Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.179
Filter
Add more filters

Publication year range
1.
Cell ; 162(5): 1016-28, 2015 Aug 27.
Article in English | MEDLINE | ID: mdl-26317468

ABSTRACT

Nuclear pore complexes (NPCs) influence gene expression besides their established function in nuclear transport. The TREX-2 complex localizes to the NPC basket and affects gene-NPC interactions, transcription, and mRNA export. How TREX-2 regulates the gene expression machinery is unknown. Here, we show that TREX-2 interacts with the Mediator complex, an essential regulator of RNA Polymerase (Pol) II. Structural and biochemical studies identify a conserved region on TREX-2, which directly binds the Mediator Med31/Med7N submodule. TREX-2 regulates assembly of Mediator with the Cdk8 kinase and is required for recruitment and site-specific phosphorylation of Pol II. Transcriptome and phenotypic profiling confirm that TREX-2 and Med31 are functionally interdependent at specific genes. TREX-2 additionally uses its Mediator-interacting surface to regulate mRNA export suggesting a mechanism for coupling transcription initiation and early steps of mRNA processing. Our data provide mechanistic insight into how an NPC-associated adaptor complex accesses the core transcription machinery.


Subject(s)
Mediator Complex/metabolism , Multiprotein Complexes/metabolism , Nucleocytoplasmic Transport Proteins/chemistry , Nucleocytoplasmic Transport Proteins/metabolism , Porins/chemistry , Porins/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Transcription, Genetic , Amino Acid Sequence , Animals , Humans , Models, Molecular , Molecular Sequence Data , Multiprotein Complexes/chemistry , Nuclear Pore/metabolism , Nucleocytoplasmic Transport Proteins/genetics , Porins/genetics , Promoter Regions, Genetic , Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/metabolism , RNA Polymerase II/metabolism , Ribonucleoproteins/chemistry , Ribonucleoproteins/metabolism , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae Proteins/genetics , Sequence Alignment , Transcriptome , X-Ray Diffraction
2.
Mol Cell ; 82(3): 629-644.e4, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35063132

ABSTRACT

The envelope of Gram-negative bacteria is a vital barrier that must balance protection and nutrient uptake. Small RNAs are crucial regulators of the envelope composition and function. Here, using RIL-seq to capture the Hfq-mediated RNA-RNA interactome in Salmonella enterica, we discover envelope-related riboregulators, including OppX. We show that OppX acts as an RNA sponge of MicF sRNA, a prototypical porin repressor. OppX originates from the 5' UTR of oppABCDF, encoding the major inner-membrane oligopeptide transporter, and sequesters MicF's seed region to derepress the synthesis of the porin OmpF. Intriguingly, OppX operates as a true sponge, storing MicF in an inactive complex without affecting its levels or stability. Conservation of the opp-OppX-MicF-ompF axis in related bacteria suggests that it serves an important mechanism, adjusting envelope porosity to specific transport capacity. These data also highlight the resource value of this Salmonella RNA interactome, which will aid in unraveling RNA-centric regulation in enteric pathogens.


Subject(s)
5' Untranslated Regions , Cell Membrane/genetics , Escherichia coli Proteins/genetics , Host Factor 1 Protein/genetics , RNA, Bacterial/genetics , Salmonella enterica/genetics , Biological Transport , Cell Membrane/metabolism , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial , Host Factor 1 Protein/metabolism , Host-Pathogen Interactions , Permeability , Porins/genetics , Porins/metabolism , RNA, Bacterial/metabolism , RNA-Seq , Salmonella enterica/metabolism , Salmonella enterica/pathogenicity
3.
Nature ; 621(7979): 586-591, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37704725

