Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
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
2.
Annu Rev Biochem ; 84: 843-64, 2015.
Article in English | MEDLINE | ID: mdl-25494301

ABSTRACT

The twin-arginine translocation (Tat) system, found in prokaryotes, chloroplasts, and some mitochondria, allows folded proteins to be moved across membranes. How this transport is achieved without significant ion leakage is an intriguing mechanistic question. Tat transport is mediated by complexes formed from small integral membrane proteins from just two protein families. Atomic-resolution structures have recently been determined for representatives of both these protein families, providing the first molecular-level glimpse of the Tat machinery. I review our current understanding of the mechanism of Tat transport in light of these new structural data.


Subject(s)
Protein Transport , Twin-Arginine-Translocation System/metabolism , Archaea/classification , Archaea/metabolism , Bacteria/classification , Bacteria/metabolism , Chloroplasts/metabolism , Mitochondria/metabolism , Prokaryotic Cells/metabolism , Proton-Motive Force , Twin-Arginine-Translocation System/chemistry
3.
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
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.
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
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.
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
8.
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
9.
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
10.
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
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 ; 293(20): 7592-7605, 2018 05 18.
Article in English | MEDLINE | ID: mdl-29535185

ABSTRACT

The twin-arginine translocation (Tat) system that comprises the TatA, TatB, and TatC components transports folded proteins across energized membranes of prokaryotes and plant plastids. It is not known, however, how the transport of this protein cargo is achieved. Favored models suggest that the TatA component supports transport by weakening the membrane upon full translocon assembly. Using Escherichia coli as a model organism, we now demonstrate in vivo that the N terminus of TatA can indeed destabilize the membrane, resulting in a lowered membrane energization in growing cells. We found that in full-length TatA, this effect is counterbalanced by its amphipathic helix. Consistent with these observations, the TatA N terminus induced proton leakage in vitro, indicating membrane destabilization. Fluorescence quenching data revealed that substrate binding causes the TatA hinge region and the N-terminal part of the TatA amphipathic helix to move toward the membrane surface. In the presence of TatBC, substrate binding also reduced the exposure of a specific region in the amphipathic helix, indicating a participation of TatBC. Of note, the substrate-induced reorientation of the TatA amphipathic helix correlated with detectable membrane weakening. We therefore propose a two-state model in which membrane-destabilizing effects of the short TatA membrane anchor are compensated by the membrane-immersed N-terminal part of the amphipathic helix in a resting state. We conclude that substrate binding to TatABC complexes switches the position of the amphipathic helix, which locally weakens the membrane on demand to allow substrate translocation across the membrane.


Subject(s)
Arginine/metabolism , Cell Membrane/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Membrane Transport Proteins/metabolism , Twin-Arginine-Translocation System/metabolism , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli Proteins/genetics , Membrane Transport Proteins/genetics , Protein Binding , Twin-Arginine-Translocation System/genetics
13.
J Biol Chem ; 293(19): 7281-7299, 2018 05 11.
Article in English | MEDLINE | ID: mdl-29593092

ABSTRACT

The twin-arginine translocation (Tat) pathway transports folded proteins across bacterial membranes. Tat precursor proteins possess a conserved twin-arginine (RR) motif in their signal peptides that is involved in the binding of the proteins to the membrane-associated TatBC receptor complex. In addition, the hydrophobic region in the Tat signal peptides also contributes to TatBC binding, but whether regions beyond the signal-peptide cleavage site are involved in this process is unknown. Here, we analyzed the contribution of the early mature protein part of the Escherichia coli trimethylamine N-oxide reductase (TorA) to productive TatBC receptor binding. We identified substitutions in the 30 amino acids immediately following the TorA signal peptide (30aa-region) that restored export of a transport-defective TorA[KQ]-30aa-MalE precursor, in which the RR residues had been replaced by a lysine-glutamine pair. Some of these substitutions increased the hydrophobicity of the N-terminal part of the 30aa-region and thereby likely enhanced hydrophobic substrate-receptor interactions within the hydrophobic TatBC substrate-binding cavity. Another class of substitutions increased the positive net charge of the region's C-terminal part, presumably leading to strengthened electrostatic interactions between the mature substrate part and the cytoplasmic TatBC regions. Furthermore, we identified substitutions in the C-terminal domains of TatB following the transmembrane segment that restored transport of various transport-defective TorA-MalE derivatives. Some of these substitutions most likely affected the orientation or conformation of the flexible, carboxy-proximal helices of TatB. Therefore, we propose that a tight accommodation of the folded mature region by TatB contributes to productive binding of Tat substrates to TatBC.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Membrane Transport Proteins/metabolism , Protein Precursors/metabolism , Receptors, Cell Surface/metabolism , Twin-Arginine-Translocation System/metabolism , Amino Acid Substitution , Arginine/metabolism , Escherichia coli Proteins/chemistry , Glutamine/metabolism , Hydrophobic and Hydrophilic Interactions , Lysine/metabolism , Membrane Transport Proteins/chemistry , Oxidoreductases, N-Demethylating/metabolism , Protein Binding , Protein Transport , Receptors, Cell Surface/chemistry , Static Electricity , Substrate Specificity
14.
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
15.
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
16.
Infect Immun ; 85(4)2017 04.
Article in English | MEDLINE | ID: mdl-28115509

ABSTRACT

The twin arginine translocation (Tat) system targets folded proteins across the inner membrane and is crucial for virulence in many important human-pathogenic bacteria. Tat has been shown to be required for the virulence of Yersinia pseudotuberculosis, and we recently showed that the system is critical for different virulence-related stress responses as well as for iron uptake. In this study, we wanted to address the role of the Tat substrates in in vivo virulence. Therefore, 22 genes encoding potential Tat substrates were mutated, and each mutant was evaluated in a competitive oral infection of mice. Interestingly, a ΔsufI mutant was essentially as attenuated for virulence as the Tat-deficient strain. We also verified that SufI was Tat dependent for membrane/periplasmic localization in Y. pseudotuberculosisIn vivo bioluminescent imaging of orally infected mice revealed that both the ΔsufI and ΔtatC mutants were able to colonize the cecum and Peyer's patches (PPs) and could spread to the mesenteric lymph nodes (MLNs). Importantly, at this point, neither the ΔtatC mutant nor the ΔsufI mutant was able to spread systemically, and they were gradually cleared. Immunostaining of MLNs revealed that both the ΔtatC and ΔsufI mutants were unable to spread from the initial infection foci and appeared to be contained by neutrophils, while wild-type bacteria readily spread to establish multiple foci from day 3 postinfection. Our results show that SufI alone is required for the establishment of systemic infection and is the major cause of the attenuation of the ΔtatC mutant.


Subject(s)
Bacterial Proteins/metabolism , Twin-Arginine-Translocation System/metabolism , Yersinia pseudotuberculosis Infections/microbiology , Yersinia pseudotuberculosis/physiology , Animals , Bacterial Load , Bacterial Proteins/genetics , Female , Gene Expression , Genes, Reporter , Mice , Mutagenesis , Neutrophils/immunology , Neutrophils/metabolism , Substrate Specificity , Twin-Arginine-Translocation System/genetics , Virulence/genetics , Yersinia pseudotuberculosis/pathogenicity
17.
Curr Microbiol ; 74(11): 1332-1336, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28779357

ABSTRACT

The twin-arginine translocase (Tat) pathway is involved in the transport of folded proteins in bacteria, and has been implicated in virulence and pathogenesis. A simple but efficient assay based on the quantification of the exopolysaccharide colanic acid was developed as a new means to study Tat function. Colanic acid contains a methylpentose (L-fucose) component, and its production is directly linked to the Tat pathway through the transport of enzymes involved in polysaccharide biosynthesis. Monitoring of L-fucose levels can be applied for identification of new Tat substrates and high-throughput screening of Tat inhibitors for therapeutic applications.


Subject(s)
Bacteria/enzymology , Enzyme Assays , High-Throughput Screening Assays , Membrane Transport Proteins/metabolism , Twin-Arginine-Translocation System/metabolism , Glucans/biosynthesis , Polysaccharides/biosynthesis , Protein Transport
18.
Curr Microbiol ; 74(6): 678-684, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28326449

ABSTRACT

The twin-arginine translocation (Tat) system, which is used for folded protein secretion, is rare in lactic acid bacteria (LAB). Previously, a Tat system composed of TatAS and TatCS subunits (the subscript S denotes a Streptococcus thermophilus origin) was identified in S. thermophilus CGMCC 7.179. In the present study, the tatA S and tatC S genes were cloned and functionally analyzed in Escherichia coli DE3 tat-deficient mutants. The E. coli tatABCDE-deficient mutant complemented with tatC S A S exhibited shortened cellular chains, but its ability to grow in the presence of sodium dodecyl sulfate (SDS) was not restored, suggesting that the S. thermophilus Tat system could partially replace that of E. coli. Surprisingly, the E. coli tatABE-deficient mutant complemented with tatA S and the E. coli tatC-deficient mutant complemented with tatC S displayed relatively normal cellular morphology and enhanced tolerance to SDS. These results suggest that TatAS and TatCS could serve as active protein translocases in E. coli DE3 tat-deficient mutants. Moreover, TatAS acted as a bifunctional subunit to fulfill the roles of both TatA and TatB of E. coli DE3. Thus, this minimal Tat system would be a promising candidate to translocate recombinant proteins in LAB.


