101
|
Niesen MJM, Marshall SS, Miller TF, Clemons WM. Improving membrane protein expression by optimizing integration efficiency. J Biol Chem 2017; 292:19537-19545. [PMID: 28918393 DOI: 10.1074/jbc.m117.813469] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 09/12/2017] [Indexed: 12/22/2022] Open
Abstract
The heterologous overexpression of integral membrane proteins in Escherichia coli often yields insufficient quantities of purifiable protein for applications of interest. The current study leverages a recently demonstrated link between co-translational membrane integration efficiency and protein expression levels to predict protein sequence modifications that improve expression. Membrane integration efficiencies, obtained using a coarse-grained simulation approach, robustly predicted effects on expression of the integral membrane protein TatC for a set of 140 sequence modifications, including loop-swap chimeras and single-residue mutations distributed throughout the protein sequence. Mutations that improve simulated integration efficiency were 4-fold enriched with respect to improved experimentally observed expression levels. Furthermore, the effects of double mutations on both simulated integration efficiency and experimentally observed expression levels were cumulative and largely independent, suggesting that multiple mutations can be introduced to yield higher levels of purifiable protein. This work provides a foundation for a general method for the rational overexpression of integral membrane proteins based on computationally simulated membrane integration efficiencies.
Collapse
Affiliation(s)
- Michiel J M Niesen
- From the Department of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California 91125
| | - Stephen S Marshall
- From the Department of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California 91125
| | - Thomas F Miller
- From the Department of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California 91125
| | - William M Clemons
- From the Department of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California 91125
| |
Collapse
|
102
|
Findik BT, Randall LL. Determination of the intracellular concentration of the export chaperone SecB in Escherichia coli. PLoS One 2017; 12:e0183231. [PMID: 28850586 PMCID: PMC5574556 DOI: 10.1371/journal.pone.0183231] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 08/01/2017] [Indexed: 11/18/2022] Open
Abstract
SecB, a small tetrameric chaperone in Escherichia coli, plays a crucial role during protein export via the general secretory pathway by binding precursor polypeptides in a nonnative conformation and passing them to SecA, the ATPase of the translocon. The dissociation constants for the interactions are known; however to relate studies in vitro to export in a living cell requires knowledge of the concentrations of the proteins in the cell. Presently in the literature there is no report of a rigorous determination of the intracellular concentration of SecB. The values available vary over 60 fold and the details of the techniques used are not given. Here we use quantitative immunoblotting to determine the level of SecB expressed from the chromosome in E.coli grown in two commonly used media. In rich medium SecB was present at 1.6 ± 0.2 μM and in minimal medium at 2.5 ± 0.6 μM. These values allow studies of SecB carried out in vitro to be applied to the situation in the cell as SecB interacts with its binding partners to move precursor polypeptides through the export pathway.
Collapse
Affiliation(s)
- Bahar T. Findik
- Department of Biochemistry, University of Missouri, Columbia, Missouri, United States of America
| | - Linda L. Randall
- Department of Biochemistry, University of Missouri, Columbia, Missouri, United States of America
- * E-mail:
| |
Collapse
|
103
|
Abstract
BamA is an essential component of the β-barrel assembly machine (BAM) that is responsible for insertion and folding of β-barrel outer membrane proteins (OMPs) in Gram-negative bacteria. BamA is an OMP itself, and its β-barrel transmembrane domain is thought to catalyze OMP insertion and folding, although the molecular mechanism remains poorly understood. Crystal structures of BamA and complementary molecular dynamics simulations have shown that its β-barrel seam (the interface between the first and last barrel strands) is destabilized. This has led to mechanistic models in which the BamA barrel seam functions as a lateral gate that opens and successively accepts β-hairpins from a nascent OMP such that a nascent barrel can bud from BamA. Consistent with this model, disulfide locking of the BamA barrel seam is lethal in Escherichia coli. Here we show that disulfide locking of the BamA barrel has no effect on its ability to catalyze folding of a model OMP into liposomes. However, disulfide trapping experiments indicate that the BamA barrel is highly dynamic in the liposome membranes, with the β-strands at the barrel seam undergoing "register sliding" by more than 14 Å both up and down the membrane. Remarkably, these extreme dynamics were also observed in the BamA barrel in the context of the native E. coli outer membrane. These results are consistent with a model in which the BamA barrel dynamics induce defects in the outer membrane that facilitate insertion of nascent OMPs.
Collapse
Affiliation(s)
- Pamela Arden Doerner
- Department of Chemistry and Biochemistry, University of Colorado, Boulder. Boulder, CO 80309-0596
| | - Marcelo C. Sousa
- Department of Chemistry and Biochemistry, University of Colorado, Boulder. Boulder, CO 80309-0596
| |
Collapse
|
104
|
Hsieh YH, Huang YJ, Zhang H, Liu Q, Lu Y, Yang H, Houghton J, Jiang C, Sui SF, Tai PC. Dissecting structures and functions of SecA-only protein-conducting channels: ATPase, pore structure, ion channel activity, protein translocation, and interaction with SecYEG/SecDF•YajC. PLoS One 2017; 12:e0178307. [PMID: 28575061 PMCID: PMC5456053 DOI: 10.1371/journal.pone.0178307] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 05/10/2017] [Indexed: 11/30/2022] Open
Abstract
SecA is an essential protein in the major bacterial Sec-dependent translocation pathways. E. coli SecA has 901 aminoacyl residues which form multi-functional domains that interact with various ligands to impart function. In this study, we constructed and purified tethered C-terminal deletion fragments of SecA to determine the requirements for N-terminal domains interacting with lipids to provide ATPase activity, pore structure, ion channel activity, protein translocation and interactions with SecYEG-SecDF•YajC. We found that the N-terminal fragment SecAN493 (SecA1-493) has low, intrinsic ATPase activity. Larger fragments have greater activity, becoming highest around N619-N632. Lipids greatly stimulated the ATPase activities of the fragments N608-N798, reaching maximal activities around N619. Three helices in amino-acyl residues SecA619-831, which includes the "Helical Scaffold" Domain (SecA619-668) are critical for pore formation, ion channel activity, and for function with SecYEG-SecDF•YajC. In the presence of liposomes, N-terminal domain fragments of SecA form pore-ring structures at fragment-size N640, ion channel activity around N798, and protein translocation capability around N831. SecA domain fragments ranging in size between N643-N669 are critical for functional interactions with SecYEG-SecDF•YajC. In the presence of liposomes, inactive C-terminal fragments complement smaller non-functional N-terminal fragments to form SecA-only pore structures with ion channel activity and protein translocation ability. Thus, SecA domain fragment interactions with liposomes defined critical structures and functional aspects of SecA-only channels. These data provide the mechanistic basis for SecA to form primitive, low-efficiency, SecA-only protein-conducting channels, as well as the minimal parameters for SecA to interact functionally with SecYEG-SecDF•YajC to form high-efficiency channels.
Collapse
Affiliation(s)
- Ying-hsin Hsieh
- Department of Biology, Center for Biotechnology and Drug Design, Georgia State University, Atlanta, GA, United States of America
| | - Ying-ju Huang
- Department of Biology, Center for Biotechnology and Drug Design, Georgia State University, Atlanta, GA, United States of America
| | - Hao Zhang
- Department of Biology, Center for Biotechnology and Drug Design, Georgia State University, Atlanta, GA, United States of America
| | - Qian Liu
- Department of Biology, Center for Biotechnology and Drug Design, Georgia State University, Atlanta, GA, United States of America
| | - Yang Lu
- Department of Biology, Center for Biotechnology and Drug Design, Georgia State University, Atlanta, GA, United States of America
| | - Hsiuchin Yang
- Department of Biology, Center for Biotechnology and Drug Design, Georgia State University, Atlanta, GA, United States of America
| | - John Houghton
- Department of Biology, Center for Biotechnology and Drug Design, Georgia State University, Atlanta, GA, United States of America
| | - Chun Jiang
- Department of Biology, Center for Biotechnology and Drug Design, Georgia State University, Atlanta, GA, United States of America
| | - Sen-Fang Sui
- State Key Laboratory of Membrane Biology, Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing China
| | - Phang C. Tai
- Department of Biology, Center for Biotechnology and Drug Design, Georgia State University, Atlanta, GA, United States of America
| |
Collapse
|
105
|
Jomaa A, Fu YHH, Boehringer D, Leibundgut M, Shan SO, Ban N. Structure of the quaternary complex between SRP, SR, and translocon bound to the translating ribosome. Nat Commun 2017; 8:15470. [PMID: 28524878 PMCID: PMC5454536 DOI: 10.1038/ncomms15470] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 03/29/2017] [Indexed: 01/05/2023] Open
Abstract
During co-translational protein targeting, the signal recognition particle (SRP) binds to the translating ribosome displaying the signal sequence to deliver it to the SRP receptor (SR) on the membrane, where the signal peptide is transferred to the translocon. Using electron cryo-microscopy, we have determined the structure of a quaternary complex of the translating Escherichia coli ribosome, the SRP–SR in the ‘activated' state and the translocon. Our structure, supported by biochemical experiments, reveals that the SRP RNA adopts a kinked and untwisted conformation to allow repositioning of the ‘activated' SRP–SR complex on the ribosome. In addition, we observe the translocon positioned through interactions with the SR in the vicinity of the ribosome exit tunnel where the signal sequence is extending beyond its hydrophobic binding groove of the SRP M domain towards the translocon. Our study provides new insights into the mechanism of signal sequence transfer from the SRP to the translocon. Membrane proteins are inserted co-transnationally through the association between ribosome, the signal recognition particle and its receptor, and the membrane-bound translocon. Here the authors present a cryo-EM reconstruction of this quaternary complex in the activated state and propose a model for signal sequence transfer to the translocon.
Collapse
Affiliation(s)
- Ahmad Jomaa
- Department of Biology, Institute of Molecular Biology and Biophysics, Otto-Stern-Weg 5, ETH, Zurich CH-8093, Switzerland
| | - Yu-Hsien Hwang Fu
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California 91125, USA
| | - Daniel Boehringer
- Department of Biology, Institute of Molecular Biology and Biophysics, Otto-Stern-Weg 5, ETH, Zurich CH-8093, Switzerland
| | - Marc Leibundgut
- Department of Biology, Institute of Molecular Biology and Biophysics, Otto-Stern-Weg 5, ETH, Zurich CH-8093, Switzerland
| | - Shu-Ou Shan
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California 91125, USA
| | - Nenad Ban
- Department of Biology, Institute of Molecular Biology and Biophysics, Otto-Stern-Weg 5, ETH, Zurich CH-8093, Switzerland
| |
Collapse
|
106
|
Matin TR, Sigdel KP, Utjesanovic M, Marsh BP, Gallazzi F, Smith VF, Kosztin I, King GM. Single-Molecule Peptide-Lipid Affinity Assay Reveals Interplay between Solution Structure and Partitioning. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2017; 33:4057-4065. [PMID: 28343391 DOI: 10.1021/acs.langmuir.7b00100] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Interactions between short protein segments and phospholipid bilayers dictate fundamental aspects of cellular activity and have important applications in biotechnology. Yet, the lack of a suitable methodology for directly probing these interactions has hindered the mechanistic understanding. We developed a precision atomic force microscopy-based single-molecule force spectroscopy assay and probed partitioning into lipid bilayers by measuring the mechanical force experienced by a peptide. Protein segments were constructed from the peripheral membrane protein SecA, a key ATPase in bacterial secretion. We focused on the first 10 amino-terminal residues of SecA (SecA2-11) that are lipophilic. In addition to the core SecA2-11 sequence, constructs with nearly identical chemical composition but with differing geometry were used: two copies of SecA2-11 linked in series and two copies SecA2-11 linked in parallel. Lipid bilayer partitioning interactions of peptides with differing structures were distinguished. To model the energetic landscape, a theory of diffusive barrier crossing was extended to incorporate a superposition of potential barriers with variable weights. Analysis revealed two dissociation pathways for the core SecA2-11 sequence with well-separated intrinsic dissociation rates. Molecular dynamics simulations showed that the three peptides had significant conformational differences in solution that correlated well with the measured variations in the propensity to partition into the bilayer. The methodology is generalizable and can be applied to other peptide and lipid species.