ABSTRACT

Many animal- and plant-pathogenic bacteria use a type III secretion system to deliver effector proteins into host cells1,2. Elucidation of how these effector proteins function in host cells is critical for understanding infectious diseases in animals and plants3-5. The widely conserved AvrE-family effectors, including DspE in Erwinia amylovora and AvrE in Pseudomonas syringae, have a central role in the pathogenesis of diverse phytopathogenic bacteria6. These conserved effectors are involved in the induction of 'water soaking' and host cell death that are conducive to bacterial multiplication in infected tissues. However, the exact biochemical functions of AvrE-family effectors have been recalcitrant to mechanistic understanding for three decades. Here we show that AvrE-family effectors fold into a ß-barrel structure that resembles bacterial porins. Expression of AvrE and DspE in Xenopus oocytes results in inward and outward currents, permeability to water and osmolarity-dependent oocyte swelling and bursting. Liposome reconstitution confirmed that the DspE channel alone is sufficient to allow the passage of small molecules such as fluorescein dye. Targeted screening of chemical blockers based on the predicted pore size (15-20 Å) of the DspE channel identified polyamidoamine dendrimers as inhibitors of the DspE/AvrE channels. Notably, polyamidoamines broadly inhibit AvrE and DspE virulence activities in Xenopus oocytes and during E. amylovora and P. syringae infections. Thus, we have unravelled the biochemical function of a centrally important family of bacterial effectors with broad conceptual and practical implications in the study of bacterial pathogenesis.


Subject(s)
Bacterial Proteins , Plant Cells , Plant Diseases , Porins , Water , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cell Death , Fluorescein/metabolism , Liposomes/metabolism , Oocytes/metabolism , Oocytes/microbiology , Plant Cells/metabolism , Plant Cells/microbiology , Plant Diseases/microbiology , Porins/chemistry , Porins/metabolism , Protein Folding , Solutions/metabolism , Water/metabolism , Xenopus laevis , Osmolar Concentration
4.
Nature ; 624(7990): 145-153, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37993720

ABSTRACT

Gram-negative antibiotic development has been hindered by a poor understanding of the types of compounds that can accumulate within these bacteria1,2. The presence of efflux pumps and substrate-specific outer-membrane porins in Pseudomonas aeruginosa renders this pathogen particularly challenging3. As a result, there are few antibiotic options for P. aeruginosa infections4 and its many porins have made the prospect of discovering general accumulation guidelines seem unlikely5. Here we assess the whole-cell accumulation of 345 diverse compounds in P. aeruginosa and Escherichia coli. Although certain positively charged compounds permeate both bacterial species, P. aeruginosa is more restrictive compared to E. coli. Computational analysis identified distinct physicochemical properties of small molecules that specifically correlate with P. aeruginosa accumulation, such as formal charge, positive polar surface area and hydrogen bond donor surface area. Mode of uptake studies revealed that most small molecules permeate P. aeruginosa using a porin-independent pathway, thus enabling discovery of general P. aeruginosa accumulation trends with important implications for future antibiotic development. Retrospective antibiotic examples confirmed these trends and these discoveries were then applied to expand the spectrum of activity of a gram-positive-only antibiotic, fusidic acid, into a version that demonstrates a dramatic improvement in antibacterial activity against P. aeruginosa. We anticipate that these discoveries will facilitate the design and development of high-permeating antipseudomonals.


Subject(s)
Anti-Bacterial Agents , Drug Design , Porins , Pseudomonas aeruginosa , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Escherichia coli/metabolism , Microbial Sensitivity Tests , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/metabolism , Retrospective Studies , Static Electricity , Hydrogen Bonding , Fusidic Acid/metabolism , Drug Design/methods
5.
Cell ; 154(3): 596-608, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23911324

ABSTRACT

The mitochondrial outer membrane harbors two protein translocases that are essential for cell viability: the translocase of the outer mitochondrial membrane (TOM) and the sorting and assembly machinery (SAM). The precursors of ß-barrel proteins use both translocases-TOM for import to the intermembrane space and SAM for export into the outer membrane. It is unknown if the translocases cooperate and where the ß-barrel of newly imported proteins is formed. We established a position-specific assay for monitoring ß-barrel formation in vivo and in organello and demonstrated that the ß-barrel was formed and membrane inserted while the precursor was bound to SAM. ß-barrel formation was inhibited by SAM mutants and, unexpectedly, by mutants of the central import receptor, Tom22. We show that the cytosolic domain of Tom22 links TOM and SAM into a supercomplex, facilitating precursor transfer on the intermembrane space side. Our study reveals receptor-mediated coupling of import and export translocases as a means of precursor channeling.


