Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Curr Biol ; 34(7): R267-R268, 2024 04 08.
Article in English | MEDLINE | ID: mdl-38593766

ABSTRACT

In this Quick guide, Palmer and Berks introduce the twin-arginine translocation (Tat) systems. Tats are found in a variety of microbes and microbe-derived organelles, and are known to translocate folded substrate proteins across biological membranes.


Subject(s)
Escherichia coli Proteins , Twin-Arginine-Translocation System , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Escherichia coli Proteins/metabolism , Twin-Arginine-Translocation System/metabolism , Cell Membrane/metabolism , Arginine/metabolism , Protein Transport , Protein Sorting Signals , Bacterial Proteins/metabolism
2.
Microbiologyopen ; 12(2): e1350, 2023 04.
Article in English | MEDLINE | ID: mdl-37186227

ABSTRACT

High-value heterologous proteins produced in Escherichia coli that contain disulfide bonds are almost invariably targeted to the periplasm via the Sec pathway as it, among other advantages, enables disulfide bond formation and simplifies downstream processing. However, the Sec system cannot transport complex or rapidly folding proteins, as it only transports proteins in an unfolded state. The Tat system also transports proteins to the periplasm, and it has significant potential as an alternative means of recombinant protein production because it transports fully folded proteins. Most of the studies related to Tat secretion have used the well-studied TorA signal peptide that is Tat-specific, but this signal peptide also tends to induce degradation of the protein of interest, resulting in lower yields. This makes it difficult to use Tat in the industry. In this study, we show that a model disulfide bond-containing protein, YebF, can be exported to the periplasm and media at a very high level by the Tat pathway in a manner almost completely dependent on cytoplasmic disulfide formation, by other two putative Tat SPs: those of MdoD and AmiC. In contrast, the TorA SP exports YebF at a low level.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Periplasm , Recombinant Proteins , Twin-Arginine-Translocation System , Protein Transport , Periplasm/metabolism , Disulfides/chemistry , Escherichia coli/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Protein Folding , Oxidoreductases, N-Demethylating/genetics , Oxidoreductases, N-Demethylating/metabolism , Protein Sorting Signals , Twin-Arginine-Translocation System/metabolism , Culture Media , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism
3.
Microbiol Spectr ; 10(5): e0162122, 2022 10 26.
Article in English | MEDLINE | ID: mdl-36036643

ABSTRACT

The twin arginine translocation system (Tat) is a protein export system that is conserved in bacteria, archaea, and plants. In Gram-negative bacteria, it is required for the export of folded proteins from the cytoplasm to the periplasm. In Salmonella, there are 30 proteins that are predicted substrates of Tat, and among these are enzymes required for anaerobic respiration and peptidoglycan remodeling. We have demonstrated that some conditions that induce bacterial envelope stress activate expression of a ΔtatABC-lacZ fusion in Salmonella enterica serovar Typhimurium. Particularly, the addition of bile salts to the growth medium causes a 3-fold induction of a ΔtatABC-lacZ reporter fusion. Our data demonstrate that this induction is mediated via the phage shock protein (Psp) stress response system protein PspA. Further, we show that deletion of tatABC increases the induction of tatABC expression in bile salts. Indeed, the data suggest significant interaction between PspA and the Tat system in the regulatory response to bile salts. Although we have not identified the precise mechanism of Psp regulation of tatABC, our work shows that PspA is involved in the activation of tatABC expression by bile salts and adds another layer of complexity to the Salmonella response to envelope stress. IMPORTANCE Salmonella species cause an array of diseases in a variety of hosts. This research is significant in showing induction of the Tat system as a defense against periplasmic stress. Understanding the underlying mechanism of this regulation broadens our understanding of the Salmonella stress response, which is critical to the ability of the organism to cause infection.


Subject(s)
Escherichia coli Proteins , Twin-Arginine-Translocation System , Twin-Arginine-Translocation System/genetics , Twin-Arginine-Translocation System/metabolism , Peptidoglycan/metabolism , Salmonella typhimurium/metabolism , Heat-Shock Proteins/metabolism , Arginine/metabolism , Bile Acids and Salts/metabolism , Bacterial Proteins/metabolism , Escherichia coli Proteins/metabolism
4.
J Biol Chem ; 298(9): 102236, 2022 09.
Article in English | MEDLINE | ID: mdl-35809643

