Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
J Bacteriol ; 205(6): e0003523, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37219427

ABSTRACT

The outer membranes (OM) of Gram-negative bacteria contain a class of proteins (TBDTs) that require energy for the import of nutrients and to serve as receptors for phages and protein toxins. Energy is derived from the proton motif force (pmf) of the cytoplasmic membrane (CM) through the action of three proteins, namely, TonB, ExbB, and ExbD, which are located in the CM and extend into the periplasm. The leaky phenotype of exbB exbD mutants is caused by partial complementation by homologous tolQ tolR. TonB, ExbB, and ExbD are genuine components of an energy transmission system from the CM into the OM. Mutant analyses, cross-linking experiments, and most recently X-ray and cryo-EM determinations were undertaken to arrive at a model that describes the energy transfer from the CM into the OM. These results are discussed in this paper. ExbB forms a pentamer with a pore inside, in which an ExbD dimer resides. This complex harvests the energy of the pmf and transmits it to TonB. TonB interacts with the TBDT at the TonB box, which triggers a conformational change in the TBDT that releases bound nutrients and opens the pore, through which nutrients pass into the periplasm. The structurally altered TBDT also changes the interactions of its periplasmic signaling domain with anti-sigma factors, with the consequence being that the sigma factors initiate transcription.


Subject(s)
Escherichia coli Proteins , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Escherichia coli/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Cell Membrane/metabolism , Biological Transport , Bacterial Proteins/genetics , Bacterial Proteins/metabolism
2.
J Bacteriol ; 202(7)2020 03 11.
Article in English | MEDLINE | ID: mdl-31932309

ABSTRACT

The TonB system actively transports vital nutrients across the unenergized outer membranes of the majority of Gram-negative bacteria. In this system, integral membrane proteins ExbB, ExbD, and TonB work together to transduce the proton motive force (PMF) of the inner membrane to customized active transporters in the outer membrane by direct and cyclic binding of TonB to the transporters. A PMF-dependent TonB-ExbD interaction is prevented by 10-residue deletions within a periplasmic disordered domain of ExbD adjacent to the cytoplasmic membrane. Here, we explored the function of the ExbD disordered domain in more detail. In vivo photo-cross-linking through sequential pBpa substitutions in the ExbD disordered domain captured five different ExbD complexes, some of which had been previously detected using in vivo formaldehyde cross-linking, a technique that lacks the residue-specific information that can be achieved through photo-cross-linking: two ExbB-ExbD heterodimers (one of which had not been detected previously), previously detected ExbD homodimers, previously detected PMF-dependent ExbD-TonB heterodimers, and for the first time, a predicted, ExbD-TonB PMF-independent interaction. The fact that multiple complexes were captured by the same pBpa substitution indicated the dynamic nature of ExbD interactions as the energy transduction cycle proceeded in vivo In this study, we also discovered that a conserved motif-V45, V47, L49, and P50-within the disordered domain was required for signal transduction to TonB and to the C-terminal domain of ExbD and was the source of motif essentiality.IMPORTANCE The TonB system is a virulence factor for Gram-negative pathogens. The mechanism by which cytoplasmic membrane proteins of the TonB system transduce an electrochemical gradient into mechanical energy is a long-standing mystery. TonB, ExbB, and ExbD primary amino acid sequences are characterized by regions of predicted intrinsic disorder, consistent with a proposed multiplicity of protein-protein contacts as TonB proceeds through an energy transduction cycle, a complex process that has yet to be recapitulated in vitro This study validates a region of intrinsic disorder near the ExbD transmembrane domain and identifies an essential conserved motif embedded within it that transduces signals to distal regions of ExbD suggested to configure TonB for productive interaction with outer membrane transporters.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Intrinsically Disordered Proteins/metabolism , Signal Transduction , Amino Acid Motifs , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Escherichia coli Proteins/chemistry , Intrinsically Disordered Proteins/chemistry , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Models, Biological , Models, Molecular , Multiprotein Complexes , Position-Specific Scoring Matrices , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Structure-Activity Relationship
3.
Int J Mol Sci ; 21(2)2020 Jan 07.
Article in English | MEDLINE | ID: mdl-31936081

ABSTRACT

The Ton complex is a molecular motor that uses the proton gradient at the inner membrane of Gram-negative bacteria to generate force and movement, which are transmitted to transporters at the outer membrane, allowing the entry of nutrients into the periplasmic space. Despite decades of investigation and the recent flurry of structures being reported by X-ray crystallography and cryoEM, the mode of action of the Ton molecular motor has remained elusive, and the precise stoichiometry of its subunits is still a matter of debate. This review summarizes the latest findings on the Ton system by presenting the recently reported structures and related reports on the stoichiometry of the fully assembled complex.


Subject(s)
Bacterial Proteins/metabolism , Gram-Negative Bacteria/metabolism , Membrane Proteins/metabolism , Proton-Motive Force , Bacterial Proteins/chemistry , Escherichia coli/chemistry , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Gram-Negative Bacteria/chemistry , Gram-Negative Bacterial Infections/microbiology , Humans , Membrane Proteins/chemistry , Models, Molecular , Protein Multimerization
4.
Front Cell Infect Microbiol ; 14: 1380976, 2024.
Article in English | MEDLINE | ID: mdl-38596648

ABSTRACT

Introduction: The hemin acquisition system is composed of an outer membrane TonB-dependent transporter that internalizes hemin into the periplasm, periplasmic hemin-binding proteins to shuttle hemin, an inner membrane transporter that transports hemin into the cytoplasm, and cytoplasmic heme oxygenase to release iron. Fur and HemP are two known regulators involved in the regulation of hemin acquisition. The hemin acquisition system of Stenotrophomonas maltophilia is poorly understood, with the exception of HemA as a TonB-dependent transporter for hemin uptake. Methods: Putative candidates responsible for hemin acquisition were selected via a homolog search and a whole-genome survey of S. maltophilia. Operon verification was performed by reverse transcription-polymerase chain reaction. The involvement of candidate genes in hemin acquisition was assessed using an in-frame deletion mutant construct and iron utilization assays. The transcript levels of candidate genes were determined using quantitative polymerase chain reaction. Results: Smlt3896-hemU-exbB2-exbD2-tonB2 and tonB1-exbB1-exbD1a-exbD1b operons were selected as candidates for hemin acquisition. Compared with the parental strain, hemU and tonB1 mutants displayed a defect in their ability to use hemin as the sole iron source for growth. However, hemin utilization by the Smlt3896 and tonB2 mutants was comparable to that of the parental strain. HemA expression was repressed by Fur in iron-replete conditions and derepressed in iron-depleted conditions. HemP negatively regulated hemA expression. Like hemA, hemU was repressed by Fur in iron-replete conditions; however, hemU was moderately derepressed in response to iron-depleted stress and fully derepressed when hemin was present. Unlike hemA and hemU, the TonB1-exbB1-exbD1a-exbD1b operon was constitutively expressed, regardless of the iron level or the presence of hemin, and Fur and HemP had no influence on its expression. Conclusion: HemA, HemU, and TonB1 contribute to hemin acquisition in S. maltophilia. Fur represses the expression of hemA and hemU in iron-replete conditions. HemA expression is regulated by low iron levels, and HemP acts as a negative regulator of this regulatory circuit. HemU expression is regulated by low iron and hemin levels in a hemP-dependent manner.


Subject(s)
Hemin , Stenotrophomonas maltophilia , Stenotrophomonas maltophilia/genetics , Stenotrophomonas maltophilia/metabolism , Bacterial Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Iron/metabolism
5.
Helicobacter ; 18(5): 363-72, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23600974

