins are transported through the cytoplasmic membrane via the Sec or Tat translocon. As they exit the transport machinery, they are recognized by a phosphatidylglycerol::prolipoprotein diacylglyceryl transferase (Lgt), which converts them to prolipoproteins by adding a diacylglyceryl group to the sulfhydryl side chain of the invariant Cys+1 residue. Lipoprotein signal peptidase (LspA or signal peptidase II) subsequently cleaves the signal peptide, liberating the α-amino group of Cys+1, which can eventually be further modified. Here, we identified the lgt and lspA genes from Corynebacterium glutamicum and found that they are unique but not essential. We found that Lgt is necessary for the acylation and membrane anchoring of two model lipoproteins expressed in this species: MusE, a C. glutamicum maltose-binding lipoprotein, and LppX, a Mycobacterium tuberculosis lipoprotein. However, Lgt is not required for these proteins' signal peptide cleavage, or for LppX glycosylation. Taken together, these data show that in C. glutamicum the association of some lipoproteins with membranes through the covalent attachment of a lipid moiety is not essential for further post-translational modification.


Subject(s)
Corynebacterium glutamicum/enzymology , Lipoproteins/metabolism , Transferases/metabolism , Acylation , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Membrane/metabolism , Corynebacterium glutamicum/genetics , Corynebacterium glutamicum/growth & development , Corynebacterium glutamicum/metabolism , Genetic Complementation Test , Maltose/metabolism , Mutation , Mycobacterium tuberculosis/genetics , Protein Processing, Post-Translational , Protein Sorting Signals , Transferases/genetics
4.
Commun Biol ; 3(1): 198, 2020 04 28.
Article in English | MEDLINE | ID: mdl-32346058

ABSTRACT

With the spreading of antibiotic resistance, the translocation of antibiotics through bacterial envelopes is crucial for their antibacterial activity. In Gram-negative bacteria, the interplay between membrane permeability and drug efflux pumps must be investigated as a whole. Here, we quantified the intracellular accumulation of a series of fluoroquinolones in population and in individual cells of Escherichia coli according to the expression of the AcrB efflux transporter. Computational results supported the accumulation levels measured experimentally and highlighted how fluoroquinolones side chains interact with specific residues of the distal pocket of the AcrB tight monomer during recognition and binding steps.


Subject(s)
Anti-Bacterial Agents/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Fluoroquinolones/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Anti-Bacterial Agents/pharmacology , Binding Sites , Biological Transport , Drug Resistance, Bacterial , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Fluoroquinolones/pharmacology , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Multidrug Resistance-Associated Proteins/chemistry , Multidrug Resistance-Associated Proteins/genetics , Protein Binding , Spectrometry, Fluorescence , Structure-Activity Relationship
5.
Article in English | MEDLINE | ID: mdl-32229490

ABSTRACT

The Cpx stress response is widespread among Enterobacteriaceae We previously reported a mutation in cpxA in a multidrug-resistant strain of Klebsiella aerogenes isolated from a patient treated with imipenem. This mutation yields a single-amino-acid substitution (Y144N) located in the periplasmic sensor domain of CpxA. In this work, we sought to characterize this mutation in Escherichia coli by using genetic and biochemical approaches. Here, we show that cpxAY144N is an activated allele that confers resistance to ß-lactams and aminoglycosides in a CpxR-dependent manner, by regulating the expression of the OmpF porin and the AcrD efflux pump, respectively. We also demonstrate the effect of the intimate interconnection between the Cpx system and peptidoglycan integrity on the expression of an exogenous AmpC ß-lactamase by using imipenem as a cell wall-active antibiotic or by inactivating penicillin-binding proteins. Moreover, our data indicate that the Y144N substitution abrogates the interaction between CpxA and CpxP and increases phosphotransfer activity on CpxR. Because the addition of a strong AmpC inducer such as imipenem is known to cause abnormal accumulation of muropeptides (disaccharide-pentapeptide and N-acetylglucosamyl-1,6-anhydro-N-acetylmuramyl-l-alanyl-d-glutamy-meso-diaminopimelic-acid-d-alanyl-d-alanine) in the periplasmic space, we propose these molecules activate the Cpx system by displacing CpxP from the sensor domain of CpxA. Altogether, these data could explain why large perturbations to peptidoglycans caused by imipenem lead to mutational activation of the Cpx system and bacterial adaptation through multidrug resistance. These results also validate the Cpx system, in particular, the interaction between CpxA and CpxP, as a promising therapeutic target.