Collapse
Affiliation(s)
| | | | | | | | | | - Virginia F Smith
- Department of Chemistry, United States Naval Academy , Annapolis, Maryland 21402, United States
| | | | | |
Collapse
|
107
|
Niesen MJM, Wang CY, Van Lehn RC, Miller TF. Structurally detailed coarse-grained model for Sec-facilitated co-translational protein translocation and membrane integration. PLoS Comput Biol 2017; 13:e1005427. [PMID: 28328943 PMCID: PMC5381951 DOI: 10.1371/journal.pcbi.1005427] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 04/05/2017] [Accepted: 02/28/2017] [Indexed: 01/05/2023] Open
Abstract
We present a coarse-grained simulation model that is capable of simulating the minute-timescale dynamics of protein translocation and membrane integration via the Sec translocon, while retaining sufficient chemical and structural detail to capture many of the sequence-specific interactions that drive these processes. The model includes accurate geometric representations of the ribosome and Sec translocon, obtained directly from experimental structures, and interactions parameterized from nearly 200 μs of residue-based coarse-grained molecular dynamics simulations. A protocol for mapping amino-acid sequences to coarse-grained beads enables the direct simulation of trajectories for the co-translational insertion of arbitrary polypeptide sequences into the Sec translocon. The model reproduces experimentally observed features of membrane protein integration, including the efficiency with which polypeptide domains integrate into the membrane, the variation in integration efficiency upon single amino-acid mutations, and the orientation of transmembrane domains. The central advantage of the model is that it connects sequence-level protein features to biological observables and timescales, enabling direct simulation for the mechanistic analysis of co-translational integration and for the engineering of membrane proteins with enhanced membrane integration efficiency.
Collapse
Affiliation(s)
- Michiel J. M. Niesen
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California, United States of America
| | - Connie Y. Wang
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California, United States of America
| | - Reid C. Van Lehn
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California, United States of America
| | - Thomas F. Miller
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California, United States of America
- * E-mail:
| |
Collapse
|
108
|
Bicalho MLS, Machado VS, Higgins CH, Lima FS, Bicalho RC. Genetic and functional analysis of the bovine uterine microbiota. Part I: Metritis versus healthy cows. J Dairy Sci 2017; 100:3850-3862. [PMID: 28259404 DOI: 10.3168/jds.2016-12058] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 01/14/2017] [Indexed: 12/21/2022]
Abstract
Metritis is a uterine disease that affects 10 to 30% of all lactating dairy cows and has detrimental effects on reproductive performance, milk production, and survival. Data regarding the identity and abundance of bacterial genes governing traits such as virulence, antibiotic resistance, and stress responses could enable identification of previously unknown agents that play a role in metritis pathogenesis. Moreover, such knowledge could lead to the development of improved treatments or preventive methods. Therefore, the objectives of this study were to characterize the uterine microbial population and to differentiate, for the first time, the microbial functional diversity in cows with metritis versus healthy cows. In addition, we aimed to identify relationships between microbial genes and postpartum uterine health. Uterine swabs were collected from 24 cows within 3 to 12 d in milk; 12 cows were diagnosed with metritis and the other 12 were healthy. Metritis was defined as a watery, reddish or brownish uterine discharge having a fetid smell, and rectal temperature greater than 39.5°C. Cows with a clear and viscous uterine discharge, not fetid or mucopurulent, were classified as healthy. Microbial metagenomic DNA from uterine swab samples was subjected to whole-genome shotgun sequencing on the Illumina MiSeq platform (Illumina Inc., San Diego, CA). The MG-RAST server (metagenomic rapid annotations using subsystems technology; http://metagenomics.anl.gov/) and STAMP software (http://kiwi.cs.dal.ca/Software/STAMP) were used to detect statistically significant differences in the abundance of taxonomic and functional features between the uterine microbial metagenomes of metritic and healthy cows. Our results showed an increased abundance of Fusobacteria and Bacteroidetes in metritic cows, confirming the potential role of those 2 taxa in the pathogenesis of metritis. The MG-RAST analysis revealed a significantly higher abundance of genes for protein transport across the cytoplasmic membrane and type VI bacterial secretion systems in the metritic microbiota. Additionally, genes coding for resistance to acid stress were exclusive to the metritis microbiota, suggesting that microbial resistance to acid stress is important for microbial survival in the infected uterus. On the other hand, genes coding for adhesion molecules, bacteriocins, and antibacterial peptides were significantly associated with the uterine microbiota of healthy cows, as was tolerance to colicin E2.
Collapse
Affiliation(s)
- M L S Bicalho
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853
| | - V S Machado
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853
| | - C H Higgins
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853
| | - F S Lima
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853
| | - R C Bicalho
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853.
| |
Collapse
|
109
|
Abstract
The insertion and assembly of proteins into the inner membrane of bacteria are crucial for many cellular processes, including cellular respiration, signal transduction, and ion and pH homeostasis. This process requires efficient membrane targeting and insertion of proteins into the lipid bilayer in their correct orientation and proper conformation. Playing center stage in these events are the targeting components, signal recognition particle (SRP) and the SRP receptor FtsY, as well as the insertion components, the Sec translocon and the YidC insertase. Here, we will discuss new insights provided from the recent high-resolution structures of these proteins. In addition, we will review the mechanism by which a variety of proteins with different topologies are inserted into the inner membrane of Gram-negative bacteria. Finally, we report on the energetics of this process and provide information on how membrane insertion occurs in Gram-positive bacteria and Archaea. It should be noted that most of what we know about membrane protein assembly in bacteria is based on studies conducted in Escherichia coli.
Collapse
Affiliation(s)
- Andreas Kuhn
- Institute for Microbiology and Molecular Biology, University of Hohenheim, 70599 Stuttgart, Germany
| | - Hans-Georg Koch
- Institute for Biochemistry and Molecular Biology, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, 79104, Freiburg, Germany
| | - Ross E Dalbey
- Department of Chemistry, The Ohio State University, Columbus, OH 43210
| |
Collapse
|
110
|
Dumitrache A, Klingeman DM, Natzke J, Rodriguez M, Giannone RJ, Hettich RL, Davison BH, Brown SD. Specialized activities and expression differences for Clostridium thermocellum biofilm and planktonic cells. Sci Rep 2017; 7:43583. [PMID: 28240279 PMCID: PMC5327387 DOI: 10.1038/srep43583] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 01/25/2017] [Indexed: 01/01/2023] Open
Abstract
Clostridium (Ruminiclostridium) thermocellum is a model organism for its ability to deconstruct plant biomass and convert the cellulose into ethanol. The bacterium forms biofilms adherent to lignocellulosic feedstocks in a continuous cell-monolayer in order to efficiently break down and uptake cellulose hydrolysates. We developed a novel bioreactor design to generate separate sessile and planktonic cell populations for omics studies. Sessile cells had significantly greater expression of genes involved in catabolism of carbohydrates by glycolysis and pyruvate fermentation, ATP generation by proton gradient, the anabolism of proteins and lipids and cellular functions critical for cell division consistent with substrate replete conditions. Planktonic cells had notably higher gene expression for flagellar motility and chemotaxis, cellulosomal cellulases and anchoring scaffoldins, and a range of stress induced homeostasis mechanisms such as oxidative stress protection by antioxidants and flavoprotein co-factors, methionine repair, Fe-S cluster assembly and repair in redox proteins, cell growth control through tRNA thiolation, recovery of damaged DNA by nucleotide excision repair and removal of terminal proteins by proteases. This study demonstrates that microbial attachment to cellulose substrate produces widespread gene expression changes for critical functions of this organism and provides physiological insights for two cells populations relevant for engineering of industrially-ready phenotypes.
Collapse
Affiliation(s)
- Alexandru Dumitrache
- BioEnergy Science Center (BESC), Oak Ridge National Laboratory, Oak Ridge, TN, 37831, U.S.A.,Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, 37831, U.S.A
| | - Dawn M Klingeman
- BioEnergy Science Center (BESC), Oak Ridge National Laboratory, Oak Ridge, TN, 37831, U.S.A.,Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, 37831, U.S.A
| | - Jace Natzke
- BioEnergy Science Center (BESC), Oak Ridge National Laboratory, Oak Ridge, TN, 37831, U.S.A.,Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, 37831, U.S.A
| | - Miguel Rodriguez
- BioEnergy Science Center (BESC), Oak Ridge National Laboratory, Oak Ridge, TN, 37831, U.S.A.,Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, 37831, U.S.A
| | - Richard J Giannone
- BioEnergy Science Center (BESC), Oak Ridge National Laboratory, Oak Ridge, TN, 37831, U.S.A.,Chemical Sciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, 37831, U.S.A
| | - Robert L Hettich
- BioEnergy Science Center (BESC), Oak Ridge National Laboratory, Oak Ridge, TN, 37831, U.S.A.,Chemical Sciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, 37831, U.S.A
| | - Brian H Davison
- BioEnergy Science Center (BESC), Oak Ridge National Laboratory, Oak Ridge, TN, 37831, U.S.A.,Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, 37831, U.S.A
| | - Steven D Brown
- BioEnergy Science Center (BESC), Oak Ridge National Laboratory, Oak Ridge, TN, 37831, U.S.A.,Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, 37831, U.S.A
| |
Collapse
|
111
|
Abstract
In Gram-negative bacteria, the biogenesis of β-barrel outer membrane proteins (OMPs) is mediated by the β-barrel assembly machinery (BAM) complex. During the past decade, structural and functional studies have collectively contributed to advancing our understanding of the structure and function of the BAM complex; however, the exact mechanism that is involved remains elusive. In this Progress article, we discuss recent structural studies that have revealed that the accessory proteins may regulate essential unprecedented conformational changes in the core component BamA during function. We also detail the mechanistic insights that have been gained from structural data, mutagenesis studies and molecular dynamics simulations, and explore two emerging models for the BAM-mediated biogenesis of OMPs in bacteria.
Collapse
Affiliation(s)
- Nicholas Noinaj
- Markey Center for Structural Biology, Department of Biological Sciences and the Purdue Institute for Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, Indiana 47907, USA
| | - James C Gumbart
- School of Physics, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Susan K Buchanan
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland 20892, USA
| |
Collapse
|
112
|
Weirich J, Bräutigam C, Mühlenkamp M, Franz-Wachtel M, Macek B, Meuskens I, Skurnik M, Leskinen K, Bohn E, Autenrieth I, Schütz M. Identifying components required for OMP biogenesis as novel targets for antiinfective drugs. Virulence 2017; 8:1170-1188. [PMID: 28118090 PMCID: PMC5711350 DOI: 10.1080/21505594.2016.1278333] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The emergence of multiresistant Gram-negative bacteria requires new therapies for combating bacterial infections. Targeting the biogenesis of virulence factors could be an alternative strategy instead of killing bacteria with antibiotics. The outer membrane (OM) of Gram-negative bacteria acts as a physical barrier. At the same time it facilitates the exchange of molecules and harbors a multitude of proteins associated with virulence. In order to insert proteins into the OM, an essential oligomeric membrane-associated protein complex, the ß-barrel assembly machinery (BAM) is required. Being essential for the biogenesis of outer membrane proteins (OMPs) the BAM and also periplasmic chaperones may serve as attractive targets to develop novel antiinfective agents. Herein, we aimed to elucidate which proteins belonging to the OMP biogenesis machinery have the most important function in granting bacterial fitness, OM barrier function, facilitating biogenesis of dedicated virulence factors and determination of overall virulence. To this end we used the enteropathogen Yersinia enterocolitica as a model system. We individually knocked out all non-essential components of the BAM (BamB, C and E) as well as the periplasmic chaperones DegP, SurA and Skp. In summary, we found that the most profound phenotypes were produced by the loss of BamB or SurA with both knockouts resulting in significant attenuation or even avirulence of Ye in a mouse infection model. Thus, we assume that both BamB and SurA are promising targets for the development of new antiinfective drugs in the future.
Collapse
Affiliation(s)
- Johanna Weirich
- a Institut für Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Tübingen , Tübingen , Germany
| | - Cornelia Bräutigam
- a Institut für Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Tübingen , Tübingen , Germany
| | - Melanie Mühlenkamp
- a Institut für Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Tübingen , Tübingen , Germany
| | | | - Boris Macek
- b Proteome Center Tübingen, Universität Tübingen , Tübingen , Germany
| | - Ina Meuskens
- a Institut für Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Tübingen , Tübingen , Germany
| | - Mikael Skurnik
- c Department of Bacteriology and Immunology , Medicum, Research Programs Unit, Immunobiology Research Program, University of Helsinki , Helsinki , Finland
| | - Katarzyna Leskinen
- c Department of Bacteriology and Immunology , Medicum, Research Programs Unit, Immunobiology Research Program, University of Helsinki , Helsinki , Finland
| | - Erwin Bohn
- a Institut für Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Tübingen , Tübingen , Germany
| | - Ingo Autenrieth
- a Institut für Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Tübingen , Tübingen , Germany
| | - Monika Schütz
- a Institut für Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Tübingen , Tübingen , Germany
| |
Collapse
|
113
|
Hsieh YH, Zhang H, Jin J, Dai C, Jiang C, Wang B, Tai PC. Biphasic actions of SecA inhibitors on Prl/Sec suppressors: Possible physiological roles of SecA-only channels. Biochem Biophys Res Commun 2017; 482:296-300. [PMID: 27856243 DOI: 10.1016/j.bbrc.2016.11.057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 11/11/2016] [Indexed: 11/30/2022]
Abstract
SecA is an essential component in the bacterial Sec-dependent protein translocation process. We previously showed that in addition to the ubiquitous, high-affinity SecYEG-SecDF·YajC protein translocation channel, there is a low-affinity SecA-only channel that elicits ion channel activity and promotes protein translocation. The SecA-only channels are less efficient, and like Prl suppressors, lack signal peptide specificity; they function in the absence of signal peptides. The presence of SecYEG-SecDF·YajC alters the sensitivity of ATPase inhibitor Rose Bengal. In this study, we found that the suppressor membranes are much more resistant to inhibition by Rose Bengal. Similar results have been found for a SecA-specific inhibitor. Moreover, biphasic responses of inhibition of ion current and protein translocation activities were observed for many PrlA/SecY and PrlG/SecE suppressor membranes, with a low IC50 value similar to that of the SecA-only channels and a very high IC50. However, the suppressor strains are as sensitive to the inhibitor as the parental strain, suggesting that SecA-only channels have some essential physiological function(s) in the cells that are inhibited by the specific SecA inhibitor.
Collapse
Affiliation(s)
- Ying-Hsin Hsieh
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Hao Zhang
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Jinshan Jin
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Chaofeng Dai
- Department of Chemistry, Georgia State University, Atlanta, GA 30303, USA
| | - Chun Jiang
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Binghe Wang
- Department of Chemistry, Georgia State University, Atlanta, GA 30303, USA
| | - Phang C Tai
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; Center for Biotechnology and Drug Design, Georgia State University, Atlanta, GA 30303, USA.
| |
Collapse
|
114
|
van Gijtenbeek LA, Robinson A, van Oijen AM, Poolman B, Kok J. On the Spatial Organization of mRNA, Plasmids, and Ribosomes in a Bacterial Host Overexpressing Membrane Proteins. PLoS Genet 2016; 12:e1006523. [PMID: 27977669 PMCID: PMC5201305 DOI: 10.1371/journal.pgen.1006523] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 12/30/2016] [Accepted: 12/06/2016] [Indexed: 01/03/2023] Open
Abstract
By using fluorescence imaging, we provide a time-resolved single-cell view on coupled defects in transcription, translation, and growth during expression of heterologous membrane proteins in Lactococcus lactis. Transcripts encoding poorly produced membrane proteins accumulate in mRNA-dense bodies at the cell poles, whereas transcripts of a well-expressed homologous membrane protein show membrane-proximal localization in a translation-dependent fashion. The presence of the aberrant polar mRNA foci correlates with cessation of cell division, which is restored once these bodies are cleared. In addition, activation of the heat-shock response and a loss of nucleoid-occluded ribosomes are observed. We show that the presence of a native-like N-terminal domain is key to SRP-dependent membrane localization and successful production of membrane proteins. The work presented gives new insights and detailed understanding of aberrant membrane protein biogenesis, which can be used for strategies to optimize membrane protein production.
Collapse
Affiliation(s)
- Lieke A. van Gijtenbeek
- Department of Molecular Genetics, University of Groningen, Groningen, The Netherlands
- * E-mail: (LAvG); (JK)
| | - Andrew Robinson
- Zernike Institute for Advanced Materials, University of Groningen, Groningen, The Netherlands
| | - Antoine M. van Oijen
- Zernike Institute for Advanced Materials, University of Groningen, Groningen, The Netherlands
| | - Bert Poolman
- Zernike Institute for Advanced Materials, University of Groningen, Groningen, The Netherlands
- Department of Biochemistry, University of Groningen, Groningen, The Netherlands
| | - Jan Kok
- Department of Molecular Genetics, University of Groningen, Groningen, The Netherlands
- * E-mail: (LAvG); (JK)
| |
Collapse
|
115
|
Serdiuk T, Balasubramaniam D, Sugihara J, Mari SA, Kaback HR, Müller DJ. YidC assists the stepwise and stochastic folding of membrane proteins. Nat Chem Biol 2016; 12:911-917. [PMID: 27595331 PMCID: PMC5069129 DOI: 10.1038/nchembio.2169] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Accepted: 06/14/2016] [Indexed: 11/30/2022]
Abstract
How chaperones, insertases and translocases facilitate insertion and folding of complex cytoplasmic proteins into cellular membranes is not fully understood. Here we utilize single-molecule force spectroscopy to observe YidC, a transmembrane chaperone and insertase, sculpting the folding trajectory of the polytopic α-helical membrane protein lactose permease (LacY). In the absence of YidC, unfolded LacY inserts individual structural segments into the membrane; however, misfolding dominates the process so that folding cannot be completed. YidC prevents LacY from misfolding by stabilizing the unfolded state from which LacY inserts structural segments stepwise into the membrane until folding is completed. During stepwise insertion, YidC and the membrane together stabilize the transient folds. Remarkably, the order of insertion of structural segments is stochastic, indicating that LacY can fold along variable pathways toward the native structure. Since YidC is essential in membrane protein biogenesis and LacY is a model for the major facilitator superfamily, our observations have general relevance.
Collapse
Affiliation(s)
- Tetiana Serdiuk
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Basel, Switzerland
| | | | - Junichi Sugihara
- Department of Physiology, University of California-Los Angeles, Los Angeles, USA
| | - Stefania A. Mari
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Basel, Switzerland
| | - H. Ronald Kaback
- Department of Physiology, University of California-Los Angeles, Los Angeles, USA
- Department of Microbiology, Immunology & Molecular Genetics, University of California-Los Angeles, Los Angeles, USA
- Molecular Biology Institute, University of California-Los Angeles, Los Angeles, USA
| | - Daniel J. Müller
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Basel, Switzerland
| |
Collapse
|
116
|
Liu X, Zhang W, Zhao Z, Dai X, Yang Y, Bai Z. Protein secretion in Corynebacterium glutamicum. Crit Rev Biotechnol 2016; 37:541-551. [PMID: 27737570 DOI: 10.1080/07388551.2016.1206059] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Corynebacterium glutamicum, a Gram-positive bacterium, has been widely used for the industrial production of amino acids, such as glutamate and lysine, for decades. Due to several characteristics - its ability to secrete properly folded and functional target proteins into culture broth, its low levels of endogenous extracellular proteins and its lack of detectable extracellular hydrolytic enzyme activity - C. glutamicum is also a very favorable host cell for the secretory production of heterologous proteins, important enzymes, and pharmaceutical proteins. The target proteins are secreted into the culture medium, which has attractive advantages over the manufacturing process for inclusion of body expression - the simplified downstream purification process. The secretory process of proteins is complicated and energy consuming. There are two major secretory pathways in C. glutamicum, the Sec pathway and the Tat pathway, both have specific signal peptides that mediate the secretion of the target proteins. In the present review, we critically discuss recent progress in the secretory production of heterologous proteins and examine in depth the mechanisms of the protein translocation process in C. glutamicum. Some successful case studies of actual applications of this secretory expression host are also evaluated. Finally, the existing issues and solutions in using C. glutamicum as a host of secretory proteins are specifically addressed.
Collapse
Affiliation(s)
- Xiuxia Liu
- a National Engineering Laboratory for Cereal Fermentation Technology , Jiangnan University , Wuxi , China.,b The Key Laboratory of Industrial Biotechnology, Ministry of Education , School of Biotechnology, Jiangnan University , Wuxi , China
| | - Wei Zhang
- a National Engineering Laboratory for Cereal Fermentation Technology , Jiangnan University , Wuxi , China.,b The Key Laboratory of Industrial Biotechnology, Ministry of Education , School of Biotechnology, Jiangnan University , Wuxi , China
| | - Zihao Zhao
- a National Engineering Laboratory for Cereal Fermentation Technology , Jiangnan University , Wuxi , China.,b The Key Laboratory of Industrial Biotechnology, Ministry of Education , School of Biotechnology, Jiangnan University , Wuxi , China
| | - Xiaofeng Dai
- a National Engineering Laboratory for Cereal Fermentation Technology , Jiangnan University , Wuxi , China.,b The Key Laboratory of Industrial Biotechnology, Ministry of Education , School of Biotechnology, Jiangnan University , Wuxi , China
| | - Yankun Yang
- a National Engineering Laboratory for Cereal Fermentation Technology , Jiangnan University , Wuxi , China.,b The Key Laboratory of Industrial Biotechnology, Ministry of Education , School of Biotechnology, Jiangnan University , Wuxi , China
| | - Zhonghu Bai
- a National Engineering Laboratory for Cereal Fermentation Technology , Jiangnan University , Wuxi , China.,b The Key Laboratory of Industrial Biotechnology, Ministry of Education , School of Biotechnology, Jiangnan University , Wuxi , China
| |
Collapse
|
117
|
Henderson RC, Gao F, Jayanthi S, Kight A, Sharma P, Goforth RL, Heyes CD, Henry RL, Suresh Kumar TK. Domain Organization in the 54-kDa Subunit of the Chloroplast Signal Recognition Particle. Biophys J 2016; 111:1151-1162. [PMID: 27653474 PMCID: PMC5034345 DOI: 10.1016/j.bpj.2016.08.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 07/29/2016] [Accepted: 08/02/2016] [Indexed: 10/21/2022] Open
Abstract
Chloroplast signal recognition particle (cpSRP) is a heterodimer composed of an evolutionarily conserved 54-kDa GTPase (cpSRP54) and a unique 43-kDa subunit (cpSRP43) responsible for delivering light-harvesting chlorophyll binding protein to the thylakoid membrane. While a nearly complete three-dimensional structure of cpSRP43 has been determined, no high-resolution structure is yet available for cpSRP54. In this study, we developed and examined an in silico three-dimensional model of the structure of cpSRP54 by homology modeling using cytosolic homologs. Model selection was guided by single-molecule Förster resonance energy transfer experiments, which revealed the presence of at least two distinct conformations. Small angle x-ray scattering showed that the linking region among the GTPase (G-domain) and methionine-rich (M-domain) domains, an M-domain loop, and the cpSRP43 binding C-terminal extension of cpSRP54 are predominantly disordered. Interestingly, the linker and loop segments were observed to play an important role in organizing the domain arrangement of cpSRP54. Further, deletion of the finger loop abolished loading of the cpSRP cargo, light-harvesting chlorophyll binding protein. These data highlight important structural dynamics relevant to cpSRP54's role in the post- and cotranslational signaling processes.
Collapse
Affiliation(s)
- Rory C Henderson
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, Arkansas
| | - Feng Gao
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, Arkansas
| | - Srinivas Jayanthi
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, Arkansas
| | - Alicia Kight
- Department of Biological Sciences, University of Arkansas, Fayetteville, Arkansas
| | - Priyanka Sharma
- Department of Biological Sciences, University of Arkansas, Fayetteville, Arkansas
| | - Robyn L Goforth
- Department of Biological Sciences, University of Arkansas, Fayetteville, Arkansas
| | - Colin D Heyes
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, Arkansas
| | - Ralph L Henry
- Department of Biological Sciences, University of Arkansas, Fayetteville, Arkansas
| | | |
Collapse
|
118
|
Braselmann E, Chaney JL, Champion MM, Clark PL. DegP Chaperone Suppresses Toxic Inner Membrane Translocation Intermediates. PLoS One 2016; 11:e0162922. [PMID: 27626276 PMCID: PMC5023192 DOI: 10.1371/journal.pone.0162922] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 08/30/2016] [Indexed: 11/18/2022] Open
Abstract
The periplasm of Gram-negative bacteria includes a variety of molecular chaperones that shepherd the folding and targeting of secreted proteins. A central player of this quality control network is DegP, a protease also suggested to have a chaperone function. We serendipitously discovered that production of the Bordetella pertussis autotransporter virulence protein pertactin is lethal in Escherichia coli ΔdegP strains. We investigated specific contributions of DegP to secretion of pertactin as a model system to test the functions of DegP in vivo. The DegP chaperone activity was sufficient to restore growth during pertactin production. This chaperone dependency could be relieved by changing the pertactin signal sequence: an E. coli signal sequence leading to co-translational inner membrane (IM) translocation was sufficient to suppress lethality in the absence of DegP, whereas an E. coli post-translational signal sequence was sufficient to recapitulate the lethal phenotype. These results identify a novel connection between the DegP chaperone and the mechanism used to translocate a protein across the IM. Lethality coincided with loss of periplasmic proteins, soluble σE, and proteins regulated by this essential stress response. These results suggest post-translational IM translocation can lead to the formation of toxic periplasmic folding intermediates, which DegP can suppress.
Collapse
Affiliation(s)
- Esther Braselmann
- Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, Indiana, United States of America
- * E-mail:
| | - Julie L. Chaney
- Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Matthew M. Champion
- Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Patricia L. Clark
- Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, Indiana, United States of America
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana, United States of America
| |
Collapse
|
119
|
Koch S, de Wit JG, Vos I, Birkner JP, Gordiichuk P, Herrmann A, van Oijen AM, Driessen AJM. Lipids Activate SecA for High Affinity Binding to the SecYEG Complex. J Biol Chem 2016; 291:22534-22543. [PMID: 27613865 DOI: 10.1074/jbc.m116.743831] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 08/30/2016] [Indexed: 11/06/2022] Open
Abstract
Protein translocation across the bacterial cytoplasmic membrane is an essential process catalyzed predominantly by the Sec translocase. This system consists of the membrane-embedded protein-conducting channel SecYEG, the motor ATPase SecA, and the heterotrimeric SecDFyajC membrane protein complex. Previous studies suggest that anionic lipids are essential for SecA activity and that the N terminus of SecA is capable of penetrating the lipid bilayer. The role of lipid binding, however, has remained elusive. By employing differently sized nanodiscs reconstituted with single SecYEG complexes and comprising varying amounts of lipids, we establish that SecA gains access to the SecYEG complex via a lipid-bound intermediate state, whereas acidic phospholipids allosterically activate SecA for ATP-dependent protein translocation.
Collapse
Affiliation(s)
- Sabrina Koch
- From the Molecular Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute and Zernike Institute for Advanced Materials and
| | - Janny G de Wit
- From the Molecular Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute and Zernike Institute for Advanced Materials and
| | - Iuliia Vos
- From the Molecular Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute and Zernike Institute for Advanced Materials and
| | - Jan Peter Birkner
- the Single-molecule Biophysics, Zernike Institute for Advanced Materials, University of Groningen, 9747 AG Groningen, The Netherlands
| | - Pavlo Gordiichuk
- the Polymer Chemistry and Bioengineering, Zernike Institute for Advanced Materials, 9747 AG, Groningen, The Netherlands, and
| | - Andreas Herrmann
- the Polymer Chemistry and Bioengineering, Zernike Institute for Advanced Materials, 9747 AG, Groningen, The Netherlands, and
| | - Antoine M van Oijen
- the Single-molecule Biophysics, Zernike Institute for Advanced Materials, University of Groningen, 9747 AG Groningen, The Netherlands.,the School of Chemistry, University of Wollongong, Wollongong, New South Wales 2522, Australia
| | - Arnold J M Driessen
- From the Molecular Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute and Zernike Institute for Advanced Materials and
| |
Collapse
|
120
|
Talukdar M, Das D, Bora C, Bora TC, Deka Boruah HP, Singh AK. Complete genome sequencing and comparative analyses of broad-spectrum antimicrobial-producing Micromonospora sp. HK10. Gene 2016; 594:97-107. [PMID: 27609432 DOI: 10.1016/j.gene.2016.09.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 08/29/2016] [Accepted: 09/02/2016] [Indexed: 01/21/2023]
Abstract
Micromonospora genus produces >700 bioactive compounds of medical relevance. In spite of its ability to produce high number of bioactive compounds, no genome sequence is available with comprehensive secondary metabolite gene clusters analysis for anti-microbial producing Micromonospora strains. Thus, here we contribute the full genome sequence of Micromonospora sp. HK10 strain, which has high antibacterial activity against several important human pathogens like, Mycobacterium abscessus, Mycobacterium smegmatis, Bacillus subtillis, Staphylococcus aureus, Proteus vulgaris, Pseudomonas aeruginosa, Salmonella and Escherichia coli. We have generated whole genome sequence data of Micromonospora sp. HK10 strain using Illumina NexSeq 500 sequencing platform (2×150bp paired end library) and assembled it de novo. The sequencing of HK10 genome enables identification of various genetic clusters associated with known- and probably unknown- antimicrobial compounds, which can pave the way for new antimicrobial scaffolds.
Collapse
Affiliation(s)
- Madhumita Talukdar
- Department of Biotechnology, CSIR-North East Institute of Science and Technology, Jorhat 785006, India
| | - Dhrubajyoti Das
- Department of Biotechnology, CSIR-North East Institute of Science and Technology, Jorhat 785006, India
| | - Chiranjeeta Bora
- Department of Biotechnology, CSIR-North East Institute of Science and Technology, Jorhat 785006, India
| | - Tarun Chandra Bora
- Department of Biotechnology, CSIR-North East Institute of Science and Technology, Jorhat 785006, India
| | - Hari Prasanna Deka Boruah
- Department of Biotechnology, CSIR-North East Institute of Science and Technology, Jorhat 785006, India
| | - Anil Kumar Singh
- Department of Biotechnology, CSIR-North East Institute of Science and Technology, Jorhat 785006, India; Academy of Scientific and Innovative Research, Rafi Marg, New Delhi 110001, India.
| |
Collapse
|
121
|
Global profiling of SRP interaction with nascent polypeptides. Nature 2016; 536:219-23. [PMID: 27487212 DOI: 10.1038/nature19070] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 06/29/2016] [Indexed: 12/23/2022]
Abstract
Signal recognition particle (SRP) is a universally conserved protein-RNA complex that mediates co-translational protein translocation and membrane insertion by targeting translating ribosomes to membrane translocons. The existence of parallel co- and post-translational transport pathways, however, raises the question of the cellular substrate pool of SRP and the molecular basis of substrate selection. Here we determine the binding sites of bacterial SRP within the nascent proteome of Escherichia coli at amino acid resolution, by sequencing messenger RNA footprints of ribosome-nascent-chain complexes associated with SRP. SRP, on the basis of its strong preference for hydrophobic transmembrane domains (TMDs), constitutes a compartment-specific targeting factor for nascent inner membrane proteins (IMPs) that efficiently excludes signal-sequence-containing precursors of periplasmic and outer membrane proteins. SRP associates with hydrophobic TMDs enriched in consecutive stretches of hydrophobic and bulky aromatic amino acids immediately on their emergence from the ribosomal exit tunnel. By contrast with current models, N-terminal TMDs are frequently skipped and TMDs internal to the polypeptide sequence are selectively recognized. Furthermore, SRP binds several TMDs in many multi-spanning membrane proteins, suggesting cycles of SRP-mediated membrane targeting. SRP-mediated targeting is not accompanied by a transient slowdown of translation and is not influenced by the ribosome-associated chaperone trigger factor (TF), which has a distinct substrate pool and acts at different stages during translation. Overall, our proteome-wide data set of SRP-binding sites reveals the underlying principles of pathway decisions for nascent chains in bacteria, with SRP acting as the dominant triaging factor, sufficient to separate IMPs from substrates of the SecA-SecB post-translational translocation and TF-assisted cytosolic protein folding pathways.
Collapse
|
122
|
Dajkovic A, Hinde E, MacKichan C, Carballido-Lopez R. Dynamic Organization of SecA and SecY Secretion Complexes in the B. subtilis Membrane. PLoS One 2016; 11:e0157899. [PMID: 27336478 PMCID: PMC4918944 DOI: 10.1371/journal.pone.0157899] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 06/07/2016] [Indexed: 12/21/2022] Open
Abstract
In prokaryotes, about one third of cellular proteins are translocated across the plasma membrane or inserted into it by concerted action of the cytoplasmic ATPase SecA and the universally conserved SecYEG heterotrimeric polypeptide-translocating pore. Secretion complexes have been reported to localize in specific subcellular sites in Bacillus subtilis. In this work, we used a combination of total internal reflection microscopy, scanning fluorescence correlation spectroscopy, and pair correlation function to study the localization and dynamics of SecA and SecY in growing Bacillus subtilis cells. Both SecA and SecY localized in transient and dynamic foci in the cytoplasmic membrane, which displayed no higher-level organization in helices. Foci of SecA and SecY were in constant flux with freely diffusing SecA and SecY molecules. Scanning FCS confirmed the existence of populations of cellular SecA and SecY molecules with a wide range of diffusion coefficients. Diffusion of SecY as an uncomplexed molecular species was short-lived and only local while SecY complexed with its protein partners traversed distances of over half a micrometer in the cell.
Collapse
Affiliation(s)
- Alex Dajkovic
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
- * E-mail:
| | - Elizabeth Hinde
- School of Medical Sciences, University of New South Wales, Sydney, Australia 2052
| | - Calum MacKichan
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Rut Carballido-Lopez
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| |
Collapse
|
123
|
Moffitt JR, Pandey S, Boettiger AN, Wang S, Zhuang X. Spatial organization shapes the turnover of a bacterial transcriptome. eLife 2016; 5. [PMID: 27198188 PMCID: PMC4874777 DOI: 10.7554/elife.13065] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 03/20/2016] [Indexed: 12/21/2022] Open
Abstract
Spatial organization of the transcriptome has emerged as a powerful means for regulating the post-transcriptional fate of RNA in eukaryotes; however, whether prokaryotes use RNA spatial organization as a mechanism for post-transcriptional regulation remains unclear. Here we used super-resolution microscopy to image the E. coli transcriptome and observed a genome-wide spatial organization of RNA: mRNAs encoding inner-membrane proteins are enriched at the membrane, whereas mRNAs encoding outer-membrane, cytoplasmic and periplasmic proteins are distributed throughout the cytoplasm. Membrane enrichment is caused by co-translational insertion of signal peptides recognized by the signal-recognition particle. Time-resolved RNA-sequencing revealed that degradation rates of inner-membrane-protein mRNAs are on average greater that those of the other mRNAs and that this selective destabilization of inner-membrane-protein mRNAs is abolished by dissociating the RNA degradosome from the membrane. Together, these results demonstrate that the bacterial transcriptome is spatially organized and suggest that this organization shapes the post-transcriptional dynamics of mRNAs. DOI:http://dx.doi.org/10.7554/eLife.13065.001 Within a cell, molecules of messenger RNA (mRNA) encode the proteins that the cell needs to survive and thrive. The amount of mRNA within a cell therefore plays an important role in determining both the amount and types of proteins that a cell contains and, thus, the behavior of the cell. In eukaryotic organisms, like humans, it has been established that it is not just the amount of mRNA that influences cell behavior, but also where the mRNA molecules are found within the cell. However, in bacteria, which are much smaller than human cells, it has long been believed that the location of an mRNA within the cell does not affect its behavior. Despite this, recent studies that have looked at small numbers of bacterial mRNAs have shown that some of these molecules are found in larger numbers than usual at certain sites inside cells. This suggests that location may actually affect the activity of some bacterial mRNAs. But do similar localization patterns occur for all of the thousands of different mRNAs that bacteria can make? To address this question, Moffitt et al. developed an approach that allows large, defined sets of mRNAs to be imaged in bacteria. Using this approach to study E. coli revealed that a considerable fraction of all the mRNAs that these bacteria can make locate themselves at specific sites within a cell. For example, mRNAs that encode proteins that reside inside the cell’s inner membrane are found enriched at this membrane. This localization also plays an important role in the life of these mRNAs, as they are degraded more quickly than those found elsewhere in the cell. This enhanced degradation rate arises partly because the enzymes that break down mRNA molecules are also found at the membrane. Thus, bacteria can shape the process by which an mRNA is made into protein by controlling where in a cell the mRNA is located. The next steps are to understand why bacteria use cell location to influence the rate of mRNA degradation. DOI:http://dx.doi.org/10.7554/eLife.13065.002
Collapse
Affiliation(s)
- Jeffrey R Moffitt
- Howard Hughes Medical Institute, Harvard University, Cambridge, United States.,Department of Chemistry and Chemical Biology, Harvard University, Cambridge, United States
| | - Shristi Pandey
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, United States
| | - Alistair N Boettiger
- Howard Hughes Medical Institute, Harvard University, Cambridge, United States.,Department of Chemistry and Chemical Biology, Harvard University, Cambridge, United States
| | - Siyuan Wang
- Howard Hughes Medical Institute, Harvard University, Cambridge, United States.,Department of Chemistry and Chemical Biology, Harvard University, Cambridge, United States
| | - Xiaowei Zhuang
- Howard Hughes Medical Institute, Harvard University, Cambridge, United States.,Department of Chemistry and Chemical Biology, Harvard University, Cambridge, United States.,Department of Physics, Harvard University, Cambridge, United States
| |
Collapse
|
124
|
Burmann BM, Holdbrook DA, Callon M, Bond PJ, Hiller S. Revisiting the interaction between the chaperone Skp and lipopolysaccharide. Biophys J 2016; 108:1516-1526. [PMID: 25809264 DOI: 10.1016/j.bpj.2015.01.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 12/22/2014] [Accepted: 01/28/2015] [Indexed: 10/23/2022] Open
Abstract
The bacterial outer membrane comprises two main classes of components, lipids and membrane proteins. These nonsoluble compounds are conveyed across the aqueous periplasm along specific molecular transport routes: the lipid lipopolysaccharide (LPS) is shuttled by the Lpt system, whereas outer membrane proteins (Omps) are transported by chaperones, including the periplasmic Skp. In this study, we revisit the specificity of the chaperone-lipid interaction of Skp and LPS. High-resolution NMR spectroscopy measurements indicate that LPS interacts with Skp nonspecifically, accompanied by destabilization of the Skp trimer and similar to denaturation by the nonnatural detergent lauryldimethylamine-N-oxide (LDAO). Bioinformatic analysis of amino acid conservation, structural analysis of LPS-binding proteins, and MD simulations further confirm the absence of a specific LPS binding site on Skp, making a biological relevance of the interaction unlikely. Instead, our analysis reveals a highly conserved salt-bridge network, which likely has a role for Skp function.
Collapse
Affiliation(s)
| | | | | | - Peter J Bond
- Bioinformatics Institute (A(∗)STAR), Singapore; Department of Biological Sciences, National University of Singapore, Singapore
| | | |
Collapse
|
125
|
Abstract
There is a consensus in the medical profession of the pressing need for novel antimicrobial agents due to issues related to drug resistance. In practice, solutions to this problem to a large degree lie with the identification of new and vital targets in bacteria and subsequently designing their inhibitors. We consider SecA a very promising antimicrobial target. In this review, we compile and analyze information available on SecA to show that inhibition of SecA has a multitude of consequences. Furthermore, we discuss issues critical to the design and evaluation of SecA inhibitors.
Collapse
|
126
|
Gagic D, Ciric M, Wen WX, Ng F, Rakonjac J. Exploring the Secretomes of Microbes and Microbial Communities Using Filamentous Phage Display. Front Microbiol 2016; 7:429. [PMID: 27092113 PMCID: PMC4823517 DOI: 10.3389/fmicb.2016.00429] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 03/17/2016] [Indexed: 01/12/2023] Open
Abstract
Microbial surface and secreted proteins (the secretome) contain a large number of proteins that interact with other microbes, host and/or environment. These proteins are exported by the coordinated activities of the protein secretion machinery present in the cell. A group of bacteriophage, called filamentous phage, have the ability to hijack bacterial protein secretion machinery in order to amplify and assemble via a secretion-like process. This ability has been harnessed in the use of filamentous phage of Escherichia coli in biotechnology applications, including screening large libraries of variants for binding to “bait” of interest, from tissues in vivo to pure proteins or even inorganic substrates. In this review we discuss the roles of secretome proteins in pathogenic and non-pathogenic bacteria and corresponding secretion pathways. We describe the basics of phage display technology and its variants applied to discovery of bacterial proteins that are implicated in colonization of host tissues and pathogenesis, as well as vaccine candidates through filamentous phage display library screening. Secretome selection aided by next-generation sequence analysis was successfully applied for selective display of the secretome at a microbial community scale, the latter revealing the richness of secretome functions of interest and surprising versatility in filamentous phage display of secretome proteins from large number of Gram-negative as well as Gram-positive bacteria and archaea.
Collapse
Affiliation(s)
- Dragana Gagic
- Institute of Fundamental Sciences, Massey UniversityPalmerston North, New Zealand; Animal Science, Grasslands Research Centre, AgResearch Ltd, Palmerston NorthNew Zealand
| | - Milica Ciric
- Institute of Fundamental Sciences, Massey UniversityPalmerston North, New Zealand; Animal Science, Grasslands Research Centre, AgResearch Ltd, Palmerston NorthNew Zealand
| | - Wesley X Wen
- Institute of Fundamental Sciences, Massey University Palmerston North, New Zealand
| | - Filomena Ng
- Animal Science, Grasslands Research Centre, AgResearch Ltd, Palmerston North New Zealand
| | - Jasna Rakonjac
- Institute of Fundamental Sciences, Massey University Palmerston North, New Zealand
| |
Collapse
|
127
|
Górska S, Buda B, Brzozowska E, Schwarzer M, Srutkova D, Kozakova H, Gamian A. Identification of Lactobacillus proteins with different recognition patterns between immune rabbit sera and nonimmune mice or human sera. BMC Microbiol 2016; 16:17. [PMID: 26861940 PMCID: PMC4748627 DOI: 10.1186/s12866-016-0631-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 01/22/2016] [Indexed: 11/23/2022] Open
Abstract
Background The genus Lactobacillus belongs to a large heterogeneous group of low G + C Gram-positive anaerobic bacteria, which are frequently used as probiotics. The health-beneficial effects, in particular the immunomodulation effect, of probiotics depend on the strain and dose used. Strain variations may be related to diversity of the cell surface architecture of bacteria and the ability to express specific antigens or secrete compounds. The use of Lactobacillus as probiotic requires a comprehensive understanding of its effect on host immune system. To evaluate the potential immunoreactive properties of proteins isolated from four Lactobacillus strains: L. johnsonii 142 and L. johnsonii 151, L. rhamnosus LOCK 0900 and L. casei LOCK 0919, the polyclonal sera obtained from mouse and human have been tested as well as with sera from rabbits immunized with whole lactobacilli cells. Results The reactivity of isolated proteins detected by SDS-PAGE and Western blotting was heterogeneous and varied between different serum samples. The proteins with the highest immunoreactivity were isolated, purified and sequenced, in particular the fractions were identified as phosphoglycerate kinase (L. johnsonii 142), glyceraldehyde 3-phosphate dehydrogenase (L. johnosnii 142, L. rhamnosus LOCK 0900), hypothetic protein JDM1_1307 (L. johnsonii 151) and fructose/tagatose-bisphosphate-aldolase (L. casei LOCK 0919). Conclusion The different prevalence of reactions against tested antigens in rabbit, mouse and human sera may indicate significant differences in immune system and commensal cross-talk in these groups. The identification of immunoreactive lactobacilli proteins opens the possibility to use them as an antigens for development of vaccines.
Collapse
Affiliation(s)
- Sabina Górska
- Department of Medical Microbiology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy of the Polish Academy of Sciences, Wroclaw, Poland.
| | - Barbara Buda
- Department of Animal Products Technology and Quality Management, Wroclaw University of Environmental and Life Sciences, Faculty of Food Science, Wroclaw, Poland
| | - Ewa Brzozowska
- Department of Medical Microbiology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy of the Polish Academy of Sciences, Wroclaw, Poland
| | - Martin Schwarzer
- Institute of Microbiology, Laboratory of Gnotobiology, Academy of Sciences of the Czech Republic v. v. i., 549 22, Novy Hradek, Czech Republic
| | - Dagmar Srutkova
- Institute of Microbiology, Laboratory of Gnotobiology, Academy of Sciences of the Czech Republic v. v. i., 549 22, Novy Hradek, Czech Republic
| | - Hana Kozakova
- Institute of Microbiology, Laboratory of Gnotobiology, Academy of Sciences of the Czech Republic v. v. i., 549 22, Novy Hradek, Czech Republic
| | - Andrzej Gamian
- Department of Medical Microbiology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy of the Polish Academy of Sciences, Wroclaw, Poland
| |
Collapse
|
128
|
Yan S, Wu G. Evolutionary evidence on suitability of SecD as a target for development of antibacterial agents against Staphylococcus aureus. Ecol Evol 2016; 6:1393-410. [PMID: 27087922 PMCID: PMC4775529 DOI: 10.1002/ece3.1951] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 12/15/2015] [Accepted: 12/16/2015] [Indexed: 11/22/2022] Open
Abstract
Staphylococcus aureus causes many infections and its drug resistance is a worrying challenge for medical care. The SecD subunit of Sec secretion system in methicillin‐resistant S. aureus is an attractive target because SecD dysfunction leads to the death of bacteria and SecD as a target is more efficient than SecA and SecF. Evolution could have made SecD to become insensitive to antibacterial agents although the drugs directly against SecD have yet to develop. So far, no detailed information on SecD evolution has been available, thus 2686 SecD sequences with full taxonomic information from kingdom to species were analyzed. First, the variance of pairwise p‐distance was evaluated for each taxonomic group. Second, the variance was further partitioned into intergroup and intragroup variances for quantification of horizontal and vertical gene transfer. Third, phylogenetic tree was built to trace the evolutionary pathway. The results showed that overall evolution of SecDs appears to have undergone horizontal and vertical gene transfer. Only 0.5% horizontal transfers were found between any two SecDs in S. aureus, 6.8% and 8.8% horizontal transfers were found between any two Staphylococcus SecDs from different and the same species, and only one SecD from S. aureus was located far away from its sister cluster. Thus, statistic and evolutionary analyses demonstrate that the SecDs from staphylococcus species have a small chance of mutating, and provide taxonomic evidence to use the SecD as a potential target for new generation of antibacterial agents against S. aureus.
Collapse
Affiliation(s)
- Shaomin Yan
- Guangxi Bioscience and Biotechnology Research Center Guangxi Academy of Sciences 98 Daling Road Nanning Guangxi 530007 China
| | - Guang Wu
- Guangxi Bioscience and Biotechnology Research Center Guangxi Academy of Sciences 98 Daling Road Nanning Guangxi 530007 China
| |
Collapse
|
129
|
Knapp A, Voget S, Gao R, Zaburannyi N, Krysciak D, Breuer M, Hauer B, Streit WR, Müller R, Daniel R, Jaeger KE. Mutations improving production and secretion of extracellular lipase by Burkholderia glumae PG1. Appl Microbiol Biotechnol 2016; 100:1265-1273. [PMID: 26476653 PMCID: PMC4717159 DOI: 10.1007/s00253-015-7041-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 09/15/2015] [Accepted: 09/24/2015] [Indexed: 01/05/2023]
Abstract
Burkholderia glumae is a Gram-negative phytopathogenic bacterium known as the causative agent of rice panicle blight. Strain B. glumae PG1 is used for the production of a biotechnologically relevant lipase, which is secreted into the culture supernatant via a type II secretion pathway. We have comparatively analyzed the genome sequences of B. glumae PG1 wild type and a lipase overproducing strain obtained by classical strain mutagenesis. Among a total number of 72 single nucleotide polymorphisms (SNPs) identified in the genome of the production strain, two were localized in front of the lipAB operon and were analyzed in detail. Both mutations contribute to a 100-fold overproduction of extracellular lipase in B. glumae PG1 by affecting transcription of the lipAB operon and efficiency of lipase secretion. We analyzed each of the two SNPs separately and observed a stronger influence of the promoter mutation than of the signal peptide modification but also a cumulative effect of both mutations. Furthermore, fusion of the mutated LipA signal peptide resulted in a 2-fold increase in secretion of the heterologous reporter alkaline phosphatase from Escherichia coli.
Collapse
Affiliation(s)
- Andreas Knapp
- Institute of Molecular Enzyme Technology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Sonja Voget
- Institute of Microbiology and Genetics, Department of Genomic and Applied Microbiology and Göttingen Genomics Laboratory, Georg-August University Göttingen, Göttingen, Germany
| | - Rong Gao
- Biocenter Klein Flottbek, Department of Microbiology and Biotechnology, University of Hamburg, Hamburg, Germany
| | - Nestor Zaburannyi
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research and Pharmaceutical Biotechnology, Saarland University, Saarbrücken, Germany
| | - Dagmar Krysciak
- Biocenter Klein Flottbek, Department of Microbiology and Biotechnology, University of Hamburg, Hamburg, Germany
| | - Michael Breuer
- BASF SE, Biocatalysis and Fine Chemicals Research, Ludwigshafen, Germany
| | - Bernhard Hauer
- BASF SE, Biocatalysis and Fine Chemicals Research, Ludwigshafen, Germany
- Institute of Technical Biochemistry, University of Stuttgart, Stuttgart, Germany
| | - Wolfgang R Streit
- Biocenter Klein Flottbek, Department of Microbiology and Biotechnology, University of Hamburg, Hamburg, Germany
| | - Rolf Müller
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research and Pharmaceutical Biotechnology, Saarland University, Saarbrücken, Germany
| | - Rolf Daniel
- Institute of Microbiology and Genetics, Department of Genomic and Applied Microbiology and Göttingen Genomics Laboratory, Georg-August University Göttingen, Göttingen, Germany
| | - Karl-Erich Jaeger
- Institute of Molecular Enzyme Technology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany.
- Forschungszentrum Jülich GmbH, Institute of Bio- and Geosciences IBG-1: Biotechnology, Jülich, Germany.
| |
Collapse
|
130
|
Jomaa A, Boehringer D, Leibundgut M, Ban N. Structures of the E. coli translating ribosome with SRP and its receptor and with the translocon. Nat Commun 2016; 7:10471. [PMID: 26804923 PMCID: PMC4737761 DOI: 10.1038/ncomms10471] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 12/14/2015] [Indexed: 12/17/2022] Open
Abstract
Co-translational protein targeting to membranes is a universally conserved process. Central steps include cargo recognition by the signal recognition particle and handover to the Sec translocon. Here we present snapshots of key co-translational-targeting complexes solved by cryo-electron microscopy at near-atomic resolution, establishing the molecular contacts between the Escherichia coli translating ribosome, the signal recognition particle and the translocon. Our results reveal the conformational changes that regulate the latching of the signal sequence, the release of the heterodimeric domains of the signal recognition particle and its receptor, and the handover of the signal sequence to the translocon. We also observe that the signal recognition particle and the translocon insert-specific structural elements into the ribosomal tunnel to remodel it, possibly to sense nascent chains. Our work provides structural evidence for a conformational state of the signal recognition particle and its receptor primed for translocon binding to the ribosome-nascent chain complex.
Collapse
Affiliation(s)
- Ahmad Jomaa
- Department of Biology, Institute of Molecular Biology and Biophysics, Otto-Stern-Weg 5, ETH Zurich CH-8093, Switzerland
| | - Daniel Boehringer
- Department of Biology, Institute of Molecular Biology and Biophysics, Otto-Stern-Weg 5, ETH Zurich CH-8093, Switzerland
| | - Marc Leibundgut
- Department of Biology, Institute of Molecular Biology and Biophysics, Otto-Stern-Weg 5, ETH Zurich CH-8093, Switzerland
| | - Nenad Ban
- Department of Biology, Institute of Molecular Biology and Biophysics, Otto-Stern-Weg 5, ETH Zurich CH-8093, Switzerland
| |
Collapse
|
131
|
Abstract
The β-barrel outer membrane proteins (OMPs) are integral membrane proteins that reside in the outer membrane of Gram-negative bacteria and perform a diverse range of biological functions. Synthesized in the cytoplasm, OMPs must be transported across the inner membrane and through the periplasmic space before they are assembled in the outer membrane. In Escherichia coli, Skp, SurA and DegP are the most prominent factors identified to guide OMPs across the periplasm and to play the role of quality control. Although extensive genetic and biochemical analyses have revealed many basic functions of these periplasmic proteins, the mechanism of their collaboration in assisting the folding and insertion of OMPs is much less understood. Recently, biophysical approaches have shed light on the identification of the intricate network. In the present review, we summarize recent advances in the characterization of these key factors, with a special emphasis on the multifunctional protein DegP. In addition, we present our proposed model on the periplasmic quality control in biogenesis of OMPs.
Collapse
|
132
|
Prabudiansyah I, Driessen AJM. The Canonical and Accessory Sec System of Gram-positive Bacteria. Curr Top Microbiol Immunol 2016; 404:45-67. [DOI: 10.1007/82_2016_9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
133
|
Snijder HJA, Hakulinen J. Membrane Protein Production in E. coli for Applications in Drug Discovery. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 896:59-77. [PMID: 27165319 DOI: 10.1007/978-3-319-27216-0_5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Producing high quality purified membrane proteins for structure-based drug design and biophysical assays compatible with typical timelines in drug discovery is a significant challenge. Escherichia coli has been an expression host of the utmost importance for soluble proteins and has applications for membrane proteins as well. However, membrane protein overexpression in E. coli may lead to toxicity and low yields of functional product. Here, we review the challenges encountered with heterologous overproduction of α-helical membrane proteins in E. coli and a range of strategies to overcome them. A detailed protocol is also provided for expression and screening of membrane proteins in E. coli using a His-specific fluorescent probe and fluorescent size-exclusion chromatography.
Collapse
Affiliation(s)
| | - Jonna Hakulinen
- Discovery Sciences, AstraZeneca R&D, SE-43183, Mölndal, Sweden
| |
Collapse
|
134
|
Horn A, Hennig J, Ahmed YL, Stier G, Wild K, Sattler M, Sinning I. Structural basis for cpSRP43 chromodomain selectivity and dynamics in Alb3 insertase interaction. Nat Commun 2015; 6:8875. [PMID: 26568381 PMCID: PMC4660199 DOI: 10.1038/ncomms9875] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 10/12/2015] [Indexed: 01/21/2023] Open
Abstract
Canonical membrane protein biogenesis requires co-translational delivery of ribosome-associated proteins to the Sec translocase and depends on the signal recognition particle (SRP) and its receptor (SR). In contrast, high-throughput delivery of abundant light-harvesting chlorophyll a,b-binding proteins (LHCPs) in chloroplasts to the Alb3 insertase occurs post-translationally via a soluble transit complex including the cpSRP43/cpSRP54 heterodimer (cpSRP). Here we describe the molecular mechanisms of tethering cpSRP to the Alb3 insertase by specific interaction of cpSRP43 chromodomain 3 with a linear motif in the Alb3 C-terminal tail. Combining NMR spectroscopy, X-ray crystallography and biochemical analyses, we dissect the structural basis for selectivity of chromodomains 2 and 3 for their respective ligands cpSRP54 and Alb3, respectively. Negative cooperativity in ligand binding can be explained by dynamics in the chromodomain interface. Our study provides a model for membrane recruitment of the transit complex and may serve as a prototype for a functional gain by the tandem arrangement of chromodomains. The chloroplast signal recognition particle delivers LHCPs to the thylakoid membrane by interaction of cpSRP43 with the Alb3 insertase. Here the authors decipher the specific recognition of the Alb3 C-terminal tail within the interface of two communicating chromodomains by structural biochemistry.
Collapse
Affiliation(s)
- Annemarie Horn
- Heidelberg University Biochemistry Center (BZH), INF 328, Heidelberg D-69120, Germany
| | - Janosch Hennig
- Center for Integrated Protein Science Munich at Biomolecular NMR Spectroscopy, Department Chemie, Technische Universität München, Lichtenbergstrasse 4, Garching DE-85747, Germany.,Institute of Structural Biology, Helmholtz Center Munich, Ingolstädter Landstrasse 1, Neuherberg D-85764, Germany
| | - Yasar L Ahmed
- Heidelberg University Biochemistry Center (BZH), INF 328, Heidelberg D-69120, Germany
| | - Gunter Stier
- Heidelberg University Biochemistry Center (BZH), INF 328, Heidelberg D-69120, Germany
| | - Klemens Wild
- Heidelberg University Biochemistry Center (BZH), INF 328, Heidelberg D-69120, Germany
| | - Michael Sattler
- Center for Integrated Protein Science Munich at Biomolecular NMR Spectroscopy, Department Chemie, Technische Universität München, Lichtenbergstrasse 4, Garching DE-85747, Germany.,Institute of Structural Biology, Helmholtz Center Munich, Ingolstädter Landstrasse 1, Neuherberg D-85764, Germany
| | - Irmgard Sinning
- Heidelberg University Biochemistry Center (BZH), INF 328, Heidelberg D-69120, Germany
| |
Collapse
|
135
|
Jin J, Cui J, Chaudhary AS, Hsieh YH, Damera K, Zhang H, Yang H, Wang B, Tai PC. Evaluation of small molecule SecA inhibitors against methicillin-resistant Staphylococcus aureus. Bioorg Med Chem 2015; 23:7061-8. [PMID: 26432604 PMCID: PMC4661110 DOI: 10.1016/j.bmc.2015.09.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Revised: 09/09/2015] [Accepted: 09/16/2015] [Indexed: 12/11/2022]
Abstract
Due to the emergence and rapid spread of drug resistance in bacteria, there is an urgent need for the development of novel antimicrobials. SecA, a key component of the general bacterial secretion system required for viability and virulence, is an attractive antimicrobial target. Earlier we reported that systematical dissection of a SecA inhibitor, Rose Bengal (RB), led to the development of novel small molecule SecA inhibitors active against Escherichia coli and Bacillus subtilis. In this study, two potent RB analogs were further evaluated for activities against methicillin-resistant Staphylococcus aureus (MRSA) strains and for their mechanism of actions. These analogs showed inhibition on the ATPase activities of S. aureus SecA1 (SaSecA1) and SecA2 (SaSecA2), and inhibition of SaSecA1-dependent protein-conducting channel. Moreover, these inhibitors reduce the secretion of three toxins from S. aureus and exert potent bacteriostatic effects against three MRSA strains. Our best inhibitor SCA-50 showed potent concentration-dependent bactericidal activity against MRSA Mu50 strain and very importantly, 2-60 fold more potent inhibitory effect on MRSA Mu50 than all the commonly used antibiotics including vancomycin, which is considered the last resort option in treating MRSA-related infections. Protein pull down experiments further confirmed SaSecA1 as a target. Deletion or overexpression of NorA and MepA efflux pumps had minimal effect on the antimicrobial activities against S. aureus, indicating that the effects of SecA inhibitors were not affected by the presence of these efflux pumps. Our studies show that these small molecule analogs target SecA functions, have potent antimicrobial activities, reduce the secretion of toxins, and have the ability to overcome the effect efflux pumps, which are responsible for multi-drug resistance. Thus, targeting SecA is an attractive antimicrobial strategy against MRSA.
Collapse
Affiliation(s)
- Jinshan Jin
- Department of Biology, Center for Biotechnology and Drug Design, Georgia State University, Atlanta, GA 30303, USA.
| | - Jianmei Cui
- Department of Chemistry, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA.
| | - Arpana Sagwal Chaudhary
- Department of Chemistry, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA.
| | - Ying-Hsin Hsieh
- Department of Biology, Center for Biotechnology and Drug Design, Georgia State University, Atlanta, GA 30303, USA.
| | - Krishna Damera
- Department of Chemistry, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA.
| | - Hao Zhang
- Department of Biology, Center for Biotechnology and Drug Design, Georgia State University, Atlanta, GA 30303, USA.
| | - Hsiuchin Yang
- Department of Biology, Center for Biotechnology and Drug Design, Georgia State University, Atlanta, GA 30303, USA.
| | - Binghe Wang
- Department of Chemistry, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA.
| | - Phang C Tai
- Department of Biology, Center for Biotechnology and Drug Design, Georgia State University, Atlanta, GA 30303, USA.
| |
Collapse
|
136
|
Goolab S, Roth RL, van Heerden H, Crampton MC. Analyzing the molecular mechanism of lipoprotein localization in Brucella. Front Microbiol 2015; 6:1189. [PMID: 26579096 PMCID: PMC4623201 DOI: 10.3389/fmicb.2015.01189] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 10/12/2015] [Indexed: 01/18/2023] Open
Abstract
Bacterial lipoproteins possess diverse structure and functionality, ranging from bacterial physiology to pathogenic processes. As such many lipoproteins, originating from Brucella are exploited as potential vaccines to countermeasure brucellosis infection in the host. These membrane proteins are translocated from the cytoplasm to the cell membrane where they are anchored peripherally by a multifaceted targeting mechanism. Although much research has focused on the identification and classification of Brucella lipoproteins and their potential use as vaccine candidates for the treatment of Brucellosis, the underlying route for the translocation of these lipoproteins to the outer surface of the Brucella (and other pathogens) outer membrane (OM) remains mostly unknown. This is partly due to the complexity of the organism and evasive tactics used to escape the host immune system, the variation in biological structure and activity of lipoproteins, combined with the complex nature of the translocation machinery. The biosynthetic pathway of Brucella lipoproteins involves a distinct secretion system aiding translocation from the cytoplasm, where they are modified by lipidation, sorted by the lipoprotein localization machinery pathway and thereafter equipped for export to the OM. Surface localized lipoproteins in Brucella may employ a lipoprotein flippase or the β-barrel assembly complex for translocation. This review provides an overview of the characterized Brucella OM proteins that form part of the OM, including a handful of other characterized bacterial lipoproteins and their mechanisms of translocation. Lipoprotein localization pathways in gram negative bacteria will be used as a model to identify gaps in Brucella lipoprotein localization and infer a potential pathway. Of particular interest are the dual topology lipoproteins identified in Escherichia coli and Haemophilus influenza. The localization and topology of these lipoproteins from other gram negative bacteria are well characterized and may be useful to infer a solution to better understand the translocation process in Brucella.
Collapse
Affiliation(s)
- Shivani Goolab
- Protein Technologies, Biosciences, Council for Scientific and Industrial ResearchPretoria, South Africa
- Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of PretoriaPretoria, South Africa
| | - Robyn L. Roth
- Protein Technologies, Biosciences, Council for Scientific and Industrial ResearchPretoria, South Africa
| | - Henriette van Heerden
- Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of PretoriaPretoria, South Africa
| | - Michael C. Crampton
- Protein Technologies, Biosciences, Council for Scientific and Industrial ResearchPretoria, South Africa
| |
Collapse
|
137
|
Mordkovich NN, Okorokova NA, Veiko VP. Structural and functional organization of the signal peptide of pro-enterotoxin B from Staphylococcus aureus. APPL BIOCHEM MICRO+ 2015. [DOI: 10.1134/s0003683815060101] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
138
|
Lipids assist the membrane insertion of a BAM-independent outer membrane protein. Sci Rep 2015; 5:15068. [PMID: 26463896 PMCID: PMC4604470 DOI: 10.1038/srep15068] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 09/14/2015] [Indexed: 02/04/2023] Open
Abstract
Like several other large, multimeric bacterial outer membrane proteins (OMPs), the assembly of the Klebsiella oxytoca OMP PulD does not rely on the universally conserved β-barrel assembly machinery (BAM) that catalyses outer membrane insertion. The only other factor known to interact with PulD prior to or during outer membrane targeting and assembly is the cognate chaperone PulS. Here, in vitro translation-transcription coupled PulD folding demonstrated that PulS does not act during the membrane insertion of PulD, and engineered in vivo site-specific cross-linking between PulD and PulS showed that PulS binding does not prevent membrane insertion. In vitro folding kinetics revealed that PulD is atypical compared to BAM-dependent OMPs by inserting more rapidly into membranes containing E. coli phospholipids than into membranes containing lecithin. PulD folding was fast in diC14:0-phosphatidylethanolamine liposomes but not diC14:0-phosphatidylglycerol liposomes, and in diC18:1-phosphatidylcholine liposomes but not in diC14:1-phosphatidylcholine liposomes. These results suggest that PulD efficiently exploits the membrane composition to complete final steps in insertion and explain how PulD can assemble independently of any protein-assembly machinery. Lipid-assisted assembly in this manner might apply to other large OMPs whose assembly is BAM-independent.
Collapse
|
139
|
Abstract
The major class of integral proteins found in the outer membrane (OM) of E. coli and Salmonella adopt a β-barrel conformation (OMPs). OMPs are synthesized in the cytoplasm with a typical signal sequence at the amino terminus, which directs them to the secretion machinery (SecYEG) located in the inner membrane for translocation to the periplasm. Chaperones such as SurA, or DegP and Skp, escort these proteins across the aqueous periplasm protecting them from aggregation. The chaperones then deliver OMPs to a highly conserved outer membrane assembly site termed the Bam complex. In E. coli, the Bam complex is composed of an essential OMP, BamA, and four associated OM lipoproteins, BamBCDE, one of which, BamD, is also essential. Here we provide an overview of what we know about the process of OMP assembly and outline the various hypotheses that have been proposed to explain how proteins might be integrated into the asymmetric OM lipid bilayer in an environment that lacks obvious energy sources. In addition, we describe the envelope stress responses that ensure the fidelity of OM biogenesis and how factors, such as phage and certain toxins, have coopted this essential machine to gain entry into the cell.
Collapse
|
140
|
Prabudiansyah I, Kusters I, Caforio A, Driessen AJ. Characterization of the annular lipid shell of the Sec translocon. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1848:2050-6. [DOI: 10.1016/j.bbamem.2015.06.024] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 06/24/2015] [Accepted: 06/26/2015] [Indexed: 11/16/2022]
|
141
|
Optimization of the secretion pathway for heterologous proteins in Bacillus subtilis. BIOTECHNOL BIOPROC E 2015. [DOI: 10.1007/s12257-014-0843-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
142
|
Abstract
Type I signal peptidase (SPase) is essential for viability in wild-type bacteria because the terminal step of the bacterial general secretory pathway requires its proteolytic activity to release proteins from their membrane-bound N-terminal leader sequences after translocation across the cytoplasmic membrane. Here, we identify the Staphylococcus aureus operon ayrRABC (SA0337 to SA0340) and show that once released from repression by AyrR, the protein products AyrABC together confer resistance to the SPase inhibitor arylomycin M131 by providing an alternate and novel method of releasing translocated proteins. Thus, the derepression of ayrRABC allows cells to bypass the essentiality of SPase. We demonstrate that AyrABC functionally complements SPase by mediating the processing of the normally secreted proteins, albeit in some cases with reduced efficiency and either without cleavage or via cleavage at a site N-terminal to the canonical SPase cleavage site. Thus, ayrRABC encodes a secretion stress-inducible alternate terminal step of the general secretory pathway. IMPORTANCE : Addressing proteins for proper localization within or outside a cell in both eukaryotes and prokaryotes is often accomplished with intrinsic signals which mediate membrane translocation and which ultimately must be removed. The canonical enzyme responsible for the removal of translocation signals is bacterial type I signal peptidase (SPase), which functions at the terminal step of the general secretory pathway and is thus essential in wild-type bacteria. Here, we identify a four-gene operon in S. aureus that encodes an alternate terminal step of the general secretory pathway and thus makes SPase nonessential. The results have important implications for protein secretion in bacteria and potentially for protein trafficking in prokaryotes and eukaryotes in general.
Collapse
|
143
|
Cournia Z, Allen TW, Andricioaei I, Antonny B, Baum D, Brannigan G, Buchete NV, Deckman JT, Delemotte L, del Val C, Friedman R, Gkeka P, Hege HC, Hénin J, Kasimova MA, Kolocouris A, Klein ML, Khalid S, Lemieux MJ, Lindow N, Roy M, Selent J, Tarek M, Tofoleanu F, Vanni S, Urban S, Wales DJ, Smith JC, Bondar AN. Membrane Protein Structure, Function, and Dynamics: a Perspective from Experiments and Theory. J Membr Biol 2015; 248:611-40. [PMID: 26063070 PMCID: PMC4515176 DOI: 10.1007/s00232-015-9802-0] [Citation(s) in RCA: 134] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 03/26/2015] [Indexed: 01/05/2023]
Abstract
Membrane proteins mediate processes that are fundamental for the flourishing of biological cells. Membrane-embedded transporters move ions and larger solutes across membranes; receptors mediate communication between the cell and its environment and membrane-embedded enzymes catalyze chemical reactions. Understanding these mechanisms of action requires knowledge of how the proteins couple to their fluid, hydrated lipid membrane environment. We present here current studies in computational and experimental membrane protein biophysics, and show how they address outstanding challenges in understanding the complex environmental effects on the structure, function, and dynamics of membrane proteins.
Collapse
Affiliation(s)
- Zoe Cournia
- Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephessiou, 11527, Athens, Greece
| | - Toby W. Allen
- School of Applied Sciences & Health Innovations Research Institute, RMIT University, GPO Box 2476, Melbourne, Vic, 3001, Australia; and Department of Chemistry, University of California, Davis. Davis, CA 95616, USA
| | - Ioan Andricioaei
- Department of Chemistry, University of California, Irvine, CA 92697
| | - Bruno Antonny
- Institut de Pharmacologie Moléculaire et Cellulaire, Université de Nice Sophia-Antipolis and Centre National de la Recherche Scientifique, UMR 7275, 06560 Valbonne, France
| | - Daniel Baum
- Department of Visualization and Data Analysis, Zuse Institute Berlin, Takustrasse 7, D-14195 Berlin, Germany
| | - Grace Brannigan
- Center for Computational and Integrative Biology and Department of Physics, Rutgers University-Camden, Camden, NJ, USA
| | - Nicolae-Viorel Buchete
- School of Physics and Complex and Adaptive Systems Laboratory, University College Dublin, Belfield, Dublin 4, Ireland
| | | | - Lucie Delemotte
- Institute of Computational and Molecular Science, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Coral del Val
- Department of Artificial Intelligence, University of Granada, E-18071 Granada, Spain
| | - Ran Friedman
- Linnæus University, Department of Chemistry and Biomedical Sciences & Centre for Biomaterials Chemistry, 391 82 Kalmar, Sweden
| | - Paraskevi Gkeka
- Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephessiou, 11527, Athens, Greece
| | - Hans-Christian Hege
- Department of Visualization and Data Analysis, Zuse Institute Berlin, Takustrasse 7, D-14195 Berlin, Germany
| | - Jérôme Hénin
- Laboratoire de Biochimie Théorique, IBPC and CNRS, Paris, France
| | - Marina A. Kasimova
- Université de Lorraine, SRSMC, UMR 7565, Vandoeuvre-lès-Nancy, F-54500, France
- Lomonosov Moscow State University, Moscow, 119991, Russian Federation
| | - Antonios Kolocouris
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, University of Athens, Panepistimioupolis-Zografou, 15771 Athens, Greece
| | - Michael L. Klein
- Institute of Computational and Molecular Science, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Syma Khalid
- Department of Chemistry, University of Southampton, Highfield, Southampton, SO17 1BJ, UK
| | - M. Joanne Lemieux
- Department of Biochemistry, Faculty of Medicine & Dentistry, Membrane Protein Disease Research Group, and Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada, T6G 2H7
| | - Norbert Lindow
- Department of Visualization and Data Analysis, Zuse Institute Berlin, Takustrasse 7, D-14195 Berlin, Germany
| | - Mahua Roy
- Department of Chemistry, University of California, Irvine
| | - Jana Selent
- Research Programme on Biomedical Informatics (GRIB), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, IMIM (Hospital del Mar Medical Research Institute), Dr. Aiguader 88, E-08003 Barcelona, Spain
| | - Mounir Tarek
- Université de Lorraine, SRSMC, UMR 7565, Vandoeuvre-lès-Nancy, F-54500, France
- CNRS, SRSMC, UMR 7565, Vandoeuvre-lès-Nancy, F-54500, France
| | - Florentina Tofoleanu
- School of Physics and Complex and Adaptive Systems Laboratory, University College Dublin, Belfield, Dublin 4, Ireland
| | - Stefano Vanni
- Institut de Pharmacologie Moléculaire et Cellulaire, Université de Nice Sophia-Antipolis and Centre National de la Recherche Scientifique, UMR 7275, 06560 Valbonne, France
| | - Sinisa Urban
- Johns Hopkins University School of Medicine, Howard Hughes Medical Institute, Department of Molecular Biology & Genetics, 725 N. Wolfe Street, 507 Preclinical Teaching Building, Baltimore, MD 21205, USA
| | - David J. Wales
- University Chemical Laboratories, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K
| | - Jeremy C. Smith
- Oak Ridge National Laboratory, PO BOX 2008 MS6309, Oak Ridge, TN 37831-6309, USA
| | - Ana-Nicoleta Bondar
- Theoretical Molecular Biophysics, Department of Physics, Freie Universität Berlin, Arnimallee 14, D-14195 Berlin, Germany
| |
Collapse
|
144
|
Dong Z, Zhang J, Du G, Chen J, Li H, Lee B. Periplasmic Export of Bile Salt Hydrolase in Escherichia coli by the Twin-Arginine Signal Peptides. Appl Biochem Biotechnol 2015. [PMID: 26198023 DOI: 10.1007/s12010-015-1755-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Bile salt hydrolase (BSH, EC 3.5.1.24) is considered as an ideal way with lower cost and less side effects to release the risk of coronary heart disease caused by hypercholesterolemia. As bile salt hydrolase from Lactobacillus plantarum BBE7 could not be efficiently exported by PelB signal peptide of the general secretory (Sec) pathway, three twin-arginine signal peptides from twin-arginine translocation (Tat) pathway were synthesized, fused with bsh gene, inserted into expression vectors pET-20b(+) and pET-22b(+), and transformed into four different Escherichia coli hosts, respectively. Among the 24 recombinant bacteria obtained, E. coli BL21 (DE3) pLysS (pET-20b(+)-dmsA-bsh) showed the highest BSH activity in periplasmic fraction, which was further increased to 1.21 ± 0.03 U/mL by orthogonal experimental design. And, signal peptide dimethyl sulfoxide reductase subunit DmsA (DMSA) had the best activity of exported BSH. More importantly, the presence of BSH in the periplasm had proven to be caused by the export rather than cell leakage. For the first time, we report the periplasmic expression of BSH by signal peptides from the Tat pathway. This will lay a solid foundation for the purification and biochemical characterization of BSH from the supernatant, and strategies adopted here could be used for the periplasmic expression of other proteins in E. coli.
Collapse
Affiliation(s)
- Zixing Dong
- College of Chemical Engineering and Material Science, Tianjin University of Science & Technology, Tianjin, 300457, China
| | | | | | | | | | | |
Collapse
|
145
|
Glas M, van den Berg van Saparoea HB, McLaughlin SH, Roseboom W, Liu F, Koningstein GM, Fish A, den Blaauwen T, Heck AJR, de Jong L, Bitter W, de Esch IJP, Luirink J. The Soluble Periplasmic Domains of Escherichia coli Cell Division Proteins FtsQ/FtsB/FtsL Form a Trimeric Complex with Submicromolar Affinity. J Biol Chem 2015; 290:21498-509. [PMID: 26160297 DOI: 10.1074/jbc.m115.654756] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Indexed: 01/10/2023] Open
Abstract
Cell division in Escherichia coli involves a set of essential proteins that assembles at midcell to form the so-called divisome. The divisome regulates the invagination of the inner membrane, cell wall synthesis, and inward growth of the outer membrane. One of the divisome proteins, FtsQ, plays a central but enigmatic role in cell division. This protein associates with FtsB and FtsL, which, like FtsQ, are bitopic inner membrane proteins with a large periplasmic domain (denoted FtsQp, FtsBp, and FtsLp) that is indispensable for the function of each protein. Considering the vital nature and accessible location of the FtsQBL complex, it is an attractive target for protein-protein interaction inhibitors intended to block bacterial cell division. In this study, we expressed FtsQp, FtsBp, and FtsLp individually and in combination. Upon co-expression, FtsQp was co-purified with FtsBp and FtsLp from E. coli extracts as a stable trimeric complex. FtsBp was also shown to interact with FtsQp in the absence of FtsLp albeit with lower affinity. Interactions were mapped at the C terminus of the respective domains by site-specific cross-linking. The binding affinity and 1:1:1 stoichiometry of the FtsQpBpLp complex and the FtsQpBp subcomplex were determined in complementary surface plasmon resonance, analytical ultracentrifugation, and native mass spectrometry experiments.
Collapse
Affiliation(s)
- Marjolein Glas
- From the Amsterdam Institute of Molecules, Medicines and Systems, VU University Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | - H Bart van den Berg van Saparoea
- From the Amsterdam Institute of Molecules, Medicines and Systems, VU University Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | - Stephen H McLaughlin
- the Medical Research Council Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, United Kingdom
| | - Winfried Roseboom
- the Swammerdam Institute for Life Sciences, Department of Mass Spectrometry of Biomacromolecules, and
| | - Fan Liu
- the Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CH Utrecht, The Netherlands, and
| | - Gregory M Koningstein
- From the Amsterdam Institute of Molecules, Medicines and Systems, VU University Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | - Alexander Fish
- the NKI Protein Facility, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Tanneke den Blaauwen
- Swammerdam Institute for Life Sciences, Department of Bacterial Cell Biology, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Albert J R Heck
- the Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CH Utrecht, The Netherlands, and
| | - Luitzen de Jong
- the Swammerdam Institute for Life Sciences, Department of Mass Spectrometry of Biomacromolecules, and
| | - Wilbert Bitter
- From the Amsterdam Institute of Molecules, Medicines and Systems, VU University Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | - Iwan J P de Esch
- From the Amsterdam Institute of Molecules, Medicines and Systems, VU University Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | - Joen Luirink
- From the Amsterdam Institute of Molecules, Medicines and Systems, VU University Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands,
| |
Collapse
|
146
|
Excretion of cytoplasmic proteins in Staphylococcus is most likely not due to cell lysis. Curr Genet 2015; 62:19-23. [DOI: 10.1007/s00294-015-0504-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 06/24/2015] [Indexed: 01/02/2023]
|
147
|
Wendler S, Otto A, Ortseifen V, Bonn F, Neshat A, Schneiker-Bekel S, Walter F, Wolf T, Zemke T, Wehmeier UF, Hecker M, Kalinowski J, Becher D, Pühler A. Comprehensive proteome analysis of Actinoplanes sp. SE50/110 highlighting the location of proteins encoded by the acarbose and the pyochelin biosynthesis gene cluster. J Proteomics 2015; 125:1-16. [DOI: 10.1016/j.jprot.2015.04.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 04/02/2015] [Accepted: 04/12/2015] [Indexed: 01/05/2023]
|
148
|
Ebner P, Prax M, Nega M, Koch I, Dube L, Yu W, Rinker J, Popella P, Flötenmeyer M, Götz F. Excretion of cytoplasmic proteins (ECP) inStaphylococcus aureus. Mol Microbiol 2015; 97:775-89. [DOI: 10.1111/mmi.13065] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/19/2015] [Indexed: 12/15/2022]
Affiliation(s)
- Patrick Ebner
- Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine (IMIT); University of Tübingen; Auf der Morgenstelle 28 72076 Tübingen Germany
| | - Marcel Prax
- Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine (IMIT); University of Tübingen; Auf der Morgenstelle 28 72076 Tübingen Germany
| | - Mulugeta Nega
- Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine (IMIT); University of Tübingen; Auf der Morgenstelle 28 72076 Tübingen Germany
| | - Iris Koch
- Max Planck Institute for Developmental Biology; Spemannstr. 35 72076 Tübingen Germany
| | - Linda Dube
- Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine (IMIT); University of Tübingen; Auf der Morgenstelle 28 72076 Tübingen Germany
| | - Wenqi Yu
- Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine (IMIT); University of Tübingen; Auf der Morgenstelle 28 72076 Tübingen Germany
| | - Janina Rinker
- Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine (IMIT); University of Tübingen; Auf der Morgenstelle 28 72076 Tübingen Germany
| | - Peter Popella
- Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine (IMIT); University of Tübingen; Auf der Morgenstelle 28 72076 Tübingen Germany
| | - Matthias Flötenmeyer
- Max Planck Institute for Developmental Biology; Spemannstr. 35 72076 Tübingen Germany
| | - Friedrich Götz
- Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine (IMIT); University of Tübingen; Auf der Morgenstelle 28 72076 Tübingen Germany
| |
Collapse
|
149
|
In Vitro Interaction of the Housekeeping SecA1 with the Accessory SecA2 Protein of Mycobacterium tuberculosis. PLoS One 2015; 10:e0128788. [PMID: 26047312 PMCID: PMC4457860 DOI: 10.1371/journal.pone.0128788] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 04/30/2015] [Indexed: 12/22/2022] Open
Abstract
The majority of proteins that are secreted across the bacterial cytoplasmic membrane leave the cell via the Sec pathway, which in its minimal form consists of the dimeric ATP-driven motor protein SecA that associates with the protein-conducting membrane pore SecYEG. Some Gram-positive bacteria contain two homologues of SecA, termed SecA1 and SecA2. SecA1 is the essential housekeeping protein, whereas SecA2 is not essential but is involved in the translocation of a subset of proteins, including various virulence factors. Some SecA2 containing bacteria also harbor a homologous SecY2 protein that may form a separate translocase. Interestingly, mycobacteria contain only one SecY protein and thus both SecA1 and SecA2 are required to interact with SecYEG, either individually or together as a heterodimer. In order to address whether SecA1 and SecA2 cooperate during secretion of SecA2 dependent proteins, we examined the oligomeric state of SecA1 and SecA2 of Mycobacterium tuberculosis and their interactions with SecA2 and the cognate SecA1, respectively. We conclude that both SecA1 and SecA2 individually form homodimers in solution but when both proteins are present simultaneously, they form dissociable heterodimers.
Collapse
|
150
|
Rana A, Kumar D, Rub A, Akhter Y. Proteome-scale identification and characterization of mitochondria targeting proteins of Mycobacterium avium subspecies paratuberculosis: Potential virulence factors modulating host mitochondrial function. Mitochondrion 2015; 23:42-54. [PMID: 26048556 DOI: 10.1016/j.mito.2015.05.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 04/21/2015] [Accepted: 05/04/2015] [Indexed: 02/03/2023]
Abstract
Mycobacterium avium subsp. paratuberculosis is the etiological agent of Johne's Disease among ruminants. During the course of infection, it expresses a number of proteins for its successful persistence inside the host that cause variety of physiological abnormalities in the host. Mitochondrion is one of the attractive targets for pathogenic bacteria. Employing a proteome-wide sequence and structural signature based approach we have identified 46 M. avium subsp. paratuberculosis proteins as potential targets for the host mitochondrial targeting. These may act as virulence factors modulating mitochondrial physiology for bacterial survival and immune evasion inside the host cells.
Collapse
Affiliation(s)
- Aarti Rana
- School of Life Sciences, Central University of Himachal Pradesh, Shahpur, District-Kangra, 176206 Himachal Pradesh, India
| | - Devender Kumar
- School of Life Sciences, Central University of Himachal Pradesh, Shahpur, District-Kangra, 176206 Himachal Pradesh, India
| | - Abdur Rub
- Infection and Immunity Lab, Department of Biotechnology, Jamia Millia Islamia (A Central University), New Delhi 110025, India
| | - Yusuf Akhter
- School of Life Sciences, Central University of Himachal Pradesh, Shahpur, District-Kangra, 176206 Himachal Pradesh, India.
| |
Collapse
|