ABSTRACT

The twin-arginine translocation (Tat) system serves to translocate folded proteins across energy-transducing membranes in bacteria, archaea, plastids, and some mitochondria. In Escherichia coli, TatA, TatB, and TatC constitute functional translocons. TatA and TatB both possess an N-terminal transmembrane helix (TMH) followed by an amphipathic helix. The TMHs of TatA and TatB generate a hydrophobic mismatch with the membrane, as the helices comprise only 12 consecutive hydrophobic residues; however, the purpose of this mismatch is unclear. Here, we shortened or extended this stretch of hydrophobic residues in either TatA, TatB, or both and analyzed effects on translocon function and assembly. We found the WT length helices functioned best, but some variation was clearly tolerated. Defects in function were exacerbated by simultaneous mutations in TatA and TatB, indicating partial compensation of mutations in each by the other. Furthermore, length variation in TatB destabilized TatBC-containing complexes, revealing that the 12-residue-length is important but not essential for this interaction and translocon assembly. To also address potential effects of helix length on TatA interactions, we characterized these interactions by molecular dynamics simulations, after having characterized the TatA assemblies by metal-tagging transmission electron microscopy. In these simulations, we found that interacting short TMHs of larger TatA assemblies were thinning the membrane and-together with laterally-aligned tilted amphipathic helices-generated a deep V-shaped membrane groove. We propose the 12 consecutive hydrophobic residues may thus serve to destabilize the membrane during Tat transport, and their conservation could represent a delicate compromise between functionality and minimization of proton leakage.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Membrane Transport Proteins , Twin-Arginine-Translocation System , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Hydrophobic and Hydrophilic Interactions , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Protein Conformation, alpha-Helical , Protons , Twin-Arginine-Translocation System/metabolism
5.
J Biol Chem ; 298(7): 102107, 2022 07.
Article in English | MEDLINE | ID: mdl-35671825

ABSTRACT

An ever-increasing number of proteins have been shown to translocate across various membranes of bacterial as well as eukaryotic cells in their folded states as a part of physiological and/or pathophysiological processes. Herein, we provide an overview of the systems/processes that are established or likely to involve the membrane translocation of folded proteins, such as protein export by the twin-arginine translocation system in bacteria and chloroplasts, unconventional protein secretion and protein import into the peroxisome in eukaryotes, and the cytosolic entry of proteins (e.g., bacterial toxins) and viruses into eukaryotes. We also discuss the various mechanistic models that have previously been proposed for the membrane translocation of folded proteins including pore/channel formation, local membrane disruption, membrane thinning, and transport by membrane vesicles. Finally, we introduce a newly discovered vesicular transport mechanism, vesicle budding and collapse, and present evidence that vesicle budding and collapse may represent a unifying mechanism that drives some (and potentially all) of folded protein translocation processes.


Subject(s)
Protein Folding , Protein Transport , Bacteria/metabolism , Bacterial Proteins/metabolism , Eukaryota/metabolism , Membrane Transport Proteins/metabolism , Peroxisomes/metabolism , Protein Sorting Signals , Twin-Arginine-Translocation System/metabolism
6.
J Bacteriol ; 204(4): e0061221, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35311558

ABSTRACT

Flagellum-mediated bacterial motility is important for bacteria to take up nutrients, adapt to environmental changes, and establish infection. The twin-arginine translocation system (Tat) is an important protein export system, playing a critical role in bacterial physiology and pathogenesis. It has been observed for a long time that the Tat system is critical for bacterial motility. However, the underlying mechanism remains unrevealed. In this study, a comparative transcriptomics analysis was performed with extraintestinal pathogenic Escherichia coli (ExPEC), which identified a considerable number of genes differentially expressed when the Tat system was disrupted. Among them, a large proportion of flagellar biosynthesis genes showed downregulation, indicating that transcription regulation plays an important role in mediating the motility defects. We further identified three Tat substrate proteins, MdoD, AmiA, and AmiC, that were responsible for the nonmotile phenotype. The Rcs system was deleted in the Δtat, the ΔmdoD, and the ΔamiAΔamiC strains, which restored the motility of ΔmdoD and partially restored the motility of Δtat and ΔamiAΔamiC. The flagella were also observed in all of the ΔtatΔrcsDB, ΔmdoDΔrcsDB, and ΔamiAΔamiCΔrcsDB strains, but not in the Δtat, ΔmdoD, and ΔamiAΔamiC strains, by using transmission electron microscopy. Quantitative reverse transcription-PCR data revealed that the regulons of the Rcs system displayed differential expression in the tat mutant, indicating that the Rcs signaling was activated. Our results suggest that the Rcs system plays an important role in mediating the motility defects of the tat mutant of ExPEC. IMPORTANCE The Tat system is an important protein export system critical for bacterial physiology and pathogenesis. It has been observed for a long time that the Tat system is critical for bacterial motility. However, the underlying mechanism remains unrevealed. In this study, we combine transcriptomics analysis and bacterial genetics, which reveal that transcription regulation plays an important role in mediating the motility defects of the tat mutant of extraintestinal pathogenic Escherichia coli. The Tat substrate proteins responsible for the motility defects are identified. We further show that the Rcs system contributes to the motility suppression. We for the first time reveal the link between the Tat system and bacterial motility, which is important for understanding the physiological functions of the Tat system.