Subject(s)
Carrier Proteins/genetics , Escherichia coli/genetics , Membrane Transport Proteins/genetics , Protein Transport/genetics , Streptococcus thermophilus/genetics , Twin-Arginine-Translocation System/genetics , Amino Acid Sequence , Cloning, Molecular , Escherichia coli/growth & development , Escherichia coli/metabolism , Gene Deletion , Genetic Complementation Test , Sequence Alignment , Sodium Dodecyl Sulfate/pharmacology , Twin-Arginine-Translocation System/metabolism
19.
J Bacteriol ; 198(20): 2876-86, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27501981

ABSTRACT

UNLABELLED: The twin-arginine translocation (Tat) system mediates the secretion of folded proteins that are identified via an N-terminal signal peptide in bacteria, plants, and archaea. Tat systems are associated with virulence in many bacterial pathogens, and our previous studies revealed that Tat-deficient Yersinia pseudotuberculosis was severely attenuated for virulence. Aiming to identify Tat-dependent pathways and phenotypes of relevance for in vivo infection, we analyzed the global transcriptome of parental and ΔtatC mutant strains of Y. pseudotuberculosis during exponential and stationary growth at 26°C and 37°C. The most significant changes in the transcriptome of the ΔtatC mutant were seen at 26°C during stationary-phase growth, and these included the altered expression of genes related to virulence, stress responses, and metabolism. Subsequent phenotypic analysis based on these transcriptome changes revealed several novel Tat-dependent phenotypes, including decreased YadA expression, impaired growth under iron-limited and high-copper conditions, as well as acidic pH and SDS. Several functionally related Tat substrates were also verified to contribute to these phenotypes. Interestingly, the phenotypic defects observed in the Tat-deficient strain were generally more pronounced than those in mutants lacking the Tat substrate predicted to contribute to that specific function. Altogether, this provides new insight into the impact of Tat deficiency on in vivo fitness and survival/replication of Y. pseudotuberculosis during infection. IMPORTANCE: In addition to its established role in mediating the secretion of housekeeping enzymes, the Tat system has been recognized as being involved in infection. In some clinically relevant bacteria, such as Pseudomonas spp., several key virulence determinants can readily be identified among the Tat substrates. In enteropathogens, such as Yersinia spp., there are no obvious virulence determinants among the Tat substrates. Tat mutants show no growth defect in vitro but are highly attenuated in in vivo This makes Tat an attractive target for the development of novel antimicrobials. Therefore, it is important to establish the causes of the attenuation. Here, we show that the attenuation is likely due to synergistic effects of different Tat-dependent phenotypes that each contributes to lowered in vivo fitness.


Subject(s)
Bacterial Proteins/genetics , Twin-Arginine-Translocation System/metabolism , Yersinia pseudotuberculosis/metabolism , Bacterial Proteins/metabolism , Copper/metabolism , Gene Expression Regulation, Bacterial , Iron/metabolism , Phenotype , Protein Transport , Transcriptome , Twin-Arginine-Translocation System/genetics , Yersinia pseudotuberculosis/genetics
20.
Mol Biol Evol ; 32(10): 2598-604, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26048548

ABSTRACT

Organisms with nonphotosynthetic plastids often retain genomes; their gene contents provide clues as to the functions of these organelles. Yet the functional roles of some retained genes-such as those coding for ATP synthase-remain mysterious. In this study, we report the complete plastid genome and transcriptome data of a nonphotosynthetic diatom and propose that its ATP synthase genes may function in ATP hydrolysis to maintain a proton gradient between thylakoids and stroma, required by the twin arginine translocator (Tat) system for translocation of particular proteins into thylakoids. Given the correlated retention of ATP synthase genes and genes for the Tat system in distantly related nonphotosynthetic plastids, we suggest that this Tat-related role for ATP synthase was a key constraint during parallel loss of photosynthesis in multiple independent lineages of algae/plants.


Subject(s)
Chloroplast Proton-Translocating ATPases/metabolism , Diatoms/genetics , Genome, Plastid , Photosynthesis , Twin-Arginine-Translocation System/metabolism , Models, Biological , Phylogeny , Physical Chromosome Mapping
SELECTION OF CITATIONS
SEARCH DETAIL