Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 289
Filter
1.
Protein Sci ; 33(7): e5025, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38864689

ABSTRACT

Polyhydroxyalkanoates are a class of biodegradable, thermoplastic polymers which represent a major carbon source for various bacteria. Proteins which mediate the translocation of polyhydroxyalkanoate breakdown products, such as ß-hydroxybutyrate (BHB)-a ketone body which in humans serves as an important biomarker, have not been well characterized. In our investigation to screen a solute-binding protein (SBP) which can act as a suitable recognition element for BHB, we uncovered insights at the intersection of bacterial metabolism and diagnostics. Herein, we identify SBPs associated with putative ATP-binding cassette transporters that specifically recognize BHB, with the potential to serve as recognition elements for continuous quantification of this analyte. Through bioinformatic analysis, we identified candidate SBPs from known metabolizers of polyhydroxybutyrate-including proteins from Cupriavidus necator, Ensifer meliloti, Paucimonas lemoignei, and Thermus thermophilus. After recombinant expression in Escherichia coli, we demonstrated with intrinsic tryptophan fluorescence spectroscopy that four candidate proteins interacted with BHB, ranging from nanomolar to micromolar affinity. Tt.2, an intrinsically thermostable protein from Thermus thermophilus, was observed to have the tightest binding and specificity for BHB, which was confirmed by isothermal calorimetry. Structural analyses facilitated by AlphaFold2, along with molecular docking and dynamics simulations, were used to hypothesize key residues in the binding pocket and to model the conformational dynamics of substrate unbinding. Overall, this study provides strong evidence identifying the cognate ligands of SBPs which we hypothesize to be involved in prokaryotic cellular translocation of polyhydroxyalkanoate breakdown products, while highlighting these proteins' promising biotechnological application.


Subject(s)
3-Hydroxybutyric Acid , 3-Hydroxybutyric Acid/metabolism , 3-Hydroxybutyric Acid/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Periplasmic Binding Proteins/metabolism , Periplasmic Binding Proteins/chemistry , Periplasmic Binding Proteins/genetics , Escherichia coli/metabolism , Escherichia coli/genetics , Ketone Bodies/metabolism , Ketone Bodies/chemistry
2.
Proc Natl Acad Sci U S A ; 121(25): e2319903121, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38870058

ABSTRACT

Biofilm formation and surface attachment in multiple Alphaproteobacteria is driven by unipolar polysaccharide (UPP) adhesins. The pathogen Agrobacterium tumefaciens produces a UPP adhesin, which is regulated by the intracellular second messenger cyclic diguanylate monophosphate (c-di-GMP). Prior studies revealed that DcpA, a diguanylate cyclase-phosphodiesterase, is crucial in control of UPP production and surface attachment. DcpA is regulated by PruR, a protein with distant similarity to enzymatic domains known to coordinate the molybdopterin cofactor (MoCo). Pterins are bicyclic nitrogen-rich compounds, several of which are produced via a nonessential branch of the folate biosynthesis pathway, distinct from MoCo. The pterin-binding protein PruR controls DcpA activity, fostering c-di-GMP breakdown and dampening its synthesis. Pterins are excreted, and we report here that PruR associates with these metabolites in the periplasm, promoting interaction with the DcpA periplasmic domain. The pteridine reductase PruA, which reduces specific dihydro-pterin molecules to their tetrahydro forms, imparts control over DcpA activity through PruR. Tetrahydromonapterin preferentially associates with PruR relative to other related pterins, and the PruR-DcpA interaction is decreased in a pruA mutant. PruR and DcpA are encoded in an operon with wide conservation among diverse Proteobacteria including mammalian pathogens. Crystal structures reveal that PruR and several orthologs adopt a conserved fold, with a pterin-specific binding cleft that coordinates the bicyclic pterin ring. These findings define a pterin-responsive regulatory mechanism that controls biofilm formation and related c-di-GMP-dependent phenotypes in A. tumefaciens and potentially acts more widely in multiple proteobacterial lineages.


Subject(s)
Agrobacterium tumefaciens , Bacterial Proteins , Biofilms , Cyclic GMP , Pterins , Biofilms/growth & development , Agrobacterium tumefaciens/metabolism , Agrobacterium tumefaciens/genetics , Pterins/metabolism , Cyclic GMP/metabolism , Cyclic GMP/analogs & derivatives , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Proteobacteria/metabolism , Proteobacteria/genetics , Molybdenum Cofactors , Periplasm/metabolism , Periplasmic Proteins/metabolism , Periplasmic Proteins/genetics , Periplasmic Binding Proteins/metabolism , Periplasmic Binding Proteins/genetics , Gene Expression Regulation, Bacterial
3.
Biochemistry ; 63(10): 1322-1334, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38696389

ABSTRACT

Periplasmic solute-binding proteins (SBPs) are key ligand recognition components of bacterial ATP-binding cassette (ABC) transporters that allow bacteria to import nutrients and metabolic precursors from the environment. Periplasmic SBPs comprise a large and diverse family of proteins, of which only a small number have been empirically characterized. In this work, we identify a set of 610 unique uncharacterized proteins within the SBP_bac_5 family that are found in conserved operons comprising genes encoding (i) ABC transport systems and (ii) putative amidases from the FmdA_AmdA family. From these uncharacterized SBP_bac_5 proteins, we characterize a representative periplasmic SBP from Mesorhizobium sp. A09 (MeAmi_SBP) and show that MeAmi_SBP binds l-amino acid amides but not the corresponding l-amino acids. An X-ray crystal structure of MeAmi_SBP bound to l-serinamide highlights the residues that impart distinct specificity for l-amino acid amides and reveals a structural Ca2+ binding site within one of the lobes of the protein. We show that the residues involved in ligand and Ca2+ binding are conserved among the 610 SBPs from experimentally uncharacterized FmdA_AmdA amidase-associated ABC transporter systems, suggesting these homologous systems are also likely to be involved in the sensing, uptake, and metabolism of l-amino acid amides across many Gram-negative nitrogen-fixing soil bacteria. We propose that MeAmi_SBP is involved in the uptake of such solutes to supplement pathways such as the citric acid cycle and the glutamine synthetase-glutamate synthase pathway. This work expands our currently limited understanding of microbial interactions with l-amino acid amides and bacterial nitrogen utilization.


Subject(s)
Amides , Periplasmic Binding Proteins , Amides/metabolism , Amides/chemistry , Crystallography, X-Ray , Periplasmic Binding Proteins/metabolism , Periplasmic Binding Proteins/chemistry , Periplasmic Binding Proteins/genetics , ATP-Binding Cassette Transporters/metabolism , ATP-Binding Cassette Transporters/chemistry , Amino Acids/metabolism , Mesorhizobium/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Binding Sites , Models, Molecular , Amidohydrolases/metabolism , Amidohydrolases/chemistry , Calcium/metabolism , Protein Binding
4.
FEBS Lett ; 598(11): 1375-1386, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38508768

ABSTRACT

Modular assembly is a compelling pathway to create new proteins, a concept supported by protein engineering and millennia of evolution. Natural evolution provided a repository of building blocks, known as domains, which trace back to even shorter segments that underwent numerous 'copy-paste' processes culminating in the scaffolds we see today. Utilizing the subdomain-database Fuzzle, we constructed a fold-chimera by integrating a flavodoxin-like fragment into a periplasmic binding protein. This chimera is well-folded and a crystal structure reveals stable interfaces between the fragments. These findings demonstrate the adaptability of α/ß-proteins and offer a stepping stone for optimization. By emphasizing the practicality of fragment databases, our work pioneers new pathways in protein engineering. Ultimately, the results substantiate the conjecture that periplasmic binding proteins originated from a flavodoxin-like ancestor.


Subject(s)
Protein Engineering , Protein Folding , Protein Engineering/methods , Models, Molecular , Flavodoxin/chemistry , Flavodoxin/metabolism , Flavodoxin/genetics , Periplasmic Binding Proteins/metabolism , Periplasmic Binding Proteins/chemistry , Periplasmic Binding Proteins/genetics , Crystallography, X-Ray , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/genetics , Protein Domains
5.
mBio ; 15(2): e0303923, 2024 Feb 14.
Article in English | MEDLINE | ID: mdl-38193657

ABSTRACT

The outer membrane (OM) is an essential organelle of Gram-negative bacteria. Lipoproteins are key to building the OM, performing essential functions in several OM assembly machines. Lipoproteins mature in the inner membrane (IM) and are then trafficked to the OM. In Escherichia coli, the LolCDE transporter is needed to extract lipoproteins from the IM to begin trafficking. Lipoproteins are then transferred from LolCDE to the periplasmic chaperone LolA which ferries them to the OM for insertion by LolB. LolA recruitment by LolC is an essential trafficking step. Structural and biochemical studies suggested that two regions (termed Hook and Pad) within a periplasmic loop of LolC worked in tandem to recruit LolA, leading to a bipartite model for recruitment. Here, we genetically examine the LolC periplasmic loop in vivo using E. coli. Our findings challenge the bipartite interaction model. We show that while the Hook is essential for lipoprotein trafficking in vivo, lipoproteins are still efficiently trafficked when the Pad residues are inactivated. We show with AlphaFold2 multimer modeling that Hook:LolA interactions are likely universal among diverse Gram-negative bacteria. Conversely, Pad:LolA interactions vary across phyla. Our in vivo data redefine LolC:LolA recruitment into a hierarchical interaction model. We propose that the Hook is the major player in LolA recruitment, while the Pad plays an ancillary role that is important for efficiency but is ultimately dispensable. Our findings expand the understanding of a fundamental step in essential lipoprotein trafficking and have implications for efforts to develop new antibacterials that target LolCDE.IMPORTANCEResistance to current antibiotics is increasingly common. New antibiotics that target essential processes are needed to expand clinical options. For Gram-negative bacteria, their cell surface-the outer membrane (OM)-is an essential organelle and antibiotic barrier that is an attractive target for new antibacterials. Lipoproteins are key to building the OM. The LolCDE transporter is needed to supply the OM with lipoproteins and has been a focus of recent antibiotic discovery. In vitro evidence recently proposed a two-part interaction of LolC with LolA lipoprotein chaperone (which traffics lipoproteins to the OM) via "Hook" and "Pad" regions. We show that this model does not reflect lipoprotein trafficking in vivo. Only the Hook is essential for lipoprotein trafficking and is remarkably robust to mutational changes. The Pad is non-essential for lipoprotein trafficking but plays an ancillary role, contributing to trafficking efficiency. These insights inform ongoing efforts to drug LolCDE.


Subject(s)
Escherichia coli Proteins , Periplasmic Binding Proteins , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Lipoproteins/genetics , Lipoproteins/metabolism , Membrane Transport Proteins/metabolism , Gram-Negative Bacteria/metabolism , Anti-Bacterial Agents/metabolism , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Periplasmic Binding Proteins/genetics , Periplasmic Binding Proteins/metabolism
6.
Proc Natl Acad Sci U S A ; 119(36): e2208662119, 2022 09 06.
Article in English | MEDLINE | ID: mdl-36037338

ABSTRACT

In gram-negative bacteria, lipoproteins are vital structural components of the outer membrane (OM) and crucial elements of machineries central to the physiology of the cell envelope. A dedicated apparatus, the Lol system, is required for the correct localization of OM lipoproteins and is essential for viability. The periplasmic chaperone LolA is central to this trafficking pathway, accepting triacylated lipoproteins from the inner membrane transporter LolCDE, before carrying them across the periplasm to the OM receptor LolB. Here, we report a crystal structure of liganded LolA, generated in vivo, revealing the molecular details of lipoprotein association. The structure highlights how LolA, initially primed to receive lipoprotein by interaction with LolC, further opens to accommodate the three ligand acyl chains in a precise conformation within its cavity. LolA forms extensive interactions with the acyl chains but not with any residue of the cargo, explaining the chaperone's ability to transport structurally diverse lipoproteins. Structural characterization of a ligandedLolA variant incapable of lipoprotein release reveals aberrant association, demonstrating the importance of the LolCDE-coordinated, sequential opening of LolA for inserting lipoprotein in a manner productive for subsequent trafficking. Comparison with existing structures of LolA in complex with LolC or LolCDE reveals substantial overlap of the lipoprotein and LolC binding sites within the LolA cavity, demonstrating that insertion of lipoprotein acyl chains physically disengages the chaperone protein from the transporter by perturbing interaction with LolC. Taken together, our data provide a key step toward a complete understanding of a fundamentally important trafficking pathway.


Subject(s)
Escherichia coli Proteins , Periplasmic Binding Proteins , Protein Transport , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Carrier Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Ligands , Lipoproteins/metabolism , Models, Molecular , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Periplasm/metabolism , Periplasmic Binding Proteins/chemistry , Periplasmic Binding Proteins/genetics , Periplasmic Binding Proteins/metabolism , Protein Structure, Tertiary , Protein Transport/genetics
7.
FEBS J ; 289(2): 436-456, 2022 01.
Article in English | MEDLINE | ID: mdl-34375507

ABSTRACT

The organic polymer lignin is a component of plant cell walls, which like (hemi)-cellulose is highly abundant in nature and relatively resistant to degradation. However, extracellular enzymes released by natural microbial consortia can cleave the ß-aryl ether linkages in lignin, releasing monoaromatic phenylpropanoids that can be further catabolised by diverse species of bacteria. Biodegradation of lignin is therefore important in global carbon cycling, and its natural abundance also makes it an attractive biotechnological feedstock for the industrial production of commodity chemicals. Whilst the pathways for degradation of lignin-derived aromatics have been extensively characterised, much less is understood about how they are recognised and taken up from the environment. The purple phototrophic bacterium Rhodopseudomonas palustris can grow on a range of phenylpropanoid monomers and is a model organism for studying their uptake and breakdown. R. palustris encodes a tripartite ATP-independent periplasmic (TRAP) transporter (TarPQM) linked to genes encoding phenylpropanoid-degrading enzymes. The periplasmic solute-binding protein component of this transporter, TarP, has previously been shown to bind aromatic substrates. Here, we determine the high-resolution crystal structure of TarP from R. palustris as well as the structures of homologous proteins from the salt marsh bacterium Sagittula stellata and the halophile Chromohalobacter salexigens, which also grow on lignin-derived aromatics. In combination with tryptophan fluorescence ligand-binding assays, our ligand-bound co-crystal structures reveal the molecular basis for high-affinity recognition of phenylpropanoids by these TRAP transporters, which have potential for improving uptake of these compounds for biotechnological transformations of lignin.


Subject(s)
Bacterial Proteins/genetics , Biodegradation, Environmental , Lignin/genetics , RNA-Binding Proteins/genetics , Rhodopseudomonas/genetics , Transcription Factors/genetics , Biological Transport/genetics , Gene Expression Regulation, Bacterial/genetics , Ligands , Lignin/chemistry , Lignin/metabolism , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Oxidoreductases/genetics , Periplasm/genetics , Periplasm/microbiology , Periplasmic Binding Proteins/genetics , Proteobacteria/genetics , Proteobacteria/growth & development , Rhodopseudomonas/growth & development
8.
Appl Environ Microbiol ; 88(2): e0211721, 2022 01 25.
Article in English | MEDLINE | ID: mdl-34757821

ABSTRACT

Periplasmic binding proteins have been previously proclaimed as a general scaffold to design sensor proteins with new recognition specificities for nonnatural compounds. Such proteins can be integrated in bacterial bioreporter chassis with hybrid chemoreceptors to produce a concentration-dependent signal after ligand binding to the sensor cell. However, computationally designed new ligand-binding properties ignore the more general properties of periplasmic binding proteins, such as their periplasmic translocation, dynamic transition of open and closed forms, and interactions with membrane receptors. In order to better understand the roles of such general properties in periplasmic signaling behavior, we studied the subcellular localization of ribose-binding protein (RbsB) in Escherichia coli in comparison to a recently evolved set of mutants designed to bind 1,3-cyclohexanediol. As proxies for localization, we calibrated and deployed C-terminal end mCherry fluorescent protein fusions. Whereas RbsB-mCherry coherently localized to the periplasmic space and accumulated in (periplasmic) polar regions depending on chemoreceptor availability, mutant RbsB-mCherry expression resulted in high fluorescence cell-to-cell variability. This resulted in higher proportions of cells devoid of clear polar foci and of cells with multiple fluorescent foci elsewhere, suggesting poorer translocation, periplasmic autoaggregation, and mislocalization. Analysis of RbsB mutants and mutant libraries at different stages of directed evolution suggested overall improvement to more RbsB-wild-type-like characteristics, which was corroborated by structure predictions. Our results show that defects in periplasmic localization of mutant RbsB proteins partly explain their poor sensing performance. Future efforts should be directed to predicting or selecting secondary mutations outside computationally designed binding pockets, taking folding, translocation, and receptor interactions into account. IMPORTANCE Biosensor engineering relies on transcription factors or signaling proteins to provide the actual sensory functions for the target chemicals. Since for many compounds there are no natural sensory proteins, there is a general interest in methods that could unlock routes to obtaining new ligand-binding properties. Bacterial periplasmic binding proteins (PBPs) form an interesting family of proteins to explore for this purpose, because there is a large natural variety suggesting evolutionary trajectories to bind new ligands. PBPs are conserved and amenable to accurate computational binding pocket predictions. However, studying ribose-binding protein in Escherichia coli, we discovered that designed variants have defects in their proper localization in the cell, which can impair appropriate sensor signaling. This indicates that functional sensing capacity of PBPs cannot be obtained solely through computational design of the ligand-binding pocket but must take other properties of the protein into account, which are currently very difficult to predict.


Subject(s)
Escherichia coli Proteins , Periplasmic Binding Proteins , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Ligands , Mutant Proteins/metabolism , Periplasmic Binding Proteins/genetics , Periplasmic Binding Proteins/metabolism , Ribose/metabolism
9.
J Biol Chem ; 298(1): 101445, 2022 01.
Article in English | MEDLINE | ID: mdl-34822841

ABSTRACT

The Escherichia coli yobA-yebZ-yebY (AZY) operon encodes the proteins YobA, YebZ, and YebY. YobA and YebZ are homologs of the CopC periplasmic copper-binding protein and the CopD putative copper importer, respectively, whereas YebY belongs to the uncharacterized Domain of Unknown Function 2511 family. Despite numerous studies of E. coli copper homeostasis and the existence of the AZY operon in a range of bacteria, the operon's proteins and their functional roles have not been explored. In this study, we present the first biochemical and functional studies of the AZY proteins. Biochemical characterization and structural modeling indicate that YobA binds a single Cu2+ ion with high affinity. Bioinformatics analysis shows that YebY is widespread and encoded either in AZY operons or in other genetic contexts unrelated to copper homeostasis. We also determined the 1.8 Å resolution crystal structure of E. coli YebY, which closely resembles that of the lantibiotic self-resistance protein MlbQ. Two strictly conserved cysteine residues form a disulfide bond, consistent with the observed periplasmic localization of YebY. Upon treatment with reductants, YebY binds Cu+ and Cu2+ with low affinity, as demonstrated by metal-binding analysis and tryptophan fluorescence. Finally, genetic manipulations show that the AZY operon is not involved in copper tolerance or antioxidant defense. Instead, YebY and YobA are required for the activity of the copper-related NADH dehydrogenase II. These results are consistent with a potential role of the AZY operon in copper delivery to membrane proteins.


Subject(s)
Copper , Escherichia coli Proteins , Escherichia coli , Operon , Periplasmic Binding Proteins , Chelating Agents/metabolism , Copper/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Periplasmic Binding Proteins/genetics , Periplasmic Binding Proteins/metabolism , Structure-Activity Relationship
10.
Commun Biol ; 4(1): 1383, 2021 12 09.
Article in English | MEDLINE | ID: mdl-34887516

ABSTRACT

As one of the most elegant biological processes developed in bacteria, the siderophore-mediated iron uptake demands the action of specific ATP-binding cassette (ABC) importers. Although extensive studies have been done on various ABC importers, the molecular basis of these iron-chelated-siderophore importers are still not fully understood. Here, we report the structure of a ferrichrome importer FhuCDB from Escherichia coli at 3.4 Å resolution determined by cryo electron microscopy. The structure revealed a monomeric membrane subunit of FhuB with a substrate translocation pathway in the middle. In the pathway, there were unique arrangements of residues, especially layers of methionines. Important residues found in the structure were interrogated by mutagenesis and functional studies. Surprisingly, the importer's ATPase activity was decreased upon FhuD binding, which deviated from the current understanding about bacterial ABC importers. In summary, to the best of our knowledge, these studies not only reveal a new structural twist in the type II ABC importer subfamily, but also provide biological insights in the transport of iron-chelated siderophores.


Subject(s)
ATP-Binding Cassette Transporters/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli/chemistry , Ferrichrome/metabolism , Membrane Transport Proteins/chemistry , Periplasmic Binding Proteins/chemistry , ATP-Binding Cassette Transporters/genetics , Biological Transport , Cryoelectron Microscopy , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Membrane Transport Proteins/genetics , Periplasmic Binding Proteins/genetics , Siderophores/metabolism
11.
J Biol Chem ; 297(6): 101419, 2021 12.
Article in English | MEDLINE | ID: mdl-34801550

ABSTRACT

A profound understanding of the molecular interactions between receptors and ligands is important throughout diverse research, such as protein design, drug discovery, or neuroscience. What determines specificity and how do proteins discriminate against similar ligands? In this study, we analyzed factors that determine binding in two homologs belonging to the well-known superfamily of periplasmic binding proteins, PotF and PotD. Building on a previously designed construct, modes of polyamine binding were swapped. This change of specificity was approached by analyzing local differences in the binding pocket as well as overall conformational changes in the protein. Throughout the study, protein variants were generated and characterized structurally and thermodynamically, leading to a specificity swap and improvement in affinity. This dataset not only enriches our knowledge applicable to rational protein design but also our results can further lay groundwork for engineering of specific biosensors as well as help to explain the adaptability of pathogenic bacteria.


Subject(s)
Escherichia coli K12/chemistry , Escherichia coli Proteins/chemistry , Periplasmic Binding Proteins/chemistry , Receptors, Biogenic Amine/chemistry , Spermidine/chemistry , Escherichia coli K12/genetics , Escherichia coli K12/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Periplasmic Binding Proteins/genetics , Periplasmic Binding Proteins/metabolism , Protein Binding , Receptors, Biogenic Amine/genetics , Receptors, Biogenic Amine/metabolism , Spermidine/metabolism
12.
Microb Cell Fact ; 20(1): 207, 2021 Oct 30.
Article in English | MEDLINE | ID: mdl-34717624

ABSTRACT

BACKGROUND: Several Rhodobacter sphaeroides have been widely applied in commercial CoQ10 production, but they have poor glucose use. Strategies for enhancing glucose use have been widely exploited in R. sphaeroides. Nevertheless, little research has focused on the role of glucose transmembrane in the improvement of production. RESULTS: There are two potential glucose transmembrane pathways in R. sphaeroides ATCC 17023: the fructose specific-phosphotransferase system (PTSFru, fruAB) and non-PTS that relied on glucokinase (glk). fruAB mutation revealed two effects on bacterial growth: inhibition at the early cultivation phase (12-24 h) and promotion since 36 h. Glucose metabolism showed a corresponding change in characteristic vs. the growth. For ΔfruAΔfruB, maximum biomass (Biomax) was increased by 44.39% and the CoQ10 content was 27.08% more than that of the WT. glk mutation caused a significant decrease in growth and glucose metabolism. Over-expressing a galactose:H+ symporter (galP) in the ΔfruAΔfruB relieved the inhibition and enhanced the growth further. Finally, a mutant with rapid growth and high CoQ10 titer was constructed (ΔfruAΔfruB/tac::galPOP) using several glucose metabolism modifications and was verified by fermentation in 1 L fermenters. CONCLUSIONS: The PTSFru mutation revealed two effects on bacterial growth: inhibition at the early cultivation phase and promotion later. Additionally, biomass yield to glucose (Yb/glc) and CoQ10 synthesis can be promoted using fruAB mutation, and glk plays a key role in glucose metabolism. Strengthening glucose transmembrane via non-PTS improves the productivity of CoQ10 fermentation.


Subject(s)
Bacterial Proteins/metabolism , Glucose/metabolism , Metabolic Engineering , Rhodobacter sphaeroides/metabolism , Ubiquinone/analogs & derivatives , Bacterial Proteins/genetics , Biological Transport , Biomass , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Escherichia coli Proteins/genetics , Fermentation , Glucokinase/genetics , Glucokinase/metabolism , Industrial Microbiology , Monosaccharide Transport Proteins/genetics , Monosaccharide Transport Proteins/metabolism , Mutation , Periplasmic Binding Proteins/genetics , Periplasmic Binding Proteins/metabolism , Protein Kinases/genetics , Rhodobacter sphaeroides/genetics , Ubiquinone/biosynthesis
13.
J Biol Chem ; 297(3): 101046, 2021 09.
Article in English | MEDLINE | ID: mdl-34358566

ABSTRACT

Bacteria require high-efficiency uptake systems to survive and proliferate in nutrient-limiting environments, such as those found in host organisms. ABC transporters in the bacterial plasma membrane provide a mechanism for transport of many substrates. In this study, we examine an operon containing a periplasmic binding protein in Actinobacillus for its potential role in nutrient acquisition. The electron density map of 1.76 Å resolution obtained from the crystal structure of the periplasmic binding protein was best fit with a molecular model containing a pyridoxal-5'-phosphate (P5P/pyridoxal phosphate/the active form of vitamin B6) ligand within the protein's binding site. The identity of the P5P bound to this periplasmic binding protein was verified by isothermal titration calorimetry, microscale thermophoresis, and mass spectrometry, leading us to name the protein P5PA and the operon P5PAB. To illustrate the functional utility of this uptake system, we introduced the P5PAB operon from Actinobacillus pleuropneumoniae into an Escherichia coli K-12 strain that was devoid of a key enzyme required for P5P synthesis. The growth of this strain at low levels of P5P supports the functional role of this operon in P5P uptake. This is the first report of a dedicated P5P bacterial uptake system, but through bioinformatics, we discovered homologs mainly within pathogenic representatives of the Pasteurellaceae family, suggesting that this operon exists more widely outside the Actinobacillus genus.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Actinobacillus pleuropneumoniae/metabolism , Bacterial Proteins/metabolism , Vitamin B 6/metabolism , ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/genetics , Actinobacillus pleuropneumoniae/chemistry , Actinobacillus pleuropneumoniae/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Binding Sites , Biological Transport , Escherichia coli/genetics , Escherichia coli/metabolism , Models, Molecular , Operon , Periplasmic Binding Proteins/chemistry , Periplasmic Binding Proteins/genetics , Periplasmic Binding Proteins/metabolism , Pyridoxal Phosphate/chemistry , Pyridoxal Phosphate/metabolism , Vitamin B 6/chemistry
14.
Int J Biol Macromol ; 187: 350-360, 2021 Sep 30.
Article in English | MEDLINE | ID: mdl-34303738

ABSTRACT

The periplasmic binding protein (PBP) BtuF plays a key role in transporting vitamin B12 from periplasm to the ATP-binding cassette (ABC) transporter BtuCD. Conformational changes of BtuF during transport can hardly be captured by traditional biophysical methods and the exact mechanism regarding B12 and BtuF recognition is still under debate. In the present work, conformational changes of BtuF upon B12 binding and release were investigated using hybrid approaches including collision-induced unfolding (CIU), hydrogen deuterium exchange mass spectrometry (HDX-MS) and molecular dynamics (MD) simulation. It was found that B12 binding increased the stability of BtuF. In addition, fast exchange regions of BtuF were localized. Most importantly, midpoint of hinge helix in BtuF was found highly flexible, and binding of B12 proceed in a manner similar to the Venus flytrap mechanism. Our study therefore delineates a clear view of BtuF delivering B12, and demonstrated a hybrid approach encompassing MS and computer based methods that holds great potential to the probing of conformational dynamics of proteins in action.


Subject(s)
Escherichia coli Proteins/metabolism , Hydrogen Deuterium Exchange-Mass Spectrometry , Molecular Dynamics Simulation , Periplasmic Binding Proteins/metabolism , Vitamin B 12/metabolism , Binding Sites , Biological Transport , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Periplasmic Binding Proteins/chemistry , Periplasmic Binding Proteins/genetics , Protein Binding , Protein Conformation , Protein Stability , Protein Unfolding , Structure-Activity Relationship , Vitamin B 12/chemistry
15.
J Mol Biol ; 433(15): 167036, 2021 07 23.
Article in English | MEDLINE | ID: mdl-33957147

ABSTRACT

Our understanding of what determines ligand affinity of proteins is poor, even with high-resolution structures available. Both the non-covalent ligand-protein interactions and the relative free energies of available conformations contribute to the affinity of a protein for a ligand. Distant, non-binding site residues can influence the ligand affinity by altering the free energy difference between a ligand-free and ligand-bound conformation. Our hypothesis is that when different ligands induce distinct ligand-bound conformations, it should be possible to tweak their affinities by changing the free energies of the available conformations. We tested this idea for the maltose-binding protein (MBP) from Escherichia coli. We used single-molecule Förster resonance energy transfer (smFRET) to distinguish several unique ligand-bound conformations of MBP. We engineered mutations, distant from the binding site, to affect the stabilities of different ligand-bound conformations. We show that ligand affinity can indeed be altered in a conformation-dependent manner. Our studies provide a framework for the tuning of ligand affinity, apart from modifying binding site residues.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Mutation , Periplasmic Binding Proteins/chemistry , Periplasmic Binding Proteins/metabolism , Binding Sites , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Fluorescence Resonance Energy Transfer , Ligands , Models, Molecular , Periplasmic Binding Proteins/genetics , Protein Binding , Protein Conformation , Protein Stability , Single Molecule Imaging
16.
J Immunol Methods ; 494: 113055, 2021 07.
Article in English | MEDLINE | ID: mdl-33857474

ABSTRACT

To develop a suitable and effective vaccine against Staphylococcus aureus (S. aureus), we selected the Hla-MntC-SACOL0723 (HMS) recombinant protein with two different formulations of alum and Monophosphoryl lipid A (MPL) adjuvants. In this study, we aimed to evaluate the potentials of alum and MPL adjuvants in stimulating the immune response of HMS vaccine candidate against S. aureus. To evaluate the type of induced immune response, anti-HMS total IgG, IgG1, IgG2a, and IFN-γ, IL-2, IL-4, and IL-17 cytokines were determined after vaccination of mice with HMS-alum, HMS-MPL candidates. Mice were challenged with Methicillin-resistant Staphylococcus aureus (MRSA) was isolated from pressure sores and evaluated for bacterial load in the kidney homogenates and survival rate. It was observed that total IgG and isotypes (IgG1 and IgG2a), IL-4, and IL-17 were significantly increased in the group that received HMS-alum vaccine compared with the group that received HMS-MPL formulation. On the other hand, the levels of IFN-γ and IL-2 cytokines in the group that received HMS-MPL were higher than the group that received HMS-alum formulation. Bacterial load in the mice who received HMS protein formulated with alum adjuvant was reduced more than the mice who received HMS protein formulated with MPL adjuvant. Histopathological analysis showed more pathological changes in kidney tissues of the group received of HMS-MPL compared with the HMS-alum formulation. The survival rate was equal in both groups of immunized with HMS-alum and HMS-MPL formulations. Finally, it could be concluded that both adjuvants of alum and MPL are suitable immune response enhancers to HMS vaccine candidate.


Subject(s)
Kidney/pathology , Methicillin-Resistant Staphylococcus aureus/physiology , Periplasmic Binding Proteins/genetics , Sepsis/immunology , Staphylococcal Infections/immunology , Staphylococcal Vaccines/immunology , Staphylococcus aureus/physiology , Alum Compounds , Animals , Female , HLA Antigens/genetics , Immunoglobulin G/metabolism , Interleukin-17/metabolism , Interleukin-4/metabolism , Lipid A/analogs & derivatives , Lipid A/immunology , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/genetics , Sepsis/prevention & control , Up-Regulation
17.
J Bacteriol ; 203(10)2021 04 21.
Article in English | MEDLINE | ID: mdl-33685971

ABSTRACT

Polyamines are essential for biofilm formation in Escherichia coli, but it is still unclear which polyamines are primarily responsible for this phenomenon. To address this issue, we constructed a series of E. coli K-12 strains with mutations in genes required for the synthesis and metabolism of polyamines. Disruption of the spermidine synthase gene (speE) caused a severe defect in biofilm formation. This defect was rescued by the addition of spermidine to the medium but not by putrescine or cadaverine. A multidrug/spermidine efflux pump membrane subunit (MdtJ)-deficient strain was anticipated to accumulate more spermidine and result in enhanced biofilm formation compared to the MdtJ+ strain. However, the mdtJ mutation did not affect intracellular spermidine or biofilm concentrations. E. coli has the spermidine acetyltransferase (SpeG) and glutathionylspermidine synthetase/amidase (Gss) to metabolize intracellular spermidine. Under biofilm-forming conditions, not Gss but SpeG plays a major role in decreasing the too-high intracellular spermidine concentrations. Additionally, PotFGHI can function as a compensatory importer of spermidine when PotABCD is absent under biofilm-forming conditions. Last, we report here that, in addition to intracellular spermidine, the periplasmic binding protein (PotD) of the spermidine preferential ABC transporter is essential for stimulating biofilm formation.IMPORTANCE Previous reports have speculated on the effect of polyamines on bacterial biofilm formation. However, the regulation of biofilm formation by polyamines in Escherichia coli has not yet been assessed. The identification of polyamines that stimulate biofilm formation is important for developing novel therapies for biofilm-forming pathogens. This study sheds light on biofilm regulation in E. coli Our findings provide conclusive evidence that only spermidine can stimulate biofilm formation in E. coli cells, not putrescine or cadaverine. Last, ΔpotD inhibits biofilm formation even though the spermidine is synthesized inside the cells from putrescine. Since PotD is significant for biofilm formation and there is no ortholog of the PotABCD transporter in humans, PotD could be a target for the development of biofilm inhibitors.


Subject(s)
Biofilms/growth & development , Escherichia coli K12/physiology , Escherichia coli Proteins/metabolism , Membrane Transport Proteins/metabolism , Periplasmic Binding Proteins/metabolism , Spermidine/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Acetyltransferases/metabolism , Amide Synthases/metabolism , Cadaverine/pharmacology , Culture Media , Escherichia coli K12/drug effects , Escherichia coli K12/genetics , Escherichia coli Proteins/genetics , Gene Deletion , Membrane Transport Proteins/genetics , Mutation , Operon , Periplasmic Binding Proteins/genetics , Putrescine/pharmacology , Spermidine/pharmacology , Spermidine Synthase/genetics , Spermidine Synthase/metabolism
18.
Infect Immun ; 89(6)2021 05 17.
Article in English | MEDLINE | ID: mdl-33782154

ABSTRACT

Helicobacter pylori infection is the leading cause of chronic gastritis, which can develop into gastric cancer. Eliminating H. pylori infection with antibiotics achieves the prevention of gastric cancer. Currently, the prevalence of H. pylori resistance to clarithromycin and metronidazole, and the dual resistance to metronidazole and clarithromycin (C_R, M_R, and C/M_R, respectively), remains at a high level worldwide. As a means of exploring new candidate proteins for the management of H. pylori infection, secreted proteins from antibiotic-susceptible and antibiotic-resistant H. pylori-associated gastritis strains were obtained by in-solution tryptic digestion coupled with nano-liquid chromatography tandem mass spectrometry (nano-LC-MS/MS). A total of 583, 582, 590, and 578 differential expressed proteins were identified from C_R, M_R, C/M_R, and antibiotic-sensitive strain (S_S) samples, respectively. Of these, 23 overlapping proteins were found by Venn diagram analysis. Based on heat map analyses, the most and least differing protein expressions were observed from C/M_R strains and S_S strains, respectively. Of the proteins secreted by the S_S strain, only nine were found. After predicting the protein interaction with metronidazole and clarithromycin via the STITCH database, the two most interesting proteins were found to be rpoBC and FBPAII. After quantitative real-time reverse transcription PCR (qRT-PCR) analysis, a downregulation of rpoB from M_R strains was observed, suggesting a relationship of rpoB to metronidazole sensitivity. Inversely, an upregulation of fba from C_R, M_R, and C/M_R strains was noticed, suggesting the paradoxical expression of FBPAII and the fba gene. This report is the first to demonstrate the association of these two novel secreted proteins, namely, rpoBC and FBPAII, with antibiotic-sensitive H. pylori-associated gastritis strains.


Subject(s)
DNA-Directed RNA Polymerases/metabolism , Gastritis/microbiology , Helicobacter Infections/microbiology , Helicobacter pylori , Periplasmic Binding Proteins/metabolism , Anti-Bacterial Agents/pharmacology , Chromatography, Liquid , DNA-Directed RNA Polymerases/genetics , Drug Resistance, Bacterial , Gastritis/epidemiology , Helicobacter Infections/epidemiology , Helicobacter pylori/drug effects , Helicobacter pylori/genetics , Humans , Microbial Sensitivity Tests , Periplasmic Binding Proteins/genetics , Proteomics/methods , Tandem Mass Spectrometry
19.
Sci Rep ; 11(1): 5290, 2021 03 05.
Article in English | MEDLINE | ID: mdl-33674702

ABSTRACT

Signal peptides and secretory carrier proteins are commonly used to secrete heterologous recombinant protein in Gram-negative bacteria. The Escherichia coli osmotically-inducible protein Y (OsmY) is a carrier protein that secretes a target protein extracellularly, and we have previously applied it in the Bacterial Extracellular Protein Secretion System (BENNY) to accelerate directed evolution. In this study, we reported the first application of random and combinatorial mutagenesis on a carrier protein to enhance total secretory target protein production. After one round of random mutagenesis followed by combining the mutations found, OsmY(M3) (L6P, V43A, S154R, V191E) was identified as the best carrier protein. OsmY(M3) produced 3.1 ± 0.3 fold and 2.9 ± 0.8 fold more secretory Tfu0937 ß-glucosidase than its wildtype counterpart in E. coli strains BL21(DE3) and C41(DE3), respectively. OsmY(M3) also produced more secretory Tfu0937 at different cultivation temperatures (37 °C, 30 °C and 25 °C) compared to the wildtype. Subcellular fractionation of the expressed protein confirmed the essential role of OsmY in protein secretion. Up to 80.8 ± 12.2% of total soluble protein was secreted after 15 h of cultivation. When fused to a red fluorescent protein or a lipase from Bacillus subtillis, OsmY(M3) also produced more secretory protein compared to the wildtype. In this study, OsmY(M3) variant improved the extracellular production of three proteins originating from diverse organisms and with diverse properties, clearly demonstrating its wide-ranging applications. The use of random and combinatorial mutagenesis on the carrier protein demonstrated in this work can also be further extended to evolve other signal peptides or carrier proteins for secretory protein production in E. coli.


Subject(s)
Bacterial Secretion Systems/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Mutagenesis , Periplasmic Binding Proteins/metabolism , Secretory Pathway/genetics , Bacillus subtilis/enzymology , Escherichia coli Proteins/genetics , Lipase/genetics , Lipase/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microorganisms, Genetically-Modified , Mutation Rate , Periplasmic Binding Proteins/genetics , Protein Sorting Signals/genetics , Protein Transport/genetics , Recombinant Fusion Proteins/metabolism , Temperature , Thermobifida/enzymology , beta-Glucosidase/genetics , beta-Glucosidase/metabolism , Red Fluorescent Protein
20.
Neuron ; 108(1): 17-32, 2020 10 14.
Article in English | MEDLINE | ID: mdl-33058762

ABSTRACT

The actions of neuromodulation are thought to mediate the ability of the mammalian brain to dynamically adjust its functional state in response to changes in the environment. Altered neurotransmitter (NT) and neuromodulator (NM) signaling is central to the pathogenesis or treatment of many human neurological and psychiatric disorders, including Parkinson's disease, schizophrenia, depression, and addiction. To reveal the precise mechanisms by which these neurochemicals regulate healthy and diseased neural circuitry, one needs to measure their spatiotemporal dynamics in the living brain with great precision. Here, we discuss recent development, optimization, and applications of optical approaches to measure the spatial and temporal profiles of NT and NM release in the brain using genetically encoded sensors for in vivo studies.


Subject(s)
Biosensing Techniques , Brain/metabolism , Neurons/metabolism , Neurotransmitter Agents/metabolism , Optical Imaging , Optogenetics , Animals , Brain/diagnostic imaging , Humans , Periplasmic Binding Proteins/genetics , Protein Engineering , Receptors, G-Protein-Coupled/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...