Subject(s)
Escherichia coli Proteins , Extraintestinal Pathogenic Escherichia coli , Twin-Arginine-Translocation System , Arginine/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Extraintestinal Pathogenic Escherichia coli/genetics , Extraintestinal Pathogenic Escherichia coli/metabolism , Flagella/metabolism , Protein Transport , Twin-Arginine-Translocation System/genetics , Twin-Arginine-Translocation System/metabolism
7.
PLoS Pathog ; 17(8): e1009425, 2021 08.
Article in English | MEDLINE | ID: mdl-34460871

ABSTRACT

Extracellular DNA (eDNA) is a major constituent of the extracellular matrix of Pseudomonas aeruginosa biofilms and its release is regulated via pseudomonas quinolone signal (PQS) dependent quorum sensing (QS). By screening a P. aeruginosa transposon library to identify factors required for DNA release, mutants with insertions in the twin-arginine translocation (Tat) pathway were identified as exhibiting reduced eDNA release, and defective biofilm architecture with enhanced susceptibility to tobramycin. P. aeruginosa tat mutants showed substantial reductions in pyocyanin, rhamnolipid and membrane vesicle (MV) production consistent with perturbation of PQS-dependent QS as demonstrated by changes in pqsA expression and 2-alkyl-4-quinolone (AQ) production. Provision of exogenous PQS to the tat mutants did not return pqsA, rhlA or phzA1 expression or pyocyanin production to wild type levels. However, transformation of the tat mutants with the AQ-independent pqs effector pqsE restored phzA1 expression and pyocyanin production. Since mutation or inhibition of Tat prevented PQS-driven auto-induction, we sought to identify the Tat substrate(s) responsible. A pqsA::lux fusion was introduced into each of 34 validated P. aeruginosa Tat substrate deletion mutants. Analysis of each mutant for reduced bioluminescence revealed that the primary signalling defect was associated with the Rieske iron-sulfur subunit of the cytochrome bc1 complex. In common with the parent strain, a Rieske mutant exhibited defective PQS signalling, AQ production, rhlA expression and eDNA release that could be restored by genetic complementation. This defect was also phenocopied by deletion of cytB or cytC1. Thus, either lack of the Rieske sub-unit or mutation of cytochrome bc1 genes results in the perturbation of PQS-dependent autoinduction resulting in eDNA deficient biofilms, reduced antibiotic tolerance and compromised virulence factor production.


Subject(s)
Biofilms/growth & development , Electron Transport Complex III/metabolism , Extracellular Vesicles/genetics , Pseudomonas aeruginosa/growth & development , Quinolones/metabolism , Quorum Sensing , Twin-Arginine-Translocation System/metabolism , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biofilms/drug effects , DNA, Bacterial/genetics , Electron Transport Complex III/genetics , Gene Expression Regulation, Bacterial , Glycolipids/metabolism , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/metabolism , Pyocyanine/metabolism , Twin-Arginine-Translocation System/genetics , Virulence Factors/genetics , Virulence Factors/metabolism
8.
Biochim Biophys Acta Mol Cell Res ; 1868(2): 118914, 2021 02.
Article in English | MEDLINE | ID: mdl-33245978

ABSTRACT

The widely conserved twin-arginine translocases (Tat) allow the transport of fully folded cofactor-containing proteins across biological membranes. In doing so, these translocases serve different biological functions ranging from energy conversion to cell division. In the Gram-positive soil bacterium Bacillus subtilis, the Tat machinery is essential for effective growth in media lacking iron or NaCl. It was previously shown that this phenomenon relates to the Tat-dependent export of the heme-containing peroxidase EfeB, which converts Fe2+ to Fe3+ at the expense of hydrogen peroxide. However, the reasons why the majority of tat mutant bacteria perish upon dilution in NaCl-deprived medium and how, after several hours, a sub-population adapts to this condition was unknown. Here we show that, upon growth in the absence of NaCl, the bacteria face two major problems, namely severe oxidative stress at the membrane and starvation leading to death. The tat mutant cells can overcome these challenges if they are fed with arginine, which implies that severe arginine depletion is a major cause of death and resumed arginine synthesis permits their survival. Altogether, our findings show that the Tat system of B. subtilis is needed to preclude severe oxidative stress and starvation upon sudden drops in the environmental Na+ concentration as caused by flooding or rain.


Subject(s)
Bacillus subtilis/metabolism , Bacterial Proteins/metabolism , Membrane Transport Proteins/metabolism , Oxidative Stress/genetics , Sodium Chloride/metabolism , Twin-Arginine-Translocation System/metabolism , Arginine/metabolism , Bacillus subtilis/genetics , Bacterial Proteins/genetics , Cell Membrane/metabolism , Membrane Transport Proteins/genetics , Microbial Viability/genetics , Mutant Proteins/metabolism , Organisms, Genetically Modified , Protein Transport/genetics , Twin-Arginine-Translocation System/genetics
9.
Infect Immun ; 88(11)2020 10 19.
Article in English | MEDLINE | ID: mdl-32778612

ABSTRACT

Brucella, the causative agent of brucellosis, is a stealthy intracellular pathogen that is highly pathogenic to a range of mammals, including humans. The twin-arginine translocation (Tat) pathway transports folded proteins across the cytoplasmic membrane and has been implicated in virulence in many bacterial pathogens. However, the roles of the Tat system and related substrates in Brucella remain unclear. We report here that disruption of Tat increases the sensitivity of Brucella melitensis M28 to the membrane stressor sodium dodecyl sulfate (SDS), indicating cell envelope defects, as well as to EDTA. In addition, mutating Tat renders M28 bacteria more sensitive to oxidative stress caused by H2O2 Further, loss of Tat significantly attenuates B. melitensis infection in murine macrophages ex vivo Using a mouse model for persistent infection, we demonstrate that Tat is required for full virulence of B. melitensis M28. Genome-wide in silico prediction combined with an in vivo amidase reporter assay indicates that at least 23 proteins are authentic Tat substrates, and they are functionally categorized into solute-binding proteins, oxidoreductases, cell envelope biosynthesis enzymes, and others. A comprehensive deletion study revealed that 6 substrates contribute significantly to Brucella virulence, including an l,d-transpeptidase, an ABC transporter solute-binding protein, and a methionine sulfoxide reductase. Collectively, our work establishes that the Tat pathway plays a critical role in Brucella virulence.


Subject(s)
Bacterial Proteins/metabolism , Brucella melitensis/pathogenicity , Brucellosis/metabolism , Twin-Arginine-Translocation System/metabolism , Virulence/physiology , Animals , Mice , Stress, Physiological/physiology
10.
Cell ; 180(6): 1144-1159.e20, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32169217

ABSTRACT

In eukaryotic cells, organelle biogenesis is pivotal for cellular function and cell survival. Chloroplasts are unique organelles with a complex internal membrane network. The mechanisms of the migration of imported nuclear-encoded chloroplast proteins across the crowded stroma to thylakoid membranes are less understood. Here, we identified two Arabidopsis ankyrin-repeat proteins, STT1 and STT2, that specifically mediate sorting of chloroplast twin arginine translocation (cpTat) pathway proteins to thylakoid membranes. STT1 and STT2 form a unique hetero-dimer through interaction of their C-terminal ankyrin domains. Binding of cpTat substrate by N-terminal intrinsically disordered regions of STT complex induces liquid-liquid phase separation. The multivalent nature of STT oligomer is critical for phase separation. STT-Hcf106 interactions reverse phase separation and facilitate cargo targeting and translocation across thylakoid membranes. Thus, the formation of phase-separated droplets emerges as a novel mechanism of intra-chloroplast cargo sorting. Our findings highlight a conserved mechanism of phase separation in regulating organelle biogenesis.


Subject(s)
Arabidopsis/metabolism , Protein Transport/physiology , Twin-Arginine-Translocation System/metabolism , Chloroplast Proteins/metabolism , Chloroplasts/metabolism , Intracellular Membranes/metabolism , Membrane Proteins/metabolism , Organelle Biogenesis , Organelles/metabolism , Phase Transition , Plant Proteins/metabolism , Thylakoids/metabolism , Twin-Arginine-Translocation System/physiology
11.
Biotechnol Lett ; 42(1): 45-55, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31679097

ABSTRACT

OBJECTIVES: The twin-arginine translocation (Tat) pathway is one of the bacterial secretory strategies which exports folded proteins across the cytoplasmic membrane. RESULTS: In the present study, we designed a novel Tat-signal peptide for secretion of human activin A used as a recombinant protein model here. In doing so, Haloferax volcanii, Halobacterium salinarum, and Escherichia coli Tat specific signal peptides were aligned by ClustalW program to determine conserved and more frequently used residues. After making the initial signal peptide sequence and doing some mutations, efficiency of this designed signal peptide was evaluated using a set of well-known software programs such as TatP, PRED-TAT, and Phobius. Then the best complex between TatC as an initiator protein in Tat secretory machine and the new designed signal peptide connected to activin A with the lowest binding energy was constructed by HADDOCK server, and ΔΔG value of - 5.5 kcal/mol was calculated by FoldX module. After that, efficiency of this novel signal peptide for secretion of human activin A to the periplasmic space of E. coli Rosetta-gami (DE3) strain was experimentally evaluated; to scrutinize the activity of the novel signal peptide, Iranian Bacillus Licheniformis α-Amylase enzyme signal peptide as a Sec pathway signal peptide was used as a positive control. The quantitative analysis of western blotting bands by ImageJ software confirmed the high secretion ability of the new designed signal peptide; translocation of 69% of the produced recombinant activin A to the periplasmic space of E. coli. Circular Dichroism (CD) spectroscopy technique also approved the proper secondary structure of activin A secreted to the periplasmic space. The biological activity of activin A was also confirmed by differentiation of K562 erythroleukemia cells to the red blood cell by measuring the amount of hemoglobin or Fe2+ ion using ICP method. CONCLUSIONS: In conclusion, this novel designed signal peptide can be used to secrete any other recombinant proteins to the periplasmic space of E. coli efficiently.


Subject(s)
Activins/metabolism , Cell Membrane/metabolism , Escherichia coli/metabolism , Periplasm/metabolism , Protein Sorting Signals/genetics , Recombinant Proteins/metabolism , Twin-Arginine-Translocation System/metabolism , Activins/chemistry , Activins/genetics , Cell Membrane/enzymology , Circular Dichroism , Escherichia coli/genetics , Halobacterium salinarum/genetics , Haloferax volcanii/genetics , Humans , Protein Folding , Protein Transport , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Sequence Alignment
12.
J Biol Chem ; 294(38): 13902-13914, 2019 09 20.
Article in English | MEDLINE | ID: mdl-31341014

ABSTRACT

Twin-arginine-dependent translocases transport folded proteins across bacterial, archaeal, and chloroplast membranes. Upon substrate binding, they assemble from hexahelical TatC and single-spanning TatA and TatB membrane proteins. Although structural and functional details of individual Tat subunits have been reported previously, the sequence and dynamics of Tat translocase assembly remain to be determined. Employing the zero-space cross-linker N,N'-dicyclohexylcarbodiimide (DCCD) in combination with LC-MS/MS, we identified as yet unknown intra- and intermolecular contact sites of TatB and TatC. In addition to their established intramembrane binding sites, both proteins were thus found to contact each other through the soluble N terminus of TatC and the interhelical linker region around the conserved glutamyl residue Glu49 of TatB from Escherichia coli Functional analyses suggested that by interacting with the TatC N terminus, TatB improves the formation of a proficient substrate recognition site of TatC. The Glu49 region of TatB was found also to contact distinct downstream sites of a neighboring TatB molecule and to thereby mediate oligomerization of TatB within the TatBC receptor complex. Finally, we show that global DCCD-mediated cross-linking of TatB and TatC in membrane vesicles or, alternatively, creating covalently linked TatC oligomers prevents TatA from occupying a position close to the TatBC-bound substrate. Collectively, our results are consistent with a circular arrangement of the TatB and TatC units within the TatBC receptor complex and with TatA entering the interior TatBC-binding cavity through lateral gates between TatBC protomers.


Subject(s)
Escherichia coli Proteins/metabolism , Membrane Transport Proteins/metabolism , Twin-Arginine-Translocation System/metabolism , Amino Acid Sequence/genetics , Binding Sites/genetics , Carrier Proteins/metabolism , Cell Membrane/metabolism , Chromatography, Liquid/methods , Escherichia coli/metabolism , Escherichia coli Proteins/physiology , Membrane Proteins/metabolism , Membrane Transport Proteins/physiology , Models, Molecular , Protein Binding/physiology , Protein Folding , Protein Sorting Signals/genetics , Protein Transport/physiology , Structure-Activity Relationship , Tandem Mass Spectrometry/methods , Twin-Arginine-Translocation System/physiology
13.
Article in English | MEDLINE | ID: mdl-31355152