Subject(s)
Escherichia coli Proteins , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Drug Resistance, Microbial , Enterobacteriaceae/genetics , Enterobacteriaceae/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial , Homeostasis , Humans , Protein Kinases/genetics , beta-Lactams/pharmacology
6.
Mol Microbiol ; 114(1): 1-16, 2020 07.
Article in English | MEDLINE | ID: mdl-32073722

ABSTRACT

The genomes of Corynebacteriales contain several genes encoding mycoloyltransferases (Myt) that are specific cell envelope enzymes essential for the biogenesis of the outer membrane. MytA is a major mycoloyltransferase of Corynebacterium glutamicum, displaying an N-terminal domain with esterase activity and a C-terminal extension containing a conserved repeated Leu-Gly-Phe-Pro (LGFP) sequence motif of unknown function. This motif is highly conserved in Corynebacteriales and found associated with cell wall hydrolases and with proteins of unknown function. In this study, we determined the crystal structure of MytA and found that its C-terminal domain is composed of five LGFP motifs and forms a long stalk perpendicular to the N-terminal catalytic α/ß-hydrolase domain. The LGFP motifs are composed of a 4-stranded ß-fold and occupy alternating orientations along the axis of the stalk. Multiple acetate binding pockets were identified in the stalk, which could correspond to putative ligand-binding sites. By using various MytA mutants and complementary in vitro and in vivo approaches, we provide evidence that the C-terminal LGFP domain interacts with the cell wall peptidoglycan-arabinogalactan polymer. We also show that the C-terminal LGFP domain is not required for the activity of MytA but rather contributes to the overall integrity of the cell envelope.


Subject(s)
Acyltransferases/metabolism , Bacterial Outer Membrane/metabolism , Cell Wall/metabolism , Corynebacterium glutamicum/metabolism , Protein Domains/physiology , Acyltransferases/genetics , Binding Sites/physiology , Corynebacterium glutamicum/genetics , Crystallography, X-Ray , Galactans/metabolism , Mycolic Acids/metabolism , Oligopeptides/metabolism , Peptidoglycan/metabolism , Protein Conformation
7.
RSC Chem Biol ; 1(5): 395-404, 2020 Dec 01.
Article in English | MEDLINE | ID: mdl-34458770

ABSTRACT

The emerging crisis of antibiotic resistance requires a multi-pronged approach in order to avert the onset of a post-antibiotic age. Studies of antibiotic uptake and localisation in live cells may inform the design of improved drugs and help develop a better understanding of bacterial resistance and persistence. To facilitate this research, we have synthesised fluorescent derivatives of the macrolide antibiotic erythromycin. These analogues exhibit a similar spectrum of antibiotic activity to the parent drug and are capable of labelling both Gram-positive and -negative bacteria for microscopy. The probes localise intracellularly, with uptake in Gram-negative bacteria dependent on the level of efflux pump activity. A plate-based assay established to quantify bacterial labelling and localisation demonstrated that the probes were taken up by both susceptible and resistant bacteria. Significant intra-strain and -species differences were observed in these preliminary studies. In order to examine uptake in real-time, the probe was used in single-cell microfluidic microscopy, revealing previously unseen heterogeneity of uptake in populations of susceptible bacteria. These studies illustrate the potential of fluorescent macrolide probes to characterise and explore drug uptake and efflux in bacteria.

8.
Nat Rev Microbiol ; 18(3): 164-176, 2020 03.
Article in English | MEDLINE | ID: mdl-31792365

ABSTRACT

Gram-negative bacteria and their complex cell envelope, which comprises an outer membrane and an inner membrane, are an important and attractive system for studying the translocation of small molecules across biological membranes. In the outer membrane of Enterobacteriaceae, trimeric porins control the cellular uptake of small molecules, including nutrients and antibacterial agents. The relatively slow porin-mediated passive uptake across the outer membrane and active efflux via efflux pumps in the inner membrane creates a permeability barrier. The synergistic action of outer membrane permeability, efflux pump activities and enzymatic degradation efficiently reduces the intracellular concentrations of small molecules and contributes to the emergence of antibiotic resistance. In this Review, we discuss recent advances in our understanding of the molecular and functional roles of general porins in small-molecule translocation in Enterobacteriaceae and consider the crucial contribution of porins in antibiotic resistance.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Cell Membrane/metabolism , Enterobacteriaceae/enzymology , Enterobacteriaceae/metabolism , Porins/metabolism , Anti-Bacterial Agents/metabolism , Biological Transport , Drug Resistance, Bacterial , Enterobacteriaceae/drug effects
9.
Medchemcomm ; 10(6): 901-906, 2019 Jun 01.
Article in English | MEDLINE | ID: mdl-31303987

ABSTRACT

Fluorescent probes derived from the fluoroquinolone antibiotic ciprofloxacin were synthesised using a Cu(i)-catalysed azide-alkyne cycloaddition (CuAAC) to link a ciprofloxacin azide derivative with alkyne-substituted green and blue fluorophores. The azide (2) and fluorophore (3 and 4) derivatives retained antimicrobial activity against Gram-positive and Gram-negative bacteria. The use of confocal fluorescent microscopy showed intracellular penetration, which was substantially enhanced in the presence of carbonyl cyanide 3-chlorophenylhydrazone as an efflux pump inhibitor in Escherichia coli.

10.
Subcell Biochem ; 92: 79-123, 2019.
Article in English | MEDLINE | ID: mdl-31214985

ABSTRACT

The transport of small molecules across membranes is essential for the import of nutrients and other energy sources into the cell and, for the export of waste and other potentially harmful byproducts out of the cell. While hydrophobic molecules are permeable to membranes, ions and other small polar molecules require transport via specialized membrane transport proteins . The two major classes of membrane transport proteins are transporters and channels. With our focus here on porins-major class of non-specific diffusion channel proteins , we will highlight some recent structural biology reports and functional assays that have substantially contributed to our understanding of the mechanism that mediates uptake of small molecules, including antibiotics, across the outer membrane of Enterobacteriaceae . We will also review advances in the regulation of porin expression and porin biogenesis and discuss these pathways as new therapeutic targets.


Subject(s)
Porins/metabolism , Anti-Bacterial Agents/metabolism , Biological Transport , Enterobacteriaceae/metabolism , Porins/biosynthesis , Porins/genetics
11.
ACS Infect Dis ; 4(10): 1487-1498, 2018 10 12.
Article in English | MEDLINE | ID: mdl-29962203

ABSTRACT

Small, hydrophilic molecules, including most important antibiotics in clinical use, cross the Gram-negative outer membrane through the water-filled channels provided by porins. We have determined the X-ray crystal structures of the principal general porins from three species of Enterobacteriaceae, namely Enterobacter aerogenes, Enterobacter cloacae, and Klebsiella pneumoniae, and determined their antibiotic permeabilities as well as those of the orthologues from Escherichia coli. Starting from the structure of the porins and molecules, we propose a physical mechanism underlying transport and condense it in a computationally efficient scoring function. The scoring function shows good agreement with in vitro penetration data and will enable the screening of virtual databases to identify molecules with optimal permeability through porins and help to guide the optimization of antibiotics with poor permeation.


Subject(s)
Anti-Bacterial Agents/metabolism , Enterobacteriaceae/metabolism , Porins/chemistry , Porins/metabolism , beta-Lactams/metabolism , Anti-Bacterial Agents/chemistry , Cations/metabolism , Cell Membrane Permeability , Crystallography, X-Ray , Facilitated Diffusion , Glycine/metabolism , Libraries, Digital , Liposomes/metabolism , Osmolar Concentration , Protein Multimerization , Static Electricity , beta-Lactams/chemistry
12.
Nat Protoc ; 13(6): 1348-1361, 2018 06.
Article in English | MEDLINE | ID: mdl-29773906

ABSTRACT

The efficacy of antibacterial molecules depends on their capacity to reach inhibitory concentrations in the vicinity of their target. This is particularly challenging for drugs directed against Gram-negative bacteria, which have a complex envelope comprising two membranes and efflux pumps. Precise determination of the bacterial drug content is an essential prerequisite for drug development. Here we describe three approaches that have been developed in our laboratories to quantify drugs accumulated in intact cells by spectrofluorimetry, microspectrofluorimetry, and kinetics microspectrofluorimetry (KMSF). These different procedures provide complementary results that highlight the contribution of membrane-associated mechanisms, including influx through the outer membrane (OM) and efflux, and the importance of the physicochemical properties of the transported drugs for the intracellular concentration of a given antibiotic in a given bacterial population. The three key stages of this protocol are preparation of the bacterial strains in the presence of the antibiotic; preparation of the whole-cell lysates (WCLs) and fluorescence readings; and data analysis, including normalization and quantitation of the intracellular antibiotic fluorescence relative to the internal standard and the antibiotic standard curve, respectively. Fluorimetry is limited to naturally fluorescent or labeled compounds, but in contrast to existing alternative methods such as mass spectrometry, it uniquely allows single-cell analysis. From culture growth to data analysis, the protocol described here takes 5 d.


Subject(s)
Anti-Bacterial Agents/analysis , Bacteria/chemistry , Spectrometry, Fluorescence/methods , Anti-Bacterial Agents/pharmacokinetics , Membranes/metabolism , Single-Cell Analysis/methods
13.
Microbiology (Reading) ; 164(3): 260-267, 2018 03.
Article in English | MEDLINE | ID: mdl-29458656

ABSTRACT

Bacteria have evolved several strategies to survive a myriad of harmful conditions in the environment and in hosts. In Gram-negative bacteria, responses to nutrient limitation, oxidative or nitrosative stress, envelope stress, exposure to antimicrobials and other growth-limiting stresses have been linked to the development of antimicrobial resistance. This results from the activation of protective changes to cell physiology (decreased outer membrane permeability), resistance transporters (drug efflux pumps), resistant lifestyles (biofilms, persistence) and/or resistance mutations (target mutations, production of antibiotic modification/degradation enzymes). In targeting and interfering with essential physiological mechanisms, antimicrobials themselves are considered as stresses to which protective responses have also evolved. In this review, we focus on envelope stress responses that affect the expression of outer membrane porins and their impact on antimicrobial resistance. We also discuss evidences that indicate the role of antimicrobials as signaling molecules in activating envelope stress responses.


Subject(s)
Cell Membrane Permeability , Drug Resistance, Multiple, Bacterial , Enterobacteriaceae/physiology , Stress, Physiological , Anti-Bacterial Agents/pharmacology , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Cell Membrane Permeability/drug effects , Cell Membrane Permeability/genetics , Drug Resistance, Multiple, Bacterial/genetics , Enterobacteriaceae/drug effects , Porins/genetics , Porins/metabolism , Stress, Physiological/drug effects , Stress, Physiological/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
14.
Res Microbiol ; 169(7-8): 432-441, 2018.
Article in English | MEDLINE | ID: mdl-29208490

ABSTRACT

To understand antibiotic resistance in Gram-negative bacteria, a key point is to investigate antibiotic accumulation, which is defined by influx and efflux. Several methods exist to evaluate membrane permeability and efflux pump activity, but they present disadvantages and limitations. An optimized spectrofluorimetric method using intrinsic tryptophan fluorescence as an internal standard, as well as a complementary microfluorimetric assay following time-course accumulation in intact individual cells, have been developed. Comparing the latter population and single cell approaches can lead to an understanding of phenotypic heterogeneity within a population. The two methodologies lead to determination of parameters, concentration, accumulation rates and localization that contribute to emerging concepts (RTC2T, SICAR) with the aim of identifying and detailing antibiotic chemotypes involved in influx/efflux.


Subject(s)
Anti-Bacterial Agents/analysis , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Gram-Negative Bacteria/metabolism , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , Multigene Family , Anti-Bacterial Agents/metabolism , Bacterial Proteins/genetics , Biological Transport , Fluorescence , Gram-Negative Bacteria/chemistry , Gram-Negative Bacteria/genetics , Membrane Transport Proteins/genetics
15.
Antibiotics (Basel) ; 6(4)2017 Dec 06.
Article in English | MEDLINE | ID: mdl-29211019

ABSTRACT

Antibiotic resistant Gram-negative bacteria are a serious threat for public health. The permeation of antibiotics through their outer membrane is largely dependent on porin, changes in which cause reduced drug uptake and efficacy. Escherichia coli produces two major porins, OmpF and OmpC. MicF and MicC are small non-coding RNAs (sRNAs) that modulate the expression of OmpF and OmpC, respectively. In this work, we investigated factors that lead to increased production of MicC. micC promoter region was fused to lacZ, and the reporter plasmid was transformed into E. coli MC4100 and derivative mutants. The response of micC-lacZ to antimicrobials was measured during growth over a 6 h time period. The data showed that the expression of micC was increased in the presence of ß-lactam antibiotics and in an rpoE depleted mutant. Interestingly, the same conditions enhanced the activity of an ompN-lacZ fusion, suggesting a dual transcriptional regulation of micC and the quiescent adjacent ompN. Increased levels of OmpN in the presence of sub-inhibitory concentrations of chemicals could not be confirmed by Western blot analysis, except when analyzed in the absence of the sigma factor σE. We suggest that the MicC sRNA acts together with the σE envelope stress response pathway to control the OmpC/N levels in response to ß-lactam antibiotics.

17.
PLoS One ; 10(9): e0138828, 2015.
Article in English | MEDLINE | ID: mdl-26398358

ABSTRACT

Infections caused by multidrug resistant (MDR) bacteria are a major concern worldwide. Changes in membrane permeability, including decreased influx and/or increased efflux of antibiotics, are known as key contributors of bacterial MDR. Therefore, it is of critical importance to understand molecular mechanisms that link membrane permeability to MDR in order to design new antimicrobial strategies. In this work, we describe genotype-phenotype correlations in Enterobacter aerogenes, a clinically problematic and antibiotic resistant bacterium. To do this, series of clinical isolates have been periodically collected from two patients during chemotherapy with imipenem. The isolates exhibited different levels of resistance towards multiple classes of antibiotics, consistently with the presence or the absence of porins and efflux pumps. Transport assays were used to characterize membrane permeability defects. Simultaneous genome-wide analysis allowed the identification of putative mutations responsible for MDR. The genome of the imipenem-susceptible isolate G7 was sequenced to closure and used as a reference for comparative genomics. This approach uncovered several loci that were specifically mutated in MDR isolates and whose products are known to control membrane permeability. These were omp35 and omp36, encoding the two major porins; rob, encoding a global AraC-type transcriptional activator; cpxA, phoQ and pmrB, encoding sensor kinases of the CpxRA, PhoPQ and PmrAB two-component regulatory systems, respectively. This report provides a comprehensive analysis of membrane alterations relative to mutational steps in the evolution of MDR of a recognized nosocomial pathogen.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Multiple, Bacterial/genetics , Enterobacter aerogenes/genetics , Enterobacteriaceae Infections/microbiology , Imipenem/pharmacology , Anti-Bacterial Agents/therapeutic use , Cell Membrane Permeability , Enterobacter aerogenes/drug effects , Enterobacteriaceae Infections/drug therapy , Evolution, Molecular , Genome, Bacterial , Humans , Imipenem/therapeutic use , Molecular Sequence Data , Sequence Analysis, DNA
18.
J Bacteriol ; 196(1): 121-8, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24142256

ABSTRACT

Proteins called secretins form large multimeric complexes that are essential for macromolecular transit across the outer membrane of Gram-negative bacteria. Evidence suggests that the channels formed by some secretin complexes are not tightly closed, but their permeability properties have not been well characterized. Here, we used cell-free synthesis coupled with spontaneous insertion into liposomes to investigate the permeability of the secretin PulD. Leakage assays using preloaded liposomes indicated that PulD allows the efflux of small fluorescent molecules with a permeation cutoff similar to that of general porins. Other secretins were also found to form similar pores. To define the polypeptide region involved in determining the pore size, we analyzed a collection of PulD variants and studied the roles of gates 1 and 2, which were previously reported to affect the pore size of filamentous phage f1 secretin pIV, in assembly and pore formation. Liposome leakage and a novel in vivo assay showed that replacement of the conserved proline residue at position 443 in PulD by leucine increased the apparent size of the pore. The in vitro approach described here could be used to study the pore properties of membrane proteins whose production in vivo is toxic.


Subject(s)
Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli/chemistry , Escherichia coli/metabolism , Porins/chemistry , Porins/metabolism , Protein Multimerization , Bacterial Outer Membrane Proteins/genetics , DNA Mutational Analysis , Escherichia coli Proteins/genetics , Fluorescent Dyes/metabolism , Liposomes/metabolism , Permeability , Porins/genetics
19.
Open Microbiol J ; 7: 22-33, 2013.
Article in English | MEDLINE | ID: mdl-23569467

ABSTRACT

Antibiotic translocation across membranes of Gram-negative bacteria is a key step for the activity on their specific intracellular targets. Resistant bacteria control their membrane permeability as a first line of defense to protect themselves against external toxic compounds such as antibiotics and biocides. On one hand, resistance to small hydrophilic antibiotics such as ß-lactams and fluoroquinolones frequently results from the « closing ¼ of their way in: the general outer membrane porins. On the other hand, an effective way out for a wide range of antibiotics is provided by TolC-like proteins, which are outer membrane components of multidrug efflux pumps. Accordingly, altered membrane permeability, including porin modifications and/or efflux pumps' overexpression, is always associated to multidrug resistance (MDR) in a number of clinical isolates. Several recent studies have highlighted our current understanding of porins/TolC structures and functions in Enterobacteriaceae. Here, we review the transport of antibiotics through the OmpF/C general porins and the TolC-like channels with regards to recent data on their structure, function, assembly, regulation and contribution to bacterial resistance. Because MDR strains have evolved global strategies to identify and fight our antibiotic arsenal, it is important to constantly update our global knowledge on antibiotic transport.

20.
PLoS One ; 7(9): e46225, 2012.
Article in English | MEDLINE | ID: mdl-23029442

ABSTRACT

BACKGROUND: Due to their contribution to bacterial virulence, lipoproteins and members of the lipoprotein biogenesis pathway represent potent drug targets. Following translocation across the inner membrane, lipoprotein precursors are acylated by lipoprotein diacylglycerol transferase (Lgt), cleaved off their signal peptides by lipoprotein signal peptidase (Lsp) and, in Gram-negative bacteria, further triacylated by lipoprotein N-acyl transferase (Lnt). The existence of an active apolipoprotein N-acyltransferase (Ms-Ppm2) involved in the N-acylation of LppX was recently reported in M. smegmatis. Ms-Ppm2 is part of the ppm operon in which Ppm1, a polyprenol-monophosphomannose synthase, has been shown to be essential in lipoglycans synthesis but whose function in lipoprotein biosynthesis is completely unknown. RESULTS: In order to clarify the role of the ppm operon in lipoprotein biosynthesis, we investigated the post-translational modifications of two model lipoproteins (AmyE and LppX) in C. glutamicum Δppm1 and Δppm2 mutants. Our results show that both proteins are anchored into the membrane and that their N-termini are N-acylated by Cg-Ppm2. The acylated N-terminal peptide of LppX was also found to be modified by hexose moieties. This O-glycosylation is localized in the N-terminal peptide of LppX and disappeared in the Δppm1 mutant. While compromised in the absence of Cg-Ppm2, LppX O-glycosylation could be restored when Cg-Ppm1, Cg-Ppm2 or the homologous Mt-Ppm1 of M. tuberculosis was overexpressed. CONCLUSION: Together, these results show for the first time that Cg-Ppm1 (Ppm synthase) and Cg-Ppm2 (Lnt) operate in a common biosynthetic pathway in which lipoprotein N-acylation and glycosylation are tightly coupled.


Subject(s)
Acyltransferases/metabolism , Bacterial Proteins/metabolism , Corynebacterium glutamicum/genetics , Lipoproteins/metabolism , Operon , Protein Processing, Post-Translational , Acylation , Acyltransferases/genetics , Amino Acid Sequence , Bacterial Proteins/genetics , Chromatography, Liquid , Corynebacterium glutamicum/enzymology , Escherichia coli/genetics , Gene Expression , Glycosylation , Isoenzymes/genetics , Isoenzymes/metabolism , Lipoproteins/genetics , Molecular Sequence Data , Mutation , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spectrometry, Mass, Electrospray Ionization
SELECTION OF CITATIONS
SEARCH DETAIL
...