ABSTRACT

BACKGROUND: Helicobacter pylori, a neutralophile, colonizes the acidic environment of the human stomach by employing acid acclimation mechanisms that regulate periplasmic and cytoplasmic pH. The regulation of urease activity is central to acid acclimation. Inactive urease apoenzyme, UreA/B, requires nickel for activation. Accessory proteins UreE, F, G, and H are required for nickel insertion into apoenzyme. The ExbB/ExbD/TonB complex transfers energy from the inner to outer membrane, providing the driving force for nickel uptake. Therefore, the aim of this study was to determine the contribution of ExbD to pH homeostasis. MATERIALS AND METHODS: A nonpolar exbD knockout was constructed and survival, growth, urease activity, and membrane potential were determined in comparison with wildtype. RESULTS: Survival of the ΔexbD strain was significantly reduced at pH 3.0. Urease activity as a function of pH and UreI activation was similar to the wildtype strain, showing normal function of the proton-gated urea channel, UreI. The increase in total urease activity over time in acid seen in the wildtype strain was abolished in the ΔexbD strain, but recovered in the presence of supraphysiologic nickel concentrations, demonstrating that the effect of the ΔexbD mutant is due to loss of a necessary constant supply of nickel. In acid, ΔexbD also decreased its ability to maintain membrane potential and periplasmic buffering in the presence of urea. CONCLUSIONS: ExbD is essential for maintenance of periplasmic buffering and membrane potential by transferring energy required for nickel uptake, making it a potential nonantibiotic target for H. pylori eradication.


Subject(s)
Adaptation, Physiological , Bacterial Proteins/metabolism , Helicobacter pylori/physiology , Homeostasis , Bacterial Proteins/genetics , Cell Membrane/physiology , Gene Knockout Techniques , Helicobacter pylori/drug effects , Helicobacter pylori/genetics , Helicobacter pylori/growth & development , Humans , Hydrogen-Ion Concentration , Membrane Potentials , Microbial Viability/drug effects , Urease/metabolism
6.
Front Microbiol ; 13: 852955, 2022.
Article in English | MEDLINE | ID: mdl-35464957

ABSTRACT

The Ton complex is a molecular motor at the inner membrane of Gram-negative bacteria that uses a proton gradient to apply forces on outer membrane (OM) proteins to permit active transport of nutrients into the periplasmic space. Recently, the structure of the ExbB-ExbD subcomplex was determined in several bacterial species, but the complete structure and stoichiometry of TonB have yet to be determined. The C-terminal end of TonB is known to cross the periplasm and interact with TonB-dependent outer membrane transport proteins with high affinity. Yet despite having significant knowledge of these transport proteins, it is not clear how the Ton motor opens a pathway across the outer membrane for nutrient import. Additionally, the mechanism by which energy is harnessed from the inner membrane subcomplex and transduced to the outer membrane via TonB is not well understood. In this review, we will discuss the gaps in the knowledge about the complete structure of the Ton motor complex and the relationship between ion flow used to generate mechanical work at the outer membrane and the nutrient transport process.

7.
FEBS Lett ; 593(6): 581-593, 2019 03.
Article in English | MEDLINE | ID: mdl-30791094

ABSTRACT

Our study aims to investigate the physiological role of organophosphate hydrolase (OPH), hitherto known for its involvement in the degradation of organophosphate insecticides and nerve agents in Sphingobium fuliginis. We find that OPH exists as part of the TonB-dependent Transport system that is involved in nutrient transport across the bacterial outer membrane. OPH interacts physically with the Ton complex components ExbD and TonB. The surface-exposed arginine residues (R91 and R96) of OPH facilitate its interaction with ExbD. OPH is targeted to the inner membrane of Escherichia coli only when it is co-expressed with either ExbD or the ExbB/ExbD complex. In the absence of ExbD, OPH remains in the cytoplasm. Our findings suggest a role for OPH in outer membrane transport.


Subject(s)
Bacterial Proteins/chemistry , Escherichia coli Proteins/chemistry , Membrane Proteins/chemistry , Organophosphonates/chemistry , Phosphoric Monoester Hydrolases/chemistry , Sphingomonadaceae/enzymology , Arginine/chemistry , Arginine/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Cell Membrane/chemistry , Cell Membrane/enzymology , Cloning, Molecular , Cytoplasm/chemistry , Cytoplasm/enzymology , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Hydrolysis , Membrane Proteins/genetics , Membrane Proteins/metabolism , Organophosphonates/metabolism , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sphingomonadaceae/genetics , Substrate Specificity , Thermodynamics
SELECTION OF CITATIONS
SEARCH DETAIL