ABSTRACT

Antibiotics are becoming less effective in treatment of infections caused by multidrug-resistant Pseudomonas aeruginosa. Antimicrobial therapies based on the inhibition of specific virulence-related traits, as opposed to growth inhibitors, constitute an innovative and appealing approach to tackle the threat of P. aeruginosa infections. The twin-arginine translocation (Tat) pathway plays an important role in the pathogenesis of P. aeruginosa, and constitutes a promising target for the development of anti-pseudomonal drugs. In this study we developed and optimized a whole-cell, one-well assay, based on native phospholipase C activity, to identify compounds active against the Tat system. Statistical robustness, sensitivity and consequently suitability for high-throughput screening (HTS) were confirmed by a dry run/pre-screening test scoring a Z' of 0.82 and a signal-to-noise ratio of 49. Using this assay, we evaluated ca. 40,000 molecules and identified 59 initial hits as possible Tat inhibitors. Since phospholipase C is exported into the periplasm by Tat, and subsequently translocated across the outer membrane by the type II secretion system (T2SS), our assay could also identify T2SS inhibitors. To validate our hits and discriminate between compounds that inhibited either Tat or T2SS, two separate counter assays were developed and optimized. Finally, three Tat inhibitors and one T2SS inhibitor were confirmed by means of dose-response analysis and additional counter and confirming assays. Although none of the identified inhibitors was suitable as a lead compound for drug development, this study validates our assay as a simple, efficient, and HTS compatible method for the identification of Tat and T2SS inhibitors.


Subject(s)
Anti-Bacterial Agents/pharmacology , High-Throughput Screening Assays , Pseudomonas aeruginosa/drug effects , Small Molecule Libraries/pharmacology , Twin-Arginine-Translocation System/drug effects , Type II Secretion Systems/drug effects , Anti-Bacterial Agents/chemistry , Dose-Response Relationship, Drug , Drug Discovery , Drug Resistance, Multiple, Bacterial/drug effects , Protein Transport/drug effects , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/growth & development , Pseudomonas aeruginosa/metabolism , Small Molecule Libraries/chemistry , Twin-Arginine-Translocation System/genetics , Twin-Arginine-Translocation System/metabolism , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/genetics , Type C Phospholipases/metabolism , Type II Secretion Systems/genetics , Type II Secretion Systems/metabolism
14.
Subcell Biochem ; 92: 337-366, 2019.
Article in English | MEDLINE | ID: mdl-31214992

ABSTRACT

The inner membrane of Gram-negative bacteria is a ~6 nm thick phospholipid bilayer. It forms a semi-permeable barrier between the cytoplasm and periplasm allowing only regulated export and import of ions, sugar polymers, DNA and proteins. Inner membrane proteins, embedded via hydrophobic transmembrane α-helices, play an essential role in this regulated trafficking: they mediate insertion into the membrane (insertases) or complete crossing of the membrane (translocases) or both. The Gram-negative inner membrane is equipped with a variety of different insertases and translocases. Many of them are specialized, taking care of the export of only a few protein substrates, while others have more general roles. Here, we focus on the three general export/insertion pathways, the secretory (Sec) pathway, YidC and the twin-arginine translocation (TAT) pathway, focusing closely on the Escherichia coli (E. coli) paradigm. We only briefly mention dedicated export pathways found in different Gram-negative bacteria. The Sec system deals with the majority of exported proteins and functions both as a translocase for secretory proteins and an insertase for membrane proteins. The insertase YidC assists the Sec system or operates independently on membrane protein clients. Sec and YidC, in common with most export pathways, require their protein clients to be in soluble non-folded states to fit through the translocation channels and grooves. The TAT pathway is an exception, as it translocates folded proteins, some loaded with prosthetic groups.


Subject(s)
Cell Membrane/enzymology , Cell Membrane/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Membrane Transport Proteins/metabolism , SEC Translocation Channels/metabolism , Twin-Arginine-Translocation System/metabolism , Escherichia coli/cytology , Escherichia coli/metabolism , Protein Transport
15.
PLoS Negl Trop Dis ; 13(6): e0007449, 2019 06.
Article in English | MEDLINE | ID: mdl-31170147

ABSTRACT

We have previously shown that the cell morphogenesis NlpD lipoprotein is essential for virulence of the plague bacteria, Yersinia pestis. To elucidate the role of NlpD in Y. pestis pathogenicity, we conducted a whole-genome comparative transcriptome analysis of the wild-type Y. pestis strain and an nlpD mutant under conditions mimicking early stages of infection. The analysis suggested that NlpD is involved in three phenomena: (i) Envelope stability/integrity evidenced by compensatory up-regulation of the Cpx and Psp membrane stress-response systems in the mutant; (ii) iron acquisition, supported by modulation of iron metabolism genes and by limited growth in iron-deprived medium; (iii) activity of the twin-arginine (Tat) system, which translocates folded proteins across the cytoplasmic membrane. Virulence studies of Y. pestis strains mutated in individual Tat components clearly indicated that the Tat system is central in Y. pestis pathogenicity and substantiated the assumption that NlpD essentiality in iron utilization involves the activity of the Tat system. This study reveals a new role for NlpD in Tat system activity and iron assimilation suggesting a modality by which this lipoprotein is involved in Y. pestis pathogenesis.


Subject(s)
Bacterial Proteins/metabolism , Gene Knockout Techniques , Iron/metabolism , Lipoproteins/metabolism , Twin-Arginine-Translocation System/metabolism , Virulence Factors/metabolism , Yersinia pestis/enzymology , Yersinia pestis/metabolism , Animals , Bacterial Proteins/genetics , Biological Transport , Female , Gene Expression Profiling , Gene Expression Regulation, Bacterial , Lipoproteins/genetics , Mice , Virulence Factors/genetics , Yersinia pestis/genetics
16.
Biotechnol Bioeng ; 116(4): 722-733, 2019 04.
Article in English | MEDLINE | ID: mdl-30536699

ABSTRACT

The large-scale production and isolation of recombinant protein is a central element of the biotechnology industry and many of the products have proved extremely beneficial for therapeutic medicine. Escherichia coli is the microorganism of choice for the expression of heterologous proteins for therapeutic application, and a range of high-value proteins have been targeted to the periplasm using the well characterized Sec protein export pathway. More recently, the ability of the second mainstream protein export system, the twin-arginine translocase, to transport fully-folded proteins into the periplasm of not only E. coli, but also other Gram-negative bacteria, has captured the interest of the biotechnology industry. In this study, we have used a novel approach to block the export of a heterologous Tat substrate in the later stages of the export process, and thereby generate a single-span membrane protein with the soluble domain positioned on the periplasmic side of the inner membrane. Biochemical and immuno-electron microscopy approaches were used to investigate the export of human growth hormone by the twin-arginine translocase, and the generation of a single-span membrane-embedded variant. This is the first time that a bonafide biotechnologically relevant protein has been exported by this machinery and visualized directly in this manner. The data presented here demonstrate a novel method for the production of single-span membrane proteins in E. coli.


Subject(s)
Escherichia coli/metabolism , Human Growth Hormone/metabolism , Industrial Microbiology , Escherichia coli/cytology , Human Growth Hormone/analysis , Humans , Industrial Microbiology/methods , Protein Sorting Signals , Protein Transport , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/metabolism , Solubility , Twin-Arginine-Translocation System/analysis , Twin-Arginine-Translocation System/metabolism
17.
J Agric Food Chem ; 66(50): 13217-13227, 2018 Dec 19.
Article in English | MEDLINE | ID: mdl-30465427

ABSTRACT

Widespread utilization of polyethylene terephthalate (PET) has caused critical environmental pollution. The enzymatic degradation of PET is a promising solution to this problem. In this study, PETase, which exhibits much higher PET-hydrolytic activity than other enzymes, was successfully secreted into extracellular milieu from Bacillus subtilis 168 under the direction of its native signal peptide (named SPPETase). SPPETase is predicted to be a twin-arginine signal peptide. Intriguingly, inactivation of twin-arginine translocation (Tat) complexes improved the secretion amount by 3.8-fold, indicating that PETase was exported via Tat-independent pathway. To the best of our knowledge, this is the first report on the improvement of Tat-independent secretion by inactivating Tat components of B. subtilis 168 in LB medium. Furthermore, PET film degradation assay showed that the secreted PETase was fully active. This study paves the first step to construct an efficient engineered strain for PET degradation.


Subject(s)
Bacillus subtilis/metabolism , Bacterial Proteins/metabolism , Hydrolases/metabolism , Polyethylene Terephthalates/metabolism , Twin-Arginine-Translocation System/metabolism , Bacillus subtilis/enzymology , Bacillus subtilis/genetics , Bacterial Proteins/genetics , Biodegradation, Environmental , Hydrolases/genetics , Metabolic Engineering , Protein Sorting Signals , Protein Transport , Twin-Arginine-Translocation System/genetics
18.
PLoS Pathog ; 14(10): e1007391, 2018 10.
Article in English | MEDLINE | ID: mdl-30379938

ABSTRACT

Salmonella enterica serovar Typhimurium (S. Tm) is a cause of food poisoning accompanied with gut inflammation. Although mucosal inflammation is generally thought to be protective against bacterial infection, S. Tm exploits the inflammation to compete with commensal microbiota, thereby growing up to high densities in the gut lumen and colonizing the gut continuously at high levels. However, the molecular mechanisms underlying the beneficial effect of gut inflammation on S. Tm competitive growth are poorly understood. Notably, the twin-arginine translocation (Tat) system, which enables the transport of folded proteins outside bacterial cytoplasm, is well conserved among many bacterial pathogens, with Tat substrates including virulence factors and virulence-associated proteins. Here, we show that Tat and Tat-exported peptidoglycan amidase, AmiA- and AmiC-dependent cell division contributes to S. Tm competitive fitness advantage in the inflamed gut. S. Tm tatC or amiA amiC mutants feature a gut colonization defect, wherein they display a chain form of cells. The chains are attributable to a cell division defect of these mutants and occur in inflamed but not in normal gut. We demonstrate that attenuated resistance to bile acids confers the colonization defect on the S. Tm amiA amiC mutant. In particular, S. Tm cell chains are highly sensitive to bile acids as compared to single or paired cells. Furthermore, we show that growth media containing high concentrations of NaCl and sublethal concentrations of antimicrobial peptides induce the S. Tm amiA amiC mutant chain form, suggesting that gut luminal conditions such as high osmolarity and the presence of antimicrobial peptides impose AmiA- and AmiC-dependent cell division on S. Tm. Together, our data indicate that Tat and the Tat-exported amidases, AmiA and AmiC, are required for S. Tm luminal fitness in the inflamed gut, suggesting that these proteins might comprise effective targets for novel antibacterial agents against infectious diarrhea.


Subject(s)
Amidohydrolases/metabolism , Gastrointestinal Tract/microbiology , Inflammation/microbiology , Peptidoglycan/metabolism , Salmonella Infections, Animal/microbiology , Salmonella typhimurium/physiology , Twin-Arginine-Translocation System/metabolism , Animals , Cell Division , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/pathology , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Inbred C57BL , Salmonella Infections, Animal/metabolism , Salmonella Infections, Animal/pathology
19.
Photosynth Res ; 138(3): 289-301, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30101370

ABSTRACT

Thylakoids are complex sub-organellar membrane systems whose role in photosynthesis makes them critical to life. Thylakoids require the coordinated expression of both nuclear- and plastid-encoded proteins to allow rapid response to changing environmental conditions. Transport of cytoplasmically synthesized proteins to thylakoids or the thylakoid lumen is complex; the process involves transport across up to three membrane systems with routing through three aqueous compartments. Protein transport in thylakoids is accomplished by conserved ancestral prokaryotic plasma membrane translocases containing novel adaptations for the sub-organellar location. This review focuses on the evolutionarily conserved chloroplast twin arginine transport (cpTat) pathway. An overview is provided of known aspects of the cpTat components, energy requirements, and mechanisms with a focus on recent discoveries. Some of the most exciting new studies have been in determining the structural architecture of the membrane complex involved in forming the point of passage for the precursor and binding features of the translocase components. The cpTat system is of particular interest because it transports folded protein domains using only the proton motive force for energy. The implications for mechanism of translocation by recent studies focusing on interactions between membrane Tat components and with the translocating precursor will be discussed.


Subject(s)
Chloroplast Proteins/metabolism , Thylakoids/metabolism , Twin-Arginine-Translocation System/metabolism , Amino Acid Sequence , Chloroplast Proteins/chemistry , Models, Molecular , Protein Transport , Twin-Arginine-Translocation System/chemistry
20.
Sci Rep ; 8(1): 11950, 2018 08 09.
Article in English | MEDLINE | ID: mdl-30093651

ABSTRACT

In bacteria, the twin-arginine translocation (Tat) pathway allows the export of folded proteins through the inner membrane. Proteins targeted to this system are synthesized with N-terminal signal peptides bearing a conserved twin-arginine motif. The Tat pathway is critical for many bacterial processes including pathogenesis and virulence. However, the full set of Tat substrates is unknown in many bacteria, and the reliability of in silico prediction methods largely uncertain. In this work, we performed a combination of in silico analysis and experimental validation to identify a core set of Tat substrates in the opportunistic pathogen Pseudomonas aeruginosa. In silico analysis predicted 44 putative Tat signal peptides in the P. aeruginosa PA14 proteome. We developed an improved amidase-based Tat reporter assay to show that 33 of these are real Tat signal peptides. In addition, in silico analysis of the full translated genome revealed a Tat candidate with a missassigned start codon. We showed that it is a new periplasmic protein in P. aeruginosa. Altogether we discovered and validated 34 Tat substrates. These show little overlap with Escherichia coli Tat substrates, and functional analysis points to a general role for the P. aeruginosa Tat system in the colonization of environmental niches and pathogenicity.


Subject(s)
Bacterial Proteins/genetics , Genome, Bacterial , Genome-Wide Association Study/methods , Pseudomonas aeruginosa/genetics , Twin-Arginine-Translocation System/genetics , Amidohydrolases/genetics , Amidohydrolases/metabolism , Amino Acid Sequence , Bacterial Proteins/metabolism , Base Sequence , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Protein Sorting Signals/genetics , Protein Transport , Proteome/genetics , Proteome/metabolism , Proteomics/methods , Pseudomonas aeruginosa/metabolism , Substrate Specificity , Twin-Arginine-Translocation System/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...