Subject(s)
Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Proteins/metabolism , Protein Transport , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Proteins/chemistry , Mutation , Porins/chemistry , Porins/metabolism , Protein Folding , Protein Structure, Secondary , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics
6.
Nat Methods ; 21(4): 609-618, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38443507

ABSTRACT

Precise identification and quantification of amino acids is crucial for many biological applications. Here we report a copper(II)-functionalized Mycobacterium smegmatis porin A (MspA) nanopore with the N91H substitution, which enables direct identification of all 20 proteinogenic amino acids when combined with a machine-learning algorithm. The validation accuracy reaches 99.1%, with 30.9% signal recovery. The feasibility of ultrasensitive quantification of amino acids was also demonstrated at the nanomolar range. Furthermore, the capability of this system for real-time analyses of two representative post-translational modifications (PTMs), one unnatural amino acid and ten synthetic peptides using exopeptidases, including clinically relevant peptides associated with Alzheimer's disease and cancer neoantigens, was demonstrated. Notably, our strategy successfully distinguishes peptides with only one amino acid difference from the hydrolysate and provides the possibility to infer the peptide sequence.


Subject(s)
Nanopores , Amino Acids/chemistry , Peptides/chemistry , Amino Acid Sequence , Porins/chemistry , Porins/metabolism
7.
Nat Methods ; 21(1): 92-101, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37749214

ABSTRACT

Natural proteins are composed of 20 proteinogenic amino acids and their post-translational modifications (PTMs). However, due to the lack of a suitable nanopore sensor that can simultaneously discriminate between all 20 amino acids and their PTMs, direct sequencing of protein with nanopores has not yet been realized. Here, we present an engineered hetero-octameric Mycobacterium smegmatis porin A (MspA) nanopore containing a sole Ni2+ modification. It enables full discrimination of all 20 proteinogenic amino acids and 4 representative modified amino acids, Nω,N'ω-dimethyl-arginine (Me-R), O-acetyl-threonine (Ac-T), N4-(ß-N-acetyl-D-glucosaminyl)-asparagine (GlcNAc-N) and O-phosphoserine (P-S). Assisted by machine learning, an accuracy of 98.6% was achieved. Amino acid supplement tablets and peptidase-digested amino acids from peptides were also analyzed using this strategy. This capacity for simultaneous discrimination of all 20 proteinogenic amino acids and their PTMs suggests the potential to achieve protein sequencing using this nanopore-based strategy.


Subject(s)
Nanopores , Amino Acids/chemistry , Proteins/metabolism , Porins/chemistry , Porins/metabolism , Peptides/chemistry
8.
Nature ; 598(7882): 657-661, 2021 10.
Article in English | MEDLINE | ID: mdl-34646015

ABSTRACT

Dimeric IgA secreted across mucous membranes in response to nonpathogenic taxa of the microbiota accounts for most antibody production in mammals. Diverse binding specificities can be detected within the polyclonal mucosal IgA antibody response1-10, but limited monoclonal hybridomas have been studied to relate antigen specificity or polyreactive binding to functional effects on microbial physiology in vivo11-17. Here we use recombinant dimeric monoclonal IgAs (mIgAs) to finely map the intestinal plasma cell response to microbial colonization with a single microorganism in mice. We identify a range of antigen-specific mIgA molecules targeting defined surface and nonsurface membrane antigens. Secretion of individual dimeric mIgAs targeting different antigens in vivo showed distinct alterations in the function and metabolism of intestinal bacteria, largely through specific binding. Even in cases in which the same microbial antigen is targeted, microbial metabolic alterations differed depending on IgA epitope specificity. By contrast, bacterial surface coating generally reduced motility and limited bile acid toxicity. The overall intestinal IgA response to a single microbe therefore contains parallel components with distinct effects on microbial carbon-source uptake, bacteriophage susceptibility, motility and membrane integrity.


Subject(s)
Immunoglobulin A, Secretory/immunology , Intestines/immunology , Microbiota/immunology , Plasma Cells/immunology , Animals , Antibodies, Monoclonal/immunology , Antigens, Bacterial/immunology , Escherichia coli , Germ-Free Life , Mice , Mice, Inbred C57BL , Porins/immunology
9.
Mol Cell ; 73(5): 1056-1065.e7, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30738704

ABSTRACT

The mitochondrial inner membrane harbors a large number of metabolite carriers. The precursors of carrier proteins are synthesized in the cytosol and imported into mitochondria by the translocase of the outer membrane (TOM) and the carrier translocase of the inner membrane (TIM22). Molecular chaperones in the cytosol and intermembrane space bind to the hydrophobic precursors to prevent their aggregation. We report that the major metabolite channel of the outer membrane, termed porin or voltage-dependent anion channel (VDAC), promotes efficient import of carrier precursors. Porin interacts with carrier precursors arriving in the intermembrane space and recruits TIM22 complexes, thus ensuring an efficient transfer of the precursors to the inner membrane translocase. Porin channel mutants impaired in metabolite transport are not disturbed in carrier import into mitochondria. We conclude that porin serves distinct functions as outer membrane channel for metabolites and as coupling factor for protein translocation into the inner membrane.


Subject(s)
Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Porins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Antiporters/genetics , Antiporters/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Mitochondria/genetics , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Precursor Protein Import Complex Proteins , Mutation , Porins/genetics , Protein Binding , Protein Transport , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
10.
Mol Cell ; 73(5): 1044-1055.e8, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30738703

ABSTRACT

Mitochondria import nearly all of their resident proteins from the cytosol, and the TOM complex functions as their entry gate. The TOM complex undergoes a dynamic conversion between the majority population of a three-channel gateway ("trimer") and the minor population that lacks Tom22 and has only two Tom40 channels ("dimer"). Here, we found that the porin Por1 acts as a sink to bind newly imported Tom22. This Por1 association thereby modulates Tom22 integration into the TOM complex, guaranteeing formation of the functional trimeric TOM complex. Por1 sequestration of Tom22 dissociated from the trimeric TOM complex also enhances the dimeric TOM complex, which is preferable for the import of TIM40/MIA-dependent proteins into mitochondria. Furthermore, Por1 appears to contribute to cell-cycle-dependent variation of the functional trimeric TOM complex by chaperoning monomeric Tom22, which arises from the cell-cycle-controlled variation of phosphorylated Tom6.


Subject(s)
Carrier Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membranes/metabolism , Porins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Biological Transport , Carrier Proteins/genetics , Cell Cycle , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Mitochondria/genetics , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Precursor Protein Import Complex Proteins , Phosphorylation , Porins/genetics , Protein Binding , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/genetics
11.
PLoS Genet ; 20(6): e1011127, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38829907

ABSTRACT

The cell envelope fortifies bacterial cells against antibiotics and other insults. Species in the Mycobacteriales order have a complex envelope that includes an outer layer of mycolic acids called the mycomembrane (MM) and a cell wall composed of peptidoglycan and arabinogalactan. This envelope architecture is unique among bacteria and contributes significantly to the virulence of pathogenic Mycobacteriales like Mycobacterium tuberculosis. Characterization of pathways that govern envelope biogenesis in these organisms is therefore critical in understanding their biology and for identifying new antibiotic targets. To better understand MM biogenesis, we developed a cell sorting-based screen for mutants defective in the surface exposure of a porin normally embedded in the MM of the model organism Corynebacterium glutamicum. The results revealed a requirement for the conserved σD envelope stress response in porin export and identified MarP as the site-1 protease, respectively, that activate the response by cleaving the membrane-embedded anti-sigma factor. A reporter system revealed that the σD pathway responds to defects in mycolic acid and arabinogalactan biosynthesis, suggesting that the stress response has the unusual property of being induced by activating signals that arise from defects in the assembly of two distinct envelope layers. Our results thus provide new insights into how C. glutamicum and related bacteria monitor envelope integrity and suggest a potential role for members of the σD regulon in protein export to the MM.


Subject(s)
Cell Membrane , Cell Wall , Corynebacterium glutamicum , Mycolic Acids , Sigma Factor , Cell Wall/metabolism , Cell Wall/genetics , Corynebacterium glutamicum/genetics , Corynebacterium glutamicum/metabolism , Mycolic Acids/metabolism , Sigma Factor/metabolism , Sigma Factor/genetics , Cell Membrane/metabolism , Stress, Physiological , Porins/metabolism , Porins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Galactans/metabolism , Gene Expression Regulation, Bacterial , Peptidoglycan/metabolism
12.
Nature ; 585(7823): 129-134, 2020 09.
Article in English | MEDLINE | ID: mdl-32848250

ABSTRACT

Transmembrane channels and pores have key roles in fundamental biological processes1 and in biotechnological applications such as DNA nanopore sequencing2-4, resulting in considerable interest in the design of pore-containing proteins. Synthetic amphiphilic peptides have been found to form ion channels5,6, and there have been recent advances in de novo membrane protein design7,8 and in redesigning naturally occurring channel-containing proteins9,10. However, the de novo design of stable, well-defined transmembrane protein pores that are capable of conducting ions selectively or are large enough to enable the passage of small-molecule fluorophores remains an outstanding challenge11,12. Here we report the computational design of protein pores formed by two concentric rings of α-helices that are stable and monodisperse in both their water-soluble and their transmembrane forms. Crystal structures of the water-soluble forms of a 12-helical pore and a 16-helical pore closely match the computational design models. Patch-clamp electrophysiology experiments show that, when expressed in insect cells, the transmembrane form of the 12-helix pore enables the passage of ions across the membrane with high selectivity for potassium over sodium; ion passage is blocked by specific chemical modification at the pore entrance. When incorporated into liposomes using in vitro protein synthesis, the transmembrane form of the 16-helix pore-but not the 12-helix pore-enables the passage of biotinylated Alexa Fluor 488. A cryo-electron microscopy structure of the 16-helix transmembrane pore closely matches the design model. The ability to produce structurally and functionally well-defined transmembrane pores opens the door to the creation of designer channels and pores for a wide variety of applications.


Subject(s)
Computer Simulation , Genes, Synthetic/genetics , Ion Channels/chemistry , Ion Channels/genetics , Models, Molecular , Synthetic Biology , Cell Line , Cryoelectron Microscopy , Crystallography, X-Ray , Electric Conductivity , Escherichia coli/genetics , Escherichia coli/metabolism , Hydrazines , Ion Channels/metabolism , Ion Transport , Liposomes/metabolism , Patch-Clamp Techniques , Porins/chemistry , Porins/genetics , Porins/metabolism , Protein Engineering , Protein Structure, Secondary , Solubility , Water/chemistry
13.
Nucleic Acids Res ; 52(13): 7429-7436, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-38884270

ABSTRACT

Nanopores are increasingly powerful tools for single molecule sensing, in particular, for sequencing DNA, RNA and peptides. This success has spurred efforts to sequence non-canonical nucleic acid bases and amino acids. While canonical DNA and RNA bases have pKas far from neutral, certain non-canonical bases, natural RNA modifications, and amino acids are known to have pKas near neutral pHs at which nanopore sequencing is typically performed. Previous reports have suggested that the nanopore signal may be sensitive to the protonation state of an individual moiety. We sequenced ion currents with the MspA nanopore using a single stranded DNA containing a single non-canonical DNA base (Z) at various pH conditions. The Z-base has a near-neutral pKa ∼ 7.8. We find that the measured ion current is remarkably sensitive to the protonation state of the Z-base. We demonstrate how nanopores can be used to localize and determine the pKa of individual moieties along a polymer. More broadly, these experiments provide a path to mapping different protonation sites along polymers and give insight in how to optimize sequencing of polymers that contain moieties with near-neutral pKas.


Subject(s)
DNA, Single-Stranded , Nanopores , Hydrogen-Ion Concentration , DNA, Single-Stranded/chemistry , DNA/chemistry , Protons , Porins/chemistry , Porins/genetics , Sequence Analysis, DNA/methods
14.
Nucleic Acids Res ; 52(15): 9161-9173, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-38842944

ABSTRACT

The ompD transcript, encoding an outer membrane porin in Salmonella, harbors a controlling element in its coding region that base-pairs imperfectly with a 'seed' region of the small regulatory RNA (sRNA) MicC. When tagged with the sRNA, the ompD mRNA is cleaved downstream of the pairing site by the conserved endoribonuclease RNase E, leading to transcript destruction. We observe that the sRNA-induced cleavage site is accessible to RNase E in vitro upon recruitment of ompD into the 30S translation pre-initiation complex (PIC) in the presence of the degradosome components. Evaluation of substrate accessibility suggests that the paused 30S PIC presents the mRNA for targeted recognition and degradation. Ribonuclease activity on PIC-bound ompD is critically dependent on the recruitment of RNase E into the multi-enzyme RNA degradosome, and our data suggest a process of substrate capture and handover to catalytic sites within the degradosome, in which sequential steps of seed matching and duplex remodelling contribute to cleavage efficiency. Our findings support a putative mechanism of surveillance at translation that potentially terminates gene expression efficiently and rapidly in response to signals provided by regulatory RNA.


Subject(s)
Endoribonucleases , Multienzyme Complexes , Polyribonucleotide Nucleotidyltransferase , Porins , RNA Helicases , RNA Stability , RNA, Messenger , Endoribonucleases/metabolism , Endoribonucleases/genetics , Polyribonucleotide Nucleotidyltransferase/metabolism , Polyribonucleotide Nucleotidyltransferase/genetics , RNA Helicases/metabolism , RNA Helicases/genetics , Multienzyme Complexes/metabolism , Multienzyme Complexes/genetics , RNA, Messenger/metabolism , RNA, Messenger/genetics , RNA Stability/genetics , Porins/metabolism , Porins/genetics , RNA, Bacterial/metabolism , RNA, Bacterial/genetics , RNA, Small Untranslated/metabolism , RNA, Small Untranslated/genetics , Gene Expression Regulation, Bacterial
15.
PLoS Genet ; 19(3): e1010490, 2023 03.
Article in English | MEDLINE | ID: mdl-36972246

ABSTRACT

Antimicrobial resistance (AMR) remains a major threat to global health. To date, tractable approaches that decipher how AMR emerges within a bacterial population remain limited. Here, we developed a framework that exploits genetic diversity from environmental bacterial populations to decode emergent phenotypes such as AMR. OmpU is a porin that can make up to 60% of the outer membrane of Vibrio cholerae, the cholera pathogen. This porin is directly associated with the emergence of toxigenic clades and confers resistance to numerous host antimicrobials. In this study, we examined naturally occurring allelic variants of OmpU in environmental V. cholerae and established associations that connected genotypic variation with phenotypic outcome. We covered the landscape of gene variability and found that the porin forms two major phylogenetic clusters with striking genetic diversity. We generated 14 isogenic mutant strains, each encoding a unique ompU allele, and found that divergent genotypes lead to convergent antimicrobial resistance profiles. We identified and characterized functional domains in OmpU unique to variants conferring AMR-associated phenotypes. Specifically, we identified four conserved domains that are linked with resistance to bile and host-derived antimicrobial peptides. Mutant strains for these domains exhibit differential susceptibility patterns to these and other antimicrobials. Interestingly, a mutant strain in which we exchanged the four domains of the clinical allele for those of a sensitive strain exhibits a resistance profile closer to a porin deletion mutant. Finally, using phenotypic microarrays, we uncovered novel functions of OmpU and their connection with allelic variability. Our findings highlight the suitability of our approach towards dissecting the specific protein domains associated with the emergence of AMR and can be naturally extended to other bacterial pathogens and biological processes.


Subject(s)
Anti-Infective Agents , Vibrio cholerae , Adhesins, Bacterial/genetics , Bacterial Outer Membrane Proteins/metabolism , Anti-Bacterial Agents/pharmacology , Alleles , Phylogeny , Protein Domains , Drug Resistance, Bacterial/genetics , Vibrio cholerae/genetics , Vibrio cholerae/metabolism , Porins/genetics , Porins/metabolism
16.
J Biol Chem ; 300(3): 105694, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38301890

ABSTRACT

Bacteriocins, which have narrow-spectrum activity and limited adverse effects, are promising alternatives to antibiotics. In this study, we identified klebicin E (KlebE), a small bacteriocin derived from Klebsiella pneumoniae. KlebE exhibited strong efficacy against multidrug-resistant K. pneumoniae isolates and conferred a significant growth advantage to the producing strain during intraspecies competition. A giant unilamellar vesicle leakage assay demonstrated the unique membrane permeabilization effect of KlebE, suggesting that it is a pore-forming toxin. In addition to a C-terminal toxic domain, KlebE also has a disordered N-terminal domain and a globular central domain. Pulldown assays and soft agar overlay experiments revealed the essential role of the outer membrane porin OmpC and the Ton system in KlebE recognition and cytotoxicity. Strong binding between KlebE and both OmpC and TonB was observed. The TonB-box, a crucial component of the toxin-TonB interaction, was identified as the 7-amino acid sequence (E3ETLTVV9) located in the N-terminal region. Further studies showed that a region near the bottom of the central domain of KlebE plays a primary role in recognizing OmpC, with eight residues surrounding this region identified as essential for KlebE toxicity. Finally, based on the discrepancies in OmpC sequences between the KlebE-resistant and sensitive strains, it was found that the 91st residue of OmpC, an aspartic acid residue, is a key determinant of KlebE toxicity. The identification and characterization of this toxin will facilitate the development of bacteriocin-based therapies targeting multidrug-resistant K. pneumoniae infections.


Subject(s)
Bacteriocins , Klebsiella pneumoniae , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Bacteriocins/genetics , Bacteriocins/metabolism , Bacteriocins/pharmacology , Bacteriocins/toxicity , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/metabolism , Porins/genetics , Porins/metabolism , Cell Membrane Permeability/drug effects , Cell Membrane Permeability/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Protein Domains , Drug Resistance, Multiple, Bacterial/drug effects
17.
EMBO J ; 40(21): e108610, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34515361

ABSTRACT

Bacteria deploy weapons to kill their neighbours during competition for resources and to aid survival within microbiomes. Colicins were the first such antibacterial system identified, yet how these bacteriocins cross the outer membrane (OM) of Escherichia coli is unknown. Here, by solving the structures of translocation intermediates via cryo-EM and by imaging toxin import, we uncover the mechanism by which the Tol-dependent nuclease colicin E9 (ColE9) crosses the bacterial OM. We show that threading of ColE9's disordered N-terminal domain through two pores of the trimeric porin OmpF causes the colicin to disengage from its primary receptor, BtuB, and reorganises the translocon either side of the membrane. Subsequent import of ColE9 through the lumen of a single OmpF subunit is driven by the proton-motive force, which is delivered by the TolQ-TolR-TolA-TolB assembly. Our study answers longstanding questions, such as why OmpF is a better translocator than OmpC, and reconciles the mechanisms by which both Tol- and Ton-dependent bacteriocins cross the bacterial outer membrane.


Subject(s)
Bacteriocins/chemistry , Colicins/chemistry , Escherichia coli/metabolism , Porins/chemistry , Bacterial Outer Membrane/chemistry , Bacterial Outer Membrane/metabolism , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Bacteriocins/genetics , Bacteriocins/metabolism , Binding Sites , Colicins/genetics , Colicins/metabolism , Cryoelectron Microscopy , Escherichia coli/chemistry , Escherichia coli/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial , Kinetics , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Models, Molecular , Periplasmic Proteins/chemistry , Periplasmic Proteins/genetics , Periplasmic Proteins/metabolism , Porins/genetics , Porins/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Domains , Protein Interaction Domains and Motifs , Protein Transport , Thermodynamics
18.
J Cell Sci ; 136(20)2023 10 15.
Article in English | MEDLINE | ID: mdl-37772444

ABSTRACT

The malaria-causing parasite, Plasmodium falciparum completely remodels its host red blood cell (RBC) through the export of several hundred parasite proteins, including transmembrane proteins, across multiple membranes to the RBC. However, the process by which these exported membrane proteins are extracted from the parasite plasma membrane for export remains unknown. To address this question, we fused the exported membrane protein, skeleton binding protein 1 (SBP1), with TurboID, a rapid, efficient and promiscuous biotin ligase (SBP1TbID). Using time-resolved proximity biotinylation and label-free quantitative proteomics, we identified two groups of SBP1TbID interactors - early interactors (pre-export) and late interactors (post-export). Notably, two promising membrane-associated proteins were identified as pre-export interactors, one of which possesses a predicted translocon domain, that could facilitate the export of membrane proteins. Further investigation using conditional mutants of these candidate proteins showed that these proteins were essential for asexual growth and localize to the host-parasite interface during early stages of the intraerythrocytic cycle. These data suggest that they might play a role in ushering membrane proteins from the parasite plasma membrane for export to the host RBC.


Subject(s)
Malaria , Plasmodium falciparum , Animals , Humans , Biotinylation , Erythrocytes/metabolism , Malaria/parasitology , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Porins/metabolism , Protein Transport , Protozoan Proteins/genetics , Protozoan Proteins/metabolism
19.
Proc Natl Acad Sci U S A ; 119(20): e2121487119, 2022 05 17.
Article in English | MEDLINE | ID: mdl-35549548

ABSTRACT

In comparison to globular proteins, the spontaneous folding and insertion of ß-barrel membrane proteins are surprisingly slow, typically occurring on the order of minutes. Using single-molecule Förster resonance energy transfer to report on the folding of fluorescently labeled outer membrane protein G we measured the real-time insertion of a ß-barrel membrane protein from an unfolded state. Folding events were rare and fast (<20 ms), occurring immediately upon arrival at the membrane. This combination of infrequent, but rapid, folding resolves this apparent dichotomy between slow ensemble kinetics and the typical timescales of biomolecular folding.


Subject(s)
Bacterial Outer Membrane Proteins , Escherichia coli Proteins , Porins , Bacterial Outer Membrane Proteins/chemistry , Escherichia coli Proteins/chemistry , Fluorescence Resonance Energy Transfer , Porins/chemistry , Protein Conformation, beta-Strand , Protein Folding , Single Molecule Imaging
20.
Proc Natl Acad Sci U S A ; 119(8)2022 02 22.
Article in English | MEDLINE | ID: mdl-35193963

ABSTRACT

Gram-negative bacteria pose a serious public health concern due to resistance to many antibiotics, caused by the low permeability of their outer membrane (OM). Effective antibiotics use porins in the OM to reach the interior of the cell; thus, understanding permeation properties of OM porins is instrumental to rationally develop broad-spectrum antibiotics. A functionally important feature of OM porins is undergoing open-closed transitions that modulate their transport properties. To characterize the molecular basis of these transitions, we performed an extensive set of molecular dynamics (MD) simulations of Escherichia coli OM porin OmpF. Markov-state analysis revealed that large-scale motion of an internal loop, L3, underlies the transition between energetically stable open and closed states. The conformation of L3 is controlled by H bonds between highly conserved acidic residues on the loop and basic residues on the OmpF ß-barrel. Mutation of key residues important for the loop's conformation shifts the equilibrium between open and closed states and regulates translocation of permeants (ions and antibiotics), as observed in the simulations and validated by our whole-cell accumulation assay. Notably, one mutant system G119D, which we find to favor the closed state, has been reported in clinically resistant bacterial strains. Overall, our accumulated ∼200 µs of simulation data (the wild type and mutants) along with experimental assays suggest the involvement of internal loop dynamics in permeability of OM porins and antibiotic resistance in Gram-negative bacteria.


Subject(s)
Anti-Bacterial Agents/metabolism , Drug Resistance, Bacterial/physiology , Porins/metabolism , Anti-Bacterial Agents/pharmacology , Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/metabolism , Drug Resistance, Bacterial/genetics , Escherichia coli/genetics , Gram-Negative Bacteria/metabolism , Microbial Sensitivity Tests , Models, Theoretical , Molecular Dynamics Simulation , Permeability , Porins/physiology , Porins/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL