Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Chem Rev ; 121(9): 5479-5596, 2021 05 12.
Article in English | MEDLINE | ID: mdl-33909410

ABSTRACT

Tripartite efflux pumps and the related type 1 secretion systems (T1SSs) in Gram-negative organisms are diverse in function, energization, and structural organization. They form continuous conduits spanning both the inner and the outer membrane and are composed of three principal components-the energized inner membrane transporters (belonging to ABC, RND, and MFS families), the outer membrane factor channel-like proteins, and linking the two, the periplasmic adaptor proteins (PAPs), also known as the membrane fusion proteins (MFPs). In this review we summarize the recent advances in understanding of structural biology, function, and regulation of these systems, highlighting the previously undescribed role of PAPs in providing a common architectural scaffold across diverse families of transporters. Despite being built from a limited number of basic structural domains, these complexes present a staggering variety of architectures. While key insights have been derived from the RND transporter systems, a closer inspection of the operation and structural organization of different tripartite systems reveals unexpected analogies between them, including those formed around MFS- and ATP-driven transporters, suggesting that they operate around basic common principles. Based on that we are proposing a new integrated model of PAP-mediated communication within the conformational cycling of tripartite systems, which could be expanded to other types of assemblies.


Subject(s)
Gram-Negative Bacteria/metabolism , Membrane Transport Proteins/metabolism , Type I Secretion Systems/metabolism , ATP-Binding Cassette Transporters , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/metabolism , Gram-Negative Bacteria/chemistry , Membrane Transport Proteins/chemistry , Molecular Dynamics Simulation , Protein Conformation , Structure-Activity Relationship , Type I Secretion Systems/chemistry
2.
Mol Microbiol ; 113(2): 521-533, 2020 02.
Article in English | MEDLINE | ID: mdl-31785114

ABSTRACT

The final step in mycolic acid biosynthesis in Mycobacterium tuberculosis is catalysed by mycolyl reductase encoded by the Rv2509 gene. Sequence analysis and homology modelling indicate that Rv2509 belongs to the short-chain fatty acid dehydrogenase/reductase (SDR) family, but with some distinct features that warrant its classification as belonging to a novel family of short-chain dehydrogenases. In particular, the predicted structure revealed a unique α-helical C-terminal region which we demonstrated to be essential for Rv2509 function, though this region did not seem to play any role in protein stabilisation or oligomerisation. We also show that unlike the M. smegmatis homologue which was not essential for growth, Rv2509 was an essential gene in slow-growing mycobacteria. A knockdown strain of the BCG2529 gene, the Rv2509 homologue in Mycobacterium bovis BCG, was unable to grow following the conditional depletion of BCG2529. This conditional depletion also led to a reduction of mature mycolic acid production and accumulation of intermediates derived from 3-oxo-mycolate precursors. Our studies demonstrate novel features of the mycolyl reductase Rv2509 and outline its role in mycobacterial growth, highlighting its potential as a new target for therapies.


Subject(s)
Mycobacterium , Mycolic Acids/metabolism , Oxidoreductases/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Wall/metabolism , Models, Molecular , Mycobacterium/genetics , Mycobacterium/growth & development , Mycobacterium/metabolism , Mycobacterium bovis/genetics , Mycobacterium bovis/growth & development , Mycobacterium bovis/metabolism , Mycobacterium smegmatis/genetics , Mycobacterium smegmatis/growth & development , Mycobacterium smegmatis/metabolism , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/metabolism , Oxidoreductases/genetics , Oxidoreductases/metabolism
3.
J Antimicrob Chemother ; 76(10): 2558-2564, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34278432

ABSTRACT

BACKGROUND: Resistance-nodulation-division (RND) efflux pumps are important mediators of antibiotic resistance. RND pumps, including the principal multidrug efflux pump AcrAB-TolC in Salmonella, are tripartite systems with an inner membrane RND transporter, a periplasmic adaptor protein (PAP) and an outer membrane factor (OMF). We previously identified the residues required for binding between the PAP AcrA and the RND transporter AcrB and have demonstrated that PAPs can function with non-cognate transporters. AcrE and AcrD/AcrF are homologues of AcrA and AcrB, respectively. Here, we show that AcrE can interact with AcrD, which does not possess its own PAP, and establish that the residues previously identified in AcrB binding are also involved in AcrD binding. METHODS: The acrD and acrE genes were expressed in a strain lacking acrABDEF (Δ3RND). PAP residues involved in promiscuous interactions were predicted based on previously defined PAP-RND interactions and corresponding mutations generated in acrA and acrE. Antimicrobial susceptibility of the mutant strains was determined. RESULTS: Co-expression of acrD and acrE significantly decreased susceptibility of the Δ3RND strain to AcrD substrates, showing that AcrE can form a functional complex with AcrD. The substrate profile of Salmonella AcrD differed from that of Escherichia coli AcrD. Mutations targeting the previously defined PAP-RND interaction sites in AcrA/AcrE impaired efflux of AcrD-dependent substrates. CONCLUSIONS: These data indicate that AcrE forms an efflux-competent pump with AcrD and thus presents an alternative PAP for this pump. Mutagenesis of the conserved RND binding sites validates the interchangeability of AcrA and AcrE, highlighting them as potential drug targets for efflux inhibition.


Subject(s)
Periplasmic Proteins , Salmonella typhimurium , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Salmonella typhimurium/genetics , Serogroup
4.
PLoS Pathog ; 15(12): e1008101, 2019 12.
Article in English | MEDLINE | ID: mdl-31877175

ABSTRACT

Active efflux due to tripartite RND efflux pumps is an important mechanism of clinically relevant antibiotic resistance in Gram-negative bacteria. These pumps are also essential for Gram-negative pathogens to cause infection and form biofilms. They consist of an inner membrane RND transporter; a periplasmic adaptor protein (PAP), and an outer membrane channel. The role of PAPs in assembly, and the identities of specific residues involved in PAP-RND binding, remain poorly understood. Using recent high-resolution structures, four 3D sites involved in PAP-RND binding within each PAP protomer were defined that correspond to nine discrete linear binding sequences or "binding boxes" within the PAP sequence. In the important human pathogen Salmonella enterica, these binding boxes are conserved within phylogenetically-related PAPs, such as AcrA and AcrE, while differing considerably between divergent PAPs such as MdsA and MdtA, despite overall conservation of the PAP structure. By analysing these binding sequences we created a predictive model of PAP-RND interaction, which suggested the determinants that may allow promiscuity between certain PAPs, but discrimination of others. We corroborated these predictions using direct phenotypic data, confirming that only AcrA and AcrE, but not MdtA or MsdA, can function with the major RND pump AcrB. Furthermore, we provide functional validation of the involvement of the binding boxes by disruptive site-directed mutagenesis. These results directly link sequence conservation within identified PAP binding sites with functional data providing mechanistic explanation for assembly of clinically relevant RND-pumps and explain how Salmonella and other pathogens maintain a degree of redundancy in efflux mediated resistance. Overall, our study provides a novel understanding of the molecular determinants driving the RND-PAP recognition by bridging the available structural information with experimental functional validation thus providing the scientific community with a predictive model of pump-contacts that could be exploited in the future for the development of targeted therapeutics and efflux pump inhibitors.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/drug effects , Membrane Transport Proteins/drug effects , Adaptor Proteins, Signal Transducing/metabolism , Animals , Bacterial Infections/drug therapy , Bacterial Proteins/drug effects , Bacterial Proteins/metabolism , Biological Transport/drug effects , Carrier Proteins/metabolism , Female , Membrane Transport Proteins/metabolism , Mice, Inbred BALB C , Periplasm/drug effects , Periplasm/metabolism , Salmonella typhimurium/drug effects , Salmonella typhimurium/metabolism
5.
J Virol ; 92(23)2018 12 01.
Article in English | MEDLINE | ID: mdl-30232185

ABSTRACT

Morbilliviruses infect a broad range of mammalian hosts, including ruminants, carnivores, and humans. The recent eradication of rinderpest virus (RPV) and the active campaigns for eradication of the human-specific measles virus (MeV) have raised significant concerns that the remaining morbilliviruses may emerge in so-called vacated ecological niches. Seeking to assess the zoonotic potential of nonhuman morbilliviruses within human populations, we found that peste des petits ruminants virus (PPRV)-the small-ruminant morbillivirus-is restricted at the point of entry into human cells due to deficient interactions with human SLAMF1-the immune cell receptor for morbilliviruses. Using a structure-guided approach, we characterized a single amino acid change, mapping to the receptor-binding domain in the PPRV hemagglutinin (H) protein, which overcomes this restriction. The same mutation allowed escape from some cross-protective, human patient, anti-MeV antibodies, raising concerns that PPRV is a pathogen with zoonotic potential. Analysis of natural variation within human and ovine SLAMF1 also identified polymorphisms that could correlate with disease resistance. Finally, the mechanistic nature of the PPRV restriction was also investigated, identifying charge incompatibility and steric hindrance between PPRV H and human SLAMF1 proteins. Importantly, this research was performed entirely using surrogate virus entry assays, negating the requirement for in situ derivation of a human-tropic PPRV and illustrating alternative strategies for identifying gain-of-function mutations in viral pathogens.IMPORTANCE A significant proportion of viral pandemics occur following zoonotic transmission events, where animal-associated viruses jump species into human populations. In order to provide forewarnings of the emergence of these viruses, it is necessary to develop a better understanding of what determines virus host range, often at the genetic and structural levels. In this study, we demonstrated that the small-ruminant morbillivirus, a close relative of measles, is unable to use human receptors to enter cells; however, a change of a single amino acid in the virus is sufficient to overcome this restriction. This information will be important for monitoring this virus's evolution in the field. Of note, this study was undertaken in vitro, without generation of a fully infectious virus with this phenotype.


Subject(s)
Antibodies, Viral/immunology , Glycoproteins/metabolism , Mutation , Peste-des-Petits-Ruminants/virology , Peste-des-petits-ruminants virus/pathogenicity , Signaling Lymphocytic Activation Molecule Family Member 1/metabolism , Virus Replication , Amino Acid Sequence , Animals , Chlorocebus aethiops , Glycoproteins/chemistry , Glycoproteins/genetics , Glycoproteins/immunology , Humans , Models, Theoretical , Mutagenesis, Site-Directed , Peste-des-Petits-Ruminants/pathology , Peste-des-Petits-Ruminants/transmission , Peste-des-petits-ruminants virus/genetics , Peste-des-petits-ruminants virus/immunology , Protein Conformation , Sequence Homology , Sheep , Signaling Lymphocytic Activation Molecule Family Member 1/chemistry , Signaling Lymphocytic Activation Molecule Family Member 1/genetics , Signaling Lymphocytic Activation Molecule Family Member 1/immunology , Vero Cells
6.
Proc Natl Acad Sci U S A ; 112(11): 3511-6, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25737552

ABSTRACT

The incidence of multidrug-resistant bacterial infections is increasing globally and the need to understand the underlying mechanisms is paramount to discover new therapeutics. The efflux pumps of Gram-negative bacteria have a broad substrate range and transport antibiotics out of the bacterium, conferring intrinsic multidrug resistance (MDR). The genomes of pre- and posttherapy MDR clinical isolates of Salmonella Typhimurium from a patient that failed antibacterial therapy and died were sequenced. In the posttherapy isolate we identified a novel G288D substitution in AcrB, the resistance-nodulation division transporter in the AcrAB-TolC tripartite MDR efflux pump system. Computational structural analysis suggested that G288D in AcrB heavily affects the structure, dynamics, and hydration properties of the distal binding pocket altering specificity for antibacterial drugs. Consistent with this hypothesis, recreation of the mutation in standard Escherichia coli and Salmonella strains showed that G288D AcrB altered substrate specificity, conferring decreased susceptibility to the fluoroquinolone antibiotic ciprofloxacin by increased efflux. At the same time, the substitution increased susceptibility to other drugs by decreased efflux. Information about drug transport is vital for the discovery of new antibacterials; the finding that one amino acid change can cause resistance to some drugs, while conferring increased susceptibility to others, could provide a basis for new drug development and treatment strategies.


Subject(s)
Amino Acid Substitution , Bacterial Proteins/genetics , Drug Resistance, Multiple, Bacterial , Escherichia coli Proteins/genetics , Membrane Transport Proteins/genetics , Multidrug Resistance-Associated Proteins/genetics , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Binding Sites , Ciprofloxacin/pharmacology , Doxorubicin/chemistry , Doxorubicin/metabolism , Drug Resistance, Multiple, Bacterial/drug effects , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli/isolation & purification , Escherichia coli Proteins/metabolism , Genetic Fitness , Genome, Bacterial , Humans , Membrane Transport Proteins/metabolism , Microbial Sensitivity Tests , Minocycline/pharmacology , Models, Molecular , Multidrug Resistance-Associated Proteins/metabolism , Mutation/genetics , Polymorphism, Single Nucleotide/genetics , Salmonella enterica/drug effects , Salmonella enterica/genetics , Salmonella enterica/isolation & purification , Substrate Specificity/drug effects , Water/chemistry
7.
Int J Mol Sci ; 19(4)2018 Mar 27.
Article in English | MEDLINE | ID: mdl-29584668

ABSTRACT

Vibrio cholerae ATP-binding cassette transporter VcaM (V. cholerae ABC multidrug resistance pump) has previously been shown to confer resistance to a variety of medically important drugs. In this study, we set to analyse its properties both in vitro in detergent-solubilised state and in vivo to differentiate its dependency on auxiliary proteins for its function. We report the first detailed kinetic parameters of purified VcaM and the rate of phosphate (Pi) production. To determine the possible functional dependencies of VcaM on the tripartite efflux pumps we then utilized different E. coli strains lacking the principal secondary transporter AcrB (Acriflavine resistance protein), as well as cells lacking the outer membrane factor (OMF) TolC (Tolerance to colicins). Consistent with the ATPase function of VcaM we found it to be susceptible to sodium orthovanadate (NaOV), however, we also found a clear dependency of VcaM function on TolC. Inhibitors targeting secondary active transporters had no effects on either VcaM-conferred resistance or Hoechst 33342 accumulation, suggesting that VcaM might be capable of engaging with the TolC-channel without periplasmic mediation by additional transporters. Our findings are indicative of VcaM being capable of a one-step substrate translocation from cytosol to extracellular space utilising the TolC-channel, making it the only multidrug ABC-transporter outside of the MacB-family with demonstrable TolC-dependency.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Bacterial Outer Membrane Proteins/genetics , Escherichia coli Proteins/genetics , Membrane Transport Proteins/genetics , Multidrug Resistance-Associated Proteins/genetics , Vibrio cholerae/metabolism , ATP-Binding Cassette Transporters/genetics , Adenosine Triphosphate/chemistry , Cloning, Molecular , Cytosol/metabolism , Drug Resistance, Multiple, Bacterial , Escherichia coli/genetics , Escherichia coli/growth & development , Extracellular Space/metabolism , Gene Knockout Techniques , Hydrolysis , Phosphates/metabolism , Vanadates/pharmacology , Vibrio cholerae/genetics
8.
J Bacteriol ; 199(18)2017 09 15.
Article in English | MEDLINE | ID: mdl-28674068

ABSTRACT

The EvgS/EvgA two-component system of Escherichia coli is activated in response to low pH and alkali metals and regulates many genes, including those for the glutamate-dependent acid resistance system and a number of efflux pumps. EvgS, the sensor kinase, is one of five unconventional histidine kinases (HKs) in E. coli and has a large periplasmic domain and a cytoplasmic PAS domain in addition to phospho-acceptor, HK and dimerization, internal receiver, and phosphotransfer domains. Mutations that constitutively activate the protein at pH 7 map to the PAS domain. Here, we built a homology model of the periplasmic region of EvgS, based on the structure of the equivalent region of the BvgS homologue, to guide mutagenesis of potential key residues in this region. We show that histidine 226 is required for induction and that it is structurally colocated with a proline residue (P522) at the top of the predicted transmembrane helix that is expected to play a key role in passing information to the cytoplasmic domains. We also show that the constitutive mutations in the PAS domain can be further activated by low external pH. Expression of the cytoplasmic part of the protein alone also gives constitutive activation, which is lost if the constitutive PAS mutations are present. These findings are consistent with a model in which EvgS senses both external and internal pH and is activated by a shift from a tight inactive to a weak active dimer, and we present an analysis of the purified cytoplasmic portion of EvgS that supports this.IMPORTANCE One of the ways bacteria sense their environment is through two-component systems, which have one membrane-bound protein to do the sensing and another inside the cell to turn genes on or off in response to what the membrane-bound protein has detected. The membrane-bound protein must thus be able to detect the stress and signal this detection event to the protein inside the cell. To understand this process, we studied a protein that helps E. coli to survive exposure to low pH, which it must do before taking up residence in the gastrointestinal tract. We describe a predicted structure for the main sensing part of the protein and identify some key residues within it that are involved in the sensing and signaling processes. We propose a mechanism for how the protein may become activated and present some evidence to support our proposal.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Histidine Kinase/metabolism , Protein Kinases/metabolism , Protein Multimerization , DNA Mutational Analysis , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Histidine Kinase/chemistry , Histidine Kinase/genetics , Hydrogen-Ion Concentration , Protein Conformation , Protein Kinases/chemistry , Protein Kinases/genetics
9.
Mol Microbiol ; 97(4): 646-59, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25943387

ABSTRACT

BAM is a conserved molecular machine, the central component of which is BamA. Orthologues of BamA are found in all Gram-negative bacteria, chloroplasts and mitochondria where it is required for the folding and insertion of ß-barrel containing integral outer membrane proteins (OMPs) into the outer membrane. BamA binds unfolded ß-barrel precursors via the five polypeptide transport-associated (POTRA) domains at its N-terminus. The C-terminus of BamA folds into a ß-barrel domain, which tethers BamA to the outer membrane and is involved in OMP insertion. BamA orthologues are found in all Gram-negative bacteria and appear to function in a species-specific manner. Here we investigate the nature of this species-specificity by examining whether chimeric Escherichia coli BamA fusion proteins, carrying either the ß-barrel or POTRA domains from various BamA orthologues, can functionally replace E. coli BamA. We demonstrate that the ß-barrel domains of many BamA orthologues are functionally interchangeable. We show that defects in the orthologous POTRA domains can be rescued by compensatory mutations within the ß-barrel. These data reveal that the POTRA and barrel domains must be precisely aligned to ensure efficient OMP insertion.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Escherichia coli Proteins/metabolism , Gram-Negative Bacteria/metabolism , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/genetics , Chimera/genetics , Chimera/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Gram-Negative Bacteria/genetics , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Folding , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Species Specificity
10.
Mol Cell ; 30(1): 114-21, 2008 Apr 11.
Article in English | MEDLINE | ID: mdl-18406332

ABSTRACT

Drugs and certain proteins are transported across the membranes of Gram-negative bacteria by energy-activated pumps. The outer membrane component of these pumps is a channel that opens from a sealed resting state during the transport process. We describe two crystal structures of the Escherichia coli outer membrane protein TolC in its partially open state. Opening is accompanied by the exposure of three shallow intraprotomer grooves in the TolC trimer, where our mutagenesis data identify a contact point with the periplasmic component of a drug efflux pump, AcrA. We suggest that the assembly of multidrug efflux pumps is accompanied by induced fit of TolC driven mainly by accommodation of the periplasmic component.


Subject(s)
Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/metabolism , Drug Resistance, Microbial/physiology , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , Multidrug Resistance-Associated Proteins/chemistry , Multidrug Resistance-Associated Proteins/metabolism , Protein Conformation , Bacterial Outer Membrane Proteins/genetics , Biological Transport/physiology , Crystallography, X-Ray , Escherichia coli Proteins/genetics , Lipoproteins , Membrane Transport Proteins/genetics , Models, Molecular , Molecular Sequence Data , Multidrug Resistance-Associated Proteins/genetics , Point Mutation
11.
J Biol Chem ; 289(1): 143-51, 2014 Jan 03.
Article in English | MEDLINE | ID: mdl-24257749

ABSTRACT

KirBac channels are prokaryotic homologs of mammalian inwardly rectifying potassium (Kir) channels, and recent structures of KirBac3.1 have provided important insights into the structural basis of gating in Kir channels. In this study, we demonstrate that KirBac3.1 channel activity is strongly pH-dependent, and we used x-ray crystallography to determine the structural changes that arise from an activatory mutation (S205L) located in the cytoplasmic domain (CTD). This mutation stabilizes a novel energetically favorable open conformation in which changes at the intersubunit interface in the CTD also alter the electrostatic potential of the inner cytoplasmic cavity. These results provide a structural explanation for the activatory effect of this mutation and provide a greater insight into the role of the CTD in Kir channel gating.


Subject(s)
Bacterial Proteins/chemistry , Magnetospirillum/chemistry , Potassium Channels, Inwardly Rectifying/chemistry , Amino Acid Substitution , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Ion Channel Gating/physiology , Magnetospirillum/genetics , Magnetospirillum/metabolism , Mutation, Missense , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels, Inwardly Rectifying/metabolism , Protein Structure, Tertiary
12.
Mol Microbiol ; 91(5): 965-75, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24386963

ABSTRACT

The tripartite AcrAB-TolC multidrug efflux pump of Escherichia coli is the central conduit for cell-toxic compounds and contributes to antibiotic resistance. While high-resolution structures of all three proteins have been solved, much remains to be learned as to how the individual components come together to form a functional complex. In this study, we investigated the importance of the AcrB ß-hairpins belonging to the DN and DC subdomains, which are presumed to dock with TolC, in complex stability and activity of the complete pump. Our data show that the DN subdomain ß-hairpin residues play a more critical role in complex stability and activity than the DC subdomain hairpin residues. The failure of the AcrB DN ß-hairpin deletion mutant to engage with TolC leads to the drug hypersensitivity phenotype, which is reversed by compensatory alterations in the lipoyl and ß-barrel domains of AcrA. Moreover, AcrA and TolC mutants that induce TolC opening also reverse the drug hypersensitivity phenotype of the AcrB ß-hairpin mutants, indicating a failure by the AcrB mutant to interact and thus induce TolC opening on its own. Together, these data suggest that both AcrB ß-hairpins and AcrA act to stabilize the tripartite complex and induce TolC opening for drug expulsion.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli/metabolism , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Amino Acids/metabolism , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Blotting, Western , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli Proteins/metabolism , Membrane Transport Proteins/metabolism , Microbial Sensitivity Tests , Multidrug Resistance-Associated Proteins/chemistry , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Stability , Protein Structure, Secondary , Structure-Activity Relationship , Suppression, Genetic
13.
Biochem Soc Trans ; 43(5): 983-94, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26517913

ABSTRACT

Membrane proteins, such as receptors, transporters and ion channels, control the vast majority of cellular signalling and metabolite exchange processes and thus are becoming key pharmacological targets. Obtaining structural information by usage of traditional structural biology techniques is limited by the requirements for the protein samples to be highly pure and stable when handled in high concentrations and in non-native buffer systems, which is often difficult to achieve for membrane targets. Hence, there is a growing requirement for the use of hybrid, integrative approaches to study the dynamic and functional aspects of membrane proteins in physiologically relevant conditions. In recent years, significant progress has been made in the field of oxidative labelling techniques and in particular the X-ray radiolytic footprinting in combination with mass spectrometry (MS) (XF-MS), which provide residue-specific information on the solvent accessibility of proteins. In combination with both low- and high-resolution data from other structural biology approaches, it is capable of providing valuable insights into dynamics of membrane proteins, which have been difficult to obtain by other structural techniques, proving a highly complementary technique to address structure and function of membrane targets. XF-MS has demonstrated a unique capability for identification of structural waters and conformational changes in proteins at both a high degree of spatial and a high degree of temporal resolution. Here, we provide a perspective on the place of XF-MS among other structural biology methods and showcase some of the latest developments in its usage for studying water-mediated transmembrane (TM) signalling, ion transport and ligand-induced allosteric conformational changes in membrane proteins.


Subject(s)
Mass Spectrometry/methods , Membrane Proteins/chemistry , Protein Footprinting/methods , Protein Structure, Tertiary , Animals , Crystallography, X-Ray , Humans , Membrane Proteins/metabolism , Models, Molecular , Oxidation-Reduction , Protein Binding
14.
Mol Microbiol ; 88(3): 590-602, 2013 May.
Article in English | MEDLINE | ID: mdl-23565750

ABSTRACT

The MtrCDE multidrug pump, from Neisseria gonorrhoeae, is assembled from the inner and outer membrane proteins MtrD and MtrE, which are connected by the periplasmic membrane fusion protein MtrC. Although it is clear that MtrD delivers drugs to the channel of MtrE, it remains unclear how drug delivery and channel opening are connected. We used a vancomycin sensitivity assay to test for opening of the MtrE channel. Cells expressing MtrE or MtrE-E434K were insensitive to vancomycin; but became moderately and highly sensitive to vancomycin respectively, when coexpressed with MtrC, suggesting that the MtrE channel opening requires MtrC binding and is energy-independent. Cells expressing wild-type MtrD, in an MtrCE background, were vancomycin-insensitive, but moderately sensitive in an MtrCE-E434K background. The mutation of residues involved in proton translocation inactivated MtrD and abolished drug efflux, rendered both MtrE and MtrE-E434K vancomycin-insensitive; imply that the pump-component interactions are preserved, and that the complex is stable in the absence of proton flux, thus sealing the open end of MtrE. Following the energy-dependent dissociation of the tripartite complex, the MtrE channel is able to reseal, while MtrE-E434K is unable to do so, resulting in the vancomycin-sensitive phenotype. Thus, our findings suggest that opening of the OMP via interaction with the MFP is energy-independent, while both drug export and complex dissociation require active proton flux.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Lipoproteins/metabolism , Membrane Transport Proteins/metabolism , Neisseria gonorrhoeae/genetics , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/genetics , Drug Resistance, Multiple, Bacterial/genetics , Escherichia coli/genetics , Escherichia coli/growth & development , Lipoproteins/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Transport Proteins/genetics , Mutation , Neisseria gonorrhoeae/drug effects , Neisseria gonorrhoeae/metabolism , Plasmids/genetics , Vancomycin/pharmacology
15.
Microb Genom ; 9(3)2023 03.
Article in English | MEDLINE | ID: mdl-36995182

ABSTRACT

Acinetobacter are generally soil-dwelling organisms that can also cause serious human infections. A. baumannii is one of the most common causative agents of Acinetobacter infections and is often multidrug resistant. However, an additional 25 species within the genus have also been associated with infection. A. baumannii encodes six resistance nodulation division (RND) efflux pumps, the most clinically relevant class of efflux pumps for antibiotic export, but the distribution and types of RND efflux pumps across the genus is currently unknown. Sixty-four species making up the genus Acinetobacter were searched for RND systems within their genomes. We also developed a novel method using conserved RND residues to predict the total number of RND proteins including currently undescribed RND pump proteins. The total number of RND proteins differed both within a species and across the genus. Species associated with infection tended to encode more pumps. AdeIJK/AdeXYZ was found in all searched species of Acinetobacter, and through genomic, structural and phenotypic work we show that these genes are actually homologues of the same system. This interpretation is further supported by structural analysis of the potential drug-binding determinants of the associated RND-transporters, which reveal their close similarity to each other, and distinctiveness from other RND-pumps in Acinetobacter, such as AdeB. Therefore, we conclude that AdeIJK is the fundamental RND system for species in the genus Acinetobacter. AdeIJK can export a broad range of antibiotics and provides crucial functions within the cell, for example lipid modulation of the cell membrane, and therefore it is likely that all Acinetobacter require AdeIJK for survival and homeostasis. In contrast, additional RND systems, such as AdeABC and AdeFGH, were only found in a subset of Acinetobacter that are associated with infection. By understanding the roles and mechanisms of RND efflux systems in Acinetobacter, treatments for infections can avoid efflux-mediated resistance and improve patient outcomes.


Subject(s)
Acinetobacter Infections , Acinetobacter baumannii , Humans , Acinetobacter baumannii/genetics , Drug Resistance, Multiple, Bacterial/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism
16.
NPJ Antimicrob Resist ; 1(1): 2, 2023.
Article in English | MEDLINE | ID: mdl-38686215

ABSTRACT

Antibiotic resistance is a pressing healthcare challenge and is mediated by various mechanisms, including the active export of drugs via multidrug efflux systems, which prevent drug accumulation within the cell. Here, we studied how Salmonella evolved resistance to two key antibiotics, cefotaxime and azithromycin, when grown planktonically or as a biofilm. Resistance to both drugs emerged in both conditions and was associated with different substitutions within the efflux-associated transporter, AcrB. Azithromycin exposure selected for an R717L substitution, while cefotaxime for Q176K. Additional mutations in ramR or envZ accumulated concurrently with the R717L or Q176K substitutions respectively, resulting in clinical resistance to the selective antibiotics and cross-resistance to other drugs. Structural, genetic, and phenotypic analysis showed the two AcrB substitutions confer their benefits in profoundly different ways. R717L reduces steric barriers associated with transit through the substrate channel 2 of AcrB. Q176K increases binding energy for cefotaxime, improving recognition in the distal binding pocket, resulting in increased efflux efficiency. Finally, we show the R717 substitution is present in isolates recovered around the world.

17.
J Biol Chem ; 286(30): 26900-12, 2011 Jul 29.
Article in English | MEDLINE | ID: mdl-21610073

ABSTRACT

The multiple transferable resistance (mTR) pump from Neisseria gonorrhoeae MtrCDE multidrug pump is assembled from the inner and outer membrane proteins MtrD and MtrE and the periplasmic membrane fusion protein MtrC. Previously we established that while there is a weak interaction of MtrD and MtrE, MtrC binds with relatively high affinity to both MtrD and MtrE. MtrD conferred antibiotic resistance only when it was expressed with MtrE and MtrC, suggesting that these proteins form a functional tripartite complex in which MtrC bridges MtrD and MtrE. Furthermore, we demonstrated that MtrC interacts with an intraprotomer groove on the surface of MtrE, inducing channel opening. However, a second groove is apparent at the interface of the MtrE subunits, which might also be capable of engaging MtrC. We have now established that MtrC can be cross-linked to cysteines placed in this interprotomer groove and that mutation of residues in the groove impair the ability of the pump to confer antibiotic resistance by locking MtrE in the closed channel conformation. Moreover, MtrE K390C forms an intermolecular disulfide bond with MtrC E149C locking MtrE in the open channel conformation, suggesting that a functional salt bridge forms between these residues during the transition from closed to open channel conformations. MtrC forms dimers that assemble into hexamers, and electron microscopy studies of single particles revealed that these hexamers are arranged into ring-like structures with an internal aperture sufficiently large to accommodate the MtrE trimer. Cross-linking of single cysteine mutants of MtrC to stabilize the dimer interface in the presence of MtrE, trapped an MtrC-MtrE complex with a molecular mass consistent with a stoichiometry of 3:6 (MtrE(3)MtrC(6)), suggesting that dimers of MtrC interact with MtrE, presumably by binding to the two grooves. As both MtrE and MtrD are trimeric, our studies suggest that the functional pump is assembled with a stoichiometry of 3:6:3.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/metabolism , Drug Resistance, Multiple, Bacterial/physiology , Lipoproteins/metabolism , Membrane Proteins/metabolism , Membrane Transport Proteins/metabolism , Multiprotein Complexes/metabolism , Neisseria gonorrhoeae/metabolism , Amino Acid Substitution , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/genetics , Lipoproteins/genetics , Membrane Proteins/genetics , Membrane Transport Proteins/genetics , Multiprotein Complexes/genetics , Mutation, Missense , Neisseria gonorrhoeae/genetics , Neisseria gonorrhoeae/ultrastructure , Protein Binding , Protein Structure, Quaternary
18.
J Biol Chem ; 286(7): 5484-93, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21115481

ABSTRACT

The multiple transferable resistance (MTR) pump, from Neisseria gonorrhoeae, is typical of the specialized machinery used to translocate drugs across the inner and outer membranes of Gram-negative bacteria. It consists of a tripartite complex composed of an inner-membrane transporter, MtrD, a periplasmic membrane fusion protein, MtrC, and an outer-membrane channel, MtrE. We have expressed the components of the pump in Escherichia coli and used the antibiotic vancomycin, which is too large to cross the outer-membrane by passive diffusion, to test for opening of the MtrE channel. Cells expressing MtrCDE are not susceptible to vancomycin, indicating that the channel is closed; but become susceptible to vancomycin in the presence of transported substrates, consistent with drug-induced opening of the MtrE channel. A mutational analysis identified residues Asn-198, Glu-434, and Gln-441, lining an intraprotomer groove on the surface of MtrE, to be important for pump function; mutation of these residues yielded cells that were sensitive to vancomycin. Pull-down assays and micro-calorimetry measurements indicated that this functional impairment is not due to the inability of MtrC to interact with the MtrE mutants; nor was it due to the MtrE mutants adopting an open conformation, because cells expressing these MtrE mutants alone are relatively insensitive to vancomycin. However, cells expressing the MtrE mutants with MtrC are sensitive to vancomycin, indicating that residues lining the intra-protomer groove control opening of the MtrE channel in response to binding of MtrC.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/metabolism , Drug Resistance, Bacterial/physiology , Lipoproteins/metabolism , Membrane Proteins/metabolism , Membrane Transport Proteins/metabolism , Neisseria gonorrhoeae/metabolism , Amino Acid Substitution , Anti-Bacterial Agents/pharmacology , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/genetics , Escherichia coli , Gene Expression , Lipoproteins/genetics , Membrane Proteins/genetics , Membrane Transport Proteins/genetics , Mutation, Missense , Neisseria gonorrhoeae/genetics , Protein Binding , Protein Structure, Quaternary , Protein Structure, Tertiary , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Vancomycin/pharmacology
19.
Nat Methods ; 6(8): 585-7, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19578383

ABSTRACT

We describe a general mass spectrometry approach to determine subunit stoichiometry and lipid binding in intact membrane protein complexes. By exploring conditions for preserving interactions during transmission into the gas phase and for optimally stripping away detergent, by subjecting the complex to multiple collisions, we released the intact complex largely devoid of detergent. This enabled us to characterize both subunit stoichiometry and lipid binding in 4 membrane protein complexes.


Subject(s)
Membrane Transport Proteins/chemistry , Multiprotein Complexes/chemistry , Protein Subunits/chemistry , Proteomics/methods , Spectrometry, Mass, Electrospray Ionization/methods , Protein Interaction Mapping
20.
Sci Rep ; 12(1): 4752, 2022 03 19.
Article in English | MEDLINE | ID: mdl-35306531

ABSTRACT

Tripartite resistance-nodulation-division (RND) efflux pumps, such as AcrAB-TolC of Salmonella Typhimurium, contribute to antibiotic resistance and comprise an inner membrane RND-transporter, an outer membrane factor, and a periplasmic adaptor protein (PAP). The role of the PAP in the assembly and active transport process remains poorly understood. Here, we identify the functionally critical residues involved in PAP-RND-transporter binding between AcrA and AcrB and show that the corresponding RND-binding residues in the closely related PAP AcrE, are also important for its interaction with AcrB. We also report a residue in the membrane-proximal domain of AcrA, that when mutated, differentially affects the transport of substrates utilising different AcrB efflux channels, namely channels 1 and 2. This supports a potential role for the PAP in sensing the substrate-occupied state of the proximal binding pocket of the transporter and substrate vetting. Understanding the PAP's role in the assembly and function of tripartite RND pumps can guide novel ways to inhibit their function to combat antibiotic resistance.


Subject(s)
Escherichia coli Proteins , Membrane Transport Proteins , Adaptor Proteins, Signal Transducing/metabolism , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Bacterial Outer Membrane Proteins/metabolism , Biological Transport , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Periplasm/metabolism , Salmonella typhimurium/genetics , Salmonella typhimurium/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL