Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 994
Filter
1.
Protein Expr Purif ; 219: 106487, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38657915

ABSTRACT

The bacterial Efe system functions as an importer of free Fe2+ into cells independently of iron-chelating compounds such as siderophores and consisted of iron-binding protein EfeO, peroxidase EfeB, and transmembrane permease EfeU. While we and other researchers reported crystal structures of EfeO and EfeB, that of EfeU remains undetermined. In this study, we constructed expression system of EfeU derived from Escherichia coli, selected E. coli Rosetta-gami 2 (DE3) as an expression host, and succeeded in purification of the proteins which were indicated to form an oligomer by blue native PAGE. We obtained preliminary data of the X-ray crystallography, suggesting that expression and purification methods we established in this study enable structural analysis of the bacterial Efe system.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Iron , Escherichia coli/genetics , Escherichia coli/metabolism , Crystallography, X-Ray , Escherichia coli Proteins/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/isolation & purification , Iron/metabolism , Iron/chemistry , Gene Expression , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Recombinant Proteins/biosynthesis , Iron-Binding Proteins/chemistry , Iron-Binding Proteins/genetics , Iron-Binding Proteins/isolation & purification , Iron-Binding Proteins/metabolism
2.
J Biol Chem ; 298(4): 101789, 2022 04.
Article in English | MEDLINE | ID: mdl-35247385

ABSTRACT

Synchronized transcription elongation complexes (TECs) are a fundamental tool for in vitro studies of transcription and RNA folding. Transcription elongation can be synchronized by omitting one or more nucleoside triphosphates from an in vitro transcription reaction so that RNA polymerase can only transcribe to the first occurrence of the omitted nucleotide(s) in the coding DNA strand. This approach was developed over four decades ago and has been applied extensively in biochemical investigations of RNA polymerase enzymes but has not been optimized for RNA-centric assays. In this work, we describe the development of a system for isolating synchronized TECs from an in vitro transcription reaction. Our approach uses a custom 5' leader sequence, called capture sequence 3-structure cassette 1 (C3-SC1), to reversibly capture synchronized TECs on magnetic beads. We first show, using electrophoretic mobility shift and high-resolution in vitro transcription assays, that complexes isolated by this procedure, called C3-SC1TECs, are >95% pure, >98% active, highly synchronous (94% of complexes chase in <15s upon addition of saturating nucleoside triphosphates), and compatible with solid-phase transcription; the yield of this purification is ∼8%. We then show that C3-SC1TECs perturb, but do not interfere with, the function of ZTP (5-aminoimidazole-4-carboxamide riboside 5'-triphosphate)-sensing and ppGpp (guanosine-3',5'-bisdiphosphate)-sensing transcriptional riboswitches. For both riboswitches, transcription using C3-SC1TECs improved the efficiency of transcription termination in the absence of ligand but did not inhibit ligand-induced transcription antitermination. Given these properties, C3-SC1TECs will likely be useful for developing biochemical and biophysical RNA assays that require high-performance, quantitative bacterial in vitro transcription.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Transcription, Genetic , Transcriptional Elongation Factors , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/isolation & purification , Escherichia coli Proteins/metabolism , Ligands , Magnetic Phenomena , Nucleosides , RNA , Riboswitch , Transcriptional Elongation Factors/chemistry , Transcriptional Elongation Factors/isolation & purification
3.
Commun Biol ; 4(1): 1337, 2021 11 25.
Article in English | MEDLINE | ID: mdl-34824357

ABSTRACT

Membrane proteins are essential for cellular growth, signalling and homeostasis, making up a large proportion of therapeutic targets. However, the necessity for a solubilising agent to extract them from the membrane creates challenges in their structural and functional study. Although amphipols have been very effective for single-particle electron cryo-microscopy (cryoEM) and mass spectrometry, they rely on initial detergent extraction before exchange into the amphipol environment. Therefore, circumventing this pre-requirement would be a big advantage. Here we use an alternative type of amphipol: a cycloalkane-modified amphiphile polymer (CyclAPol) to extract Escherichia coli AcrB directly from the membrane and demonstrate that the protein can be isolated in a one-step purification with the resultant cryoEM structure achieving 3.2 Å resolution. Together this work shows that cycloalkane amphipols provide a powerful approach for the study of membrane proteins, allowing native extraction and high-resolution structure determination by cryoEM.


Subject(s)
Cryoelectron Microscopy/methods , Cycloparaffins/chemistry , Escherichia coli Proteins/isolation & purification , Escherichia coli/physiology , Multidrug Resistance-Associated Proteins/isolation & purification , Polymers/chemistry , Cryoelectron Microscopy/instrumentation
4.
Epidemiol Infect ; 149: e244, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34726137

ABSTRACT

In June 2020, a large-scale food poisoning outbreak involving about 3000 elementary and junior high school students occurred in Yashio, Saitama, Japan. A school lunch was the only food stuff ingested by all of the patients. Escherichia coli serotype O7:H4 carrying the astA gene for enteroaggregative E. coli (EAggEC) heat-stable enterotoxin 1 (EAST1) was detected in faecal specimens from the patients, and sample inspection revealed its presence in a seaweed salad and red seaweed (Gigartina tenella) as one of the raw materials. Analysis of the antibiotic sensitivity of the isolates revealed resistance to ampicillin and cefotaxime. All isolates were confirmed to be of the same origin by pulsed-field gel electrophoresis after digestion with the restriction enzyme XbaI, and single nucleotide polymorphism analysis using whole genome sequencing. To our knowledge, this is the first report of a large-scale food poisoning caused by E. coli O7:H4, which lacks well-characterized virulence genes other than astA.


Subject(s)
Disease Outbreaks , Escherichia coli/isolation & purification , Foodborne Diseases/epidemiology , Foodborne Diseases/microbiology , Bacterial Toxins/genetics , Bacterial Toxins/isolation & purification , DNA, Bacterial/genetics , Drug Resistance, Bacterial , Enterotoxins/genetics , Enterotoxins/isolation & purification , Escherichia coli/genetics , Escherichia coli Infections/epidemiology , Escherichia coli Infections/microbiology , Escherichia coli Proteins/genetics , Escherichia coli Proteins/isolation & purification , Food Contamination , Food Services , Foodborne Diseases/etiology , Humans , Japan/epidemiology , Rhodophyta , Whole Genome Sequencing
5.
Sci Rep ; 11(1): 18885, 2021 09 23.
Article in English | MEDLINE | ID: mdl-34556749

ABSTRACT

Since 1993, when the structure of Escherichia coli type II L-asparaginase (EcAII) in complex with L-aspartate was firstly reported, many structures of the wild type and mutated enzyme have been deposited in the Protein Data Bank. None of them report the full structure of the monomer in its ligand-free, open conformation, mainly because of the high dynamic and flexibility of the active site flexible loop. Here we report for the first time the structure of EcAII wild type in its open conformation comprising, for at least one protomer, clear electron density for the active site flexible loop (PDB ID: 6YZI). The structural element is highly mobile and it is transposed onto the rigid part of the active site upon substrate binding to allow completion of the enzyme catalytic center, thanks to key residues that serve as hinges and anchoring points. In the substrate binding pocket, several highly conserved water molecules are coordinated by residues involved in substrate binding, comprising two water molecules very likely involved in the enzyme catalytic process. We also describe, by molecular dynamics simulations, how the transposition of the loop, besides providing the proximity of residues needed for catalysis, causes a general stabilization of the protein.


Subject(s)
Asparaginase/ultrastructure , Escherichia coli Proteins/ultrastructure , Recombinant Proteins/ultrastructure , Asparaginase/isolation & purification , Catalytic Domain , Escherichia coli/enzymology , Escherichia coli Proteins/isolation & purification , Molecular Dynamics Simulation , Protein Stability , Recombinant Proteins/isolation & purification , X-Ray Diffraction
6.
Nat Commun ; 12(1): 5277, 2021 09 06.
Article in English | MEDLINE | ID: mdl-34489474

ABSTRACT

The pyruvate dehydrogenase complex (PDHc) links glycolysis to the citric acid cycle by converting pyruvate into acetyl-coenzyme A. PDHc encompasses three enzymatically active subunits, namely pyruvate dehydrogenase, dihydrolipoyl transacetylase, and dihydrolipoyl dehydrogenase. Dihydrolipoyl transacetylase is a multidomain protein comprising a varying number of lipoyl domains, a peripheral subunit-binding domain, and a catalytic domain. It forms the structural core of the complex, provides binding sites for the other enzymes, and shuffles reaction intermediates between the active sites through covalently bound lipoyl domains. The molecular mechanism by which this shuttling occurs has remained elusive. Here, we report a cryo-EM reconstruction of the native E. coli dihydrolipoyl transacetylase core in a resting state. This structure provides molecular details of the assembly of the core and reveals how the lipoyl domains interact with the core at the active site.


Subject(s)
Escherichia coli Proteins/chemistry , Pyruvate Dehydrogenase Complex/chemistry , Pyruvate Dehydrogenase Complex/metabolism , Catalytic Domain , Cryoelectron Microscopy , Dihydrolipoyllysine-Residue Acetyltransferase/chemistry , Dihydrolipoyllysine-Residue Acetyltransferase/metabolism , Escherichia coli Proteins/isolation & purification , Escherichia coli Proteins/metabolism , Hydrophobic and Hydrophilic Interactions , Lysine/analogs & derivatives , Lysine/chemistry , Lysine/metabolism , Models, Molecular , Protein Domains , Pyruvate Dehydrogenase Complex/isolation & purification , Thioctic Acid/analogs & derivatives , Thioctic Acid/chemistry , Thioctic Acid/metabolism
7.
Foodborne Pathog Dis ; 18(8): 616-625, 2021 08.
Article in English | MEDLINE | ID: mdl-34403269

ABSTRACT

While Shiga toxin-producing Escherichia coli (STEC) is a major foodborne pathogen worldwide, data on the molecular and phylogenetic properties of STEC isolates from retail beef samples in China remain scant. Fresh retail beef samples (n = 1062) were collected from eight provinces, and STEC isolates were recovered and characterized. PCR data showed that more than 50% of the samples were stx positive, and 82 STEC isolates were recovered from 14.8% (79/535) stx-positive enriched broths. In contrast, all ciprofloxacin resistant isolates (n = 19) and 13 cefotaxime (CTX) resistant isolates were eae positive and belonged to three serotypes: O111:H8, O26:H11, or O157:H7. Point mutations in quinolone resistance-determining regions and plasmid-mediated quinolone resistance determinants were identified in 16 and 20 isolates, respectively. BlaCTX-M and a point mutation (C-42T) in ampC promoter were detected in 15 and 8 of the CTX resistant isolates, respectively. In addition, macrolide resistance gene mphA was identified in eight azithromycin resistant O111:H8 isolates and one O26:H11 isolate. Single nucleotide polymorphism analysis demonstrated that the O26 and O157 isolates had multiple origins, but the O111 isolates were closely related. Taken together, our data demonstrated that several sequence types associated with hemolytic uremic syndrome from the retail beef samples in China had developed into dangerous multidrug resistant pathogens. The resistant phenotype can facilitate their transmission among the farm animals and human beings when there is an antimicrobial selective pressure.


Subject(s)
Escherichia coli Proteins/isolation & purification , Food Microbiology/statistics & numerical data , Red Meat/microbiology , Shiga-Toxigenic Escherichia coli/genetics , Shiga-Toxigenic Escherichia coli/isolation & purification , Animals , China , Drug Resistance, Multiple/genetics , Humans , Phylogeny , Polymerase Chain Reaction , Polymorphism, Single Nucleotide , Serogroup , Virulence Factors/genetics
8.
Nat Commun ; 12(1): 4174, 2021 07 07.
Article in English | MEDLINE | ID: mdl-34234105

ABSTRACT

The folding of ß-barrel outer membrane proteins (OMPs) in Gram-negative bacteria is catalysed by the ß-barrel assembly machinery (BAM). How lateral opening in the ß-barrel of the major subunit BamA assists in OMP folding, and the contribution of membrane disruption to BAM catalysis remain unresolved. Here, we use an anti-BamA monoclonal antibody fragment (Fab1) and two disulphide-crosslinked BAM variants (lid-locked (LL), and POTRA-5-locked (P5L)) to dissect these roles. Despite being lethal in vivo, we show that all complexes catalyse folding in vitro, albeit less efficiently than wild-type BAM. CryoEM reveals that while Fab1 and BAM-P5L trap an open-barrel state, BAM-LL contains a mixture of closed and contorted, partially-open structures. Finally, all three complexes globally destabilise the lipid bilayer, while BamA does not, revealing that the BAM lipoproteins are required for this function. Together the results provide insights into the role of BAM structure and lipid dynamics in OMP folding.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Escherichia coli Proteins/metabolism , Hydrolases/metabolism , Liposomes/metabolism , Protein Folding , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/isolation & purification , Bacterial Outer Membrane Proteins/ultrastructure , Cryoelectron Microscopy , Dynamic Light Scattering , Escherichia coli Proteins/genetics , Escherichia coli Proteins/isolation & purification , Escherichia coli Proteins/ultrastructure , Hydrolases/genetics , Hydrolases/isolation & purification , Hydrolases/ultrastructure , Lipid Metabolism , Liposomes/ultrastructure , Molecular Dynamics Simulation , Protein Conformation, beta-Strand , Proteolipids/metabolism , Proteolipids/ultrastructure , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure
9.
Biosci Rep ; 41(7)2021 07 30.
Article in English | MEDLINE | ID: mdl-34142112

ABSTRACT

Protein purification is the vital basis to study the function, structure and interaction of proteins. Widely used methods are affinity chromatography-based purifications, which require different chromatography columns and harsh conditions, such as acidic pH and/or adding imidazole or high salt concentration, to elute and collect the purified proteins. Here we established an easy and fast purification method for soluble proteins under mild conditions, based on the light-induced protein dimerization system improved light-induced dimer (iLID), which regulates protein binding and release with light. We utilize the biological membrane, which can be easily separated by centrifugation, as the port to anchor the target proteins. In Xenopus laevis oocyte and Escherichia coli, the blue light-sensitive part of iLID, AsLOV2-SsrA, was targeted to the plasma membrane by different membrane anchors. The other part of iLID, SspB, was fused with the protein of interest (POI) and expressed in the cytosol. The SspB-POI can be captured to the membrane fraction through light-induced binding to AsLOV2-SsrA and then released purely to fresh buffer in the dark after simple centrifugation and washing. This method, named mem-iLID, is very flexible in scale and economic. We demonstrate the quickly obtained yield of two pure and fully functional enzymes: a DNA polymerase and a light-activated adenylyl cyclase. Furthermore, we also designed a new SspB mutant for better dissociation and less interference with the POI, which could potentially facilitate other optogenetic manipulations of protein-protein interaction.


Subject(s)
Adenylyl Cyclases/isolation & purification , DNA-Directed DNA Polymerase/isolation & purification , Escherichia coli Proteins/isolation & purification , Optogenetics , Protein Engineering , Xenopus Proteins/isolation & purification , Adenylyl Cyclases/genetics , Animals , Cell Membrane/enzymology , Cell Membrane/genetics , Cost-Benefit Analysis , DNA-Directed DNA Polymerase/genetics , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Light , Mutation , Optogenetics/economics , Protein Binding , Protein Engineering/economics , Protein Multimerization , Recombinant Fusion Proteins/isolation & purification , Time Factors , Workflow , Xenopus Proteins/genetics , Xenopus laevis/genetics , Xenopus laevis/metabolism
10.
Methods Mol Biol ; 2323: 67-73, 2021.
Article in English | MEDLINE | ID: mdl-34086274

ABSTRACT

For structural, biochemical, or pharmacological studies, it is required to have pure RNA in large quantities. We previously devised a generic approach that allows for efficient in vivo expression of recombinant RNA in Escherichia coli. We have extended the "tRNA scaffold" method to RNA-protein coexpression in order to express and purify RNA by affinity in native condition. As a proof of concept, we present the expression and the purification of the AtRNA-mala in complex with the MS2 coat protein.


Subject(s)
Chromatography, Affinity/methods , Cloning, Molecular/methods , Escherichia coli Proteins/isolation & purification , Escherichia coli/chemistry , RNA-Binding Proteins/isolation & purification , RNA/isolation & purification , Ampicillin/pharmacology , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/genetics , Capsid , Chloramphenicol/pharmacology , Computer Simulation , Drug Resistance, Microbial/genetics , Electrophoresis, Polyacrylamide Gel/methods , Escherichia coli/genetics , Escherichia coli Proteins/biosynthesis , Levivirus/genetics , Models, Molecular , Nucleic Acid Conformation , Operator Regions, Genetic , Plasmids/genetics , RNA/biosynthesis , RNA, Bacterial/genetics , RNA, Bacterial/isolation & purification , RNA, Viral/genetics , RNA, Viral/isolation & purification , RNA-Binding Proteins/biosynthesis
11.
Nat Commun ; 12(1): 2363, 2021 04 22.
Article in English | MEDLINE | ID: mdl-33888690

ABSTRACT

Cell-free gene expression (CFE) systems from crude cellular extracts have attracted much attention for biomanufacturing and synthetic biology. However, activating membrane-dependent functionality of cell-derived vesicles in bacterial CFE systems has been limited. Here, we address this limitation by characterizing native membrane vesicles in Escherichia coli-based CFE extracts and describing methods to enrich vesicles with heterologous, membrane-bound machinery. As a model, we focus on bacterial glycoengineering. We first use multiple, orthogonal techniques to characterize vesicles and show how extract processing methods can be used to increase concentrations of membrane vesicles in CFE systems. Then, we show that extracts enriched in vesicle number also display enhanced concentrations of heterologous membrane protein cargo. Finally, we apply our methods to enrich membrane-bound oligosaccharyltransferases and lipid-linked oligosaccharides for improving cell-free N-linked and O-linked glycoprotein synthesis. We anticipate that these methods will facilitate on-demand glycoprotein production and enable new CFE systems with membrane-associated activities.


Subject(s)
Cell-Derived Microparticles/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/cytology , Glycoproteins/biosynthesis , Hexosyltransferases/metabolism , Membrane Proteins/metabolism , Protein Biosynthesis , Cell Membrane/genetics , Cell Membrane/metabolism , Cell-Derived Microparticles/genetics , Chromatography, High Pressure Liquid/methods , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/isolation & purification , Glycoproteins/isolation & purification , Hexosyltransferases/genetics , Hexosyltransferases/isolation & purification , Mass Spectrometry/methods , Membrane Proteins/genetics , Membrane Proteins/isolation & purification , Oligosaccharides/metabolism , Protein Engineering , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
12.
ACS Appl Mater Interfaces ; 13(10): 11571-11578, 2021 Mar 17.
Article in English | MEDLINE | ID: mdl-33661597

ABSTRACT

The membrane proteins of microbes are at the forefront of host and parasite interactions. Having a general view of the functions of microbial membrane proteins is vital for many biomedical studies on microbiota. Nevertheless, due to the strong hydrophobicity and low concentration of membrane proteins, it is hard to efficiently enrich and digest the proteins for mass spectrometry analysis. Herein, we design an enzymatic nanoreactor for the digestion of membrane proteins using methylated well-ordered hexagonal mesoporous silica (Met-SBA-15). The material can efficiently extract hydrophobic membrane proteins and host the proteolysis in nanopores. The performance of the enzymatic nanoreactor is first demonstrated using standard hydrophobic proteins and then validated using membrane proteins extracted from Escherichia coli (E. coli) or a mixed bacterial sample of eight strains. Using the nanoreactor, 431 membrane proteins are identified from E. coli, accounting for 38.5% of all membrane proteins of the species, which is much more than that by the widely used in-solution digestion protocol. From the mixed bacterial sample of eight strains, 1395 membrane proteins are identified using the nanoreactor. On the contrary, the traditional in-solution proteolysis workflow only leads to the identification of 477 membrane proteins, demonstrating that the Met-SBA-15 can be offered as an excellent tool for microbial membrane proteome research and is expected to be used in human microbiota studies, e.g. host-microbe interactions.


Subject(s)
Bacterial Outer Membrane Proteins/isolation & purification , Escherichia coli Proteins/isolation & purification , Escherichia coli/chemistry , Proteomics/methods , Silicon Dioxide/chemistry , Adsorption , Bacterial Outer Membrane Proteins/analysis , Escherichia coli Proteins/analysis , Porosity
13.
Int J Biol Macromol ; 180: 242-251, 2021 Jun 01.
Article in English | MEDLINE | ID: mdl-33737181

ABSTRACT

Fatty acid desaturase catalyzes the desaturation reactions by insertion of double bonds into the fatty acyl chain, producing unsaturated fatty acids. Though soluble fatty acid desaturases have been studied widely in advanced organisms, there are very limited studies of membrane fatty acid desaturases due to the difficulty of generating recombinant desaturase. Brassica napus is a rapeseed, which possesses a range of different membrane-bound desaturases capable of producing fatty acids including Δ3, Δ4, Δ8, Δ9, Δ12, and Δ15 fatty acids. The 1155 bp open reading frame of Δ12 fatty acid desaturase (FAD12) from Brassica napus codes for 383 amino acid residues with a molecular weight of 44 kDa. It was expressed in Escherichia coli at 37 °C in soluble and insoluble forms when induced with 0.5 mM IPTG. Soluble FAD12 has been purified using Ni2+-Sepharose affinity chromatography with a total protein yield of 0.728 mg/mL. Gas chromatography-mass spectrometry (GC-MS) analysis revealed that desaturase activity of FAD12 could produce linoleic acid from oleic acid at a retention time of 17.6 with a conversion rate of 47%. Characterization of purified FAD12 revealed the optimal temperature of FAD12 was 50 °C with 2 mM preferred substrate concentration of oleic acid. Analysis of circular dichroism (CD) showed FAD12 was made up of 47.3% and 0.9% of alpha-helix and ß-sheet secondary structures. The predicted Tm value was 50.2 °C.


Subject(s)
Brassica napus/enzymology , Escherichia coli Proteins/chemistry , Escherichia coli/enzymology , Fatty Acid Desaturases/chemistry , Membrane Proteins/chemistry , Plant Proteins/chemistry , Amino Acid Sequence , Brassica napus/genetics , Chromatography, Affinity , Circular Dichroism , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/isolation & purification , Escherichia coli Proteins/metabolism , Fatty Acid Desaturases/genetics , Fatty Acid Desaturases/isolation & purification , Fatty Acid Desaturases/metabolism , Gas Chromatography-Mass Spectrometry , Genes, Bacterial , Genes, Plant , Hot Temperature , Linoleic Acid/metabolism , Membrane Proteins/genetics , Membrane Proteins/isolation & purification , Membrane Proteins/metabolism , Molecular Weight , Oleic Acid/metabolism , Open Reading Frames , Plant Proteins/genetics , Plant Proteins/isolation & purification , Plant Proteins/metabolism , Protein Structure, Secondary , Solubility
14.
Sci Rep ; 11(1): 3940, 2021 02 16.
Article in English | MEDLINE | ID: mdl-33594158

ABSTRACT

The membrane insertase YidC inserts newly synthesized proteins by its hydrophobic slide consisting of the two transmembrane (TM) segments TM3 and TM5. Mutations in this part of the protein affect the insertion of the client proteins. We show here that a quintuple mutation, termed YidC-5S, inhibits the insertion of the subunit a of the FoF1 ATP synthase but has no effect on the insertion of the Sec-independent M13 procoat protein and the C-tail protein SciP. Further investigations show that the interaction of YidC-5S with SecY is inhibited. The purified and fluorescently labeled YidC-5S did not approach SecYEG when both were co-reconstituted in proteoliposomes in contrast to the co-reconstituted YidC wild type. These results suggest that TM3 and TM5 are involved in the formation of a common YidC-SecYEG complex that is required for the insertion of Sec/YidC-dependent client proteins.


Subject(s)
Escherichia coli Proteins/metabolism , Membrane Transport Proteins/metabolism , SEC Translocation Channels/metabolism , Escherichia coli , Escherichia coli Proteins/genetics , Escherichia coli Proteins/isolation & purification , Membrane Transport Proteins/genetics , Membrane Transport Proteins/isolation & purification , Proteolipids/metabolism , SEC Translocation Channels/isolation & purification
15.
Curr Genet ; 67(2): 295-303, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33386487

ABSTRACT

DNA polymerase IV (pol IV) is expressed at increased levels in Escherichia coli cells that suffer DNA damage. In a recent live-cell single-molecule fluorescence microscopy study, we demonstrated that the formation of pol IV foci is strongly recB-dependent in cells treated with the DNA break-inducing antibiotic ciprofloxacin. The results of that study support a model in which pol IV acts to extend D-loop structures during recombinational repair of DNA double-strand breaks. In the present study, we extend upon this work, investigating the UmuD and UmuD' proteins as potential modulators of pol IV activity in ciprofloxacin-treated cells. We found that the non-cleavable mutant UmuD(K97A) promotes long-lived association of pol IV with the nucleoid, whereas its cleaved form, UmuD', which accumulates in DNA-damaged cells, reduces binding. The results provide additional support for a model in which UmuD and UmuD' directly modulate pol IV-binding to the nucleoid.


Subject(s)
DNA Polymerase beta/genetics , DNA-Directed DNA Polymerase/genetics , Escherichia coli Proteins/genetics , Escherichia coli/genetics , Mutant Proteins/genetics , DNA Damage/genetics , DNA Polymerase beta/isolation & purification , DNA Repair/genetics , DNA Replication/genetics , DNA-Directed DNA Polymerase/isolation & purification , Escherichia coli Proteins/isolation & purification , Microscopy, Fluorescence , Protein Binding/genetics , Single Molecule Imaging
16.
Biol Pharm Bull ; 44(1): 125-130, 2021.
Article in English | MEDLINE | ID: mdl-33390539

ABSTRACT

The constitutive active/androstane receptor (CAR) is a nuclear receptor that functions as a xenobiotic sensor, which regulates the expression of enzymes involved in drug metabolism and of efflux transporters. Evaluation of the binding properties between CAR and a drug was assumed to facilitate the prediction of drug-drug interaction, thereby contributing to drug discovery. The purpose of this study is to construct a system for the rapid evaluation of interactions between CAR and drugs. We prepared recombinant CAR protein using the Escherichia coli expression system. Since isolated CAR protein is known to be unstable, we designed a fusion protein with the CAR binding sequence of the nuclear receptor coactivator 1 (NCOA1), which was expressed as a fusion protein with maltose binding protein (MBP), and purified it by several chromatography steps. The thus-obtained CAR/NCOA1 tethered protein (CAR-NCOA1) was used to evaluate the interactions of CAR with agonists and inverse agonists by a thermal denaturation experiment using differential scanning fluorometry (DSF) in the presence and absence of drugs. An increase in the melting temperature was observed with the addition of the drugs, confirming the direct interaction between them and CAR. DSF is easy to set up and compatible with multiwell plate devices (such as 96-well plates). The use of DSF and the CAR-NCOA1 fusion protein together allows for the rapid evaluation of the interaction between a drug and CAR, and is thereby considered to be useful in drug discovery.


Subject(s)
Escherichia coli Proteins/biosynthesis , Gelatin/chemical synthesis , Maltose-Binding Proteins/chemical synthesis , Nuclear Receptor Coactivator 1/biosynthesis , Receptors, Cytoplasmic and Nuclear/biosynthesis , Starch/chemical synthesis , Constitutive Androstane Receptor , Escherichia coli , Escherichia coli Proteins/genetics , Escherichia coli Proteins/isolation & purification , Gene Expression , Nuclear Receptor Coactivator 1/genetics , Nuclear Receptor Coactivator 1/isolation & purification , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/isolation & purification
17.
J Biomol NMR ; 75(1): 25-38, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33501610

ABSTRACT

G protein-coupled receptors (GPCRs) are transmembrane signal transducers which regulate many key physiological process. Since their discovery, their analysis has been limited by difficulties in obtaining sufficient amounts of the receptors in high-quality, functional form from heterologous expression hosts. Albeit highly attractive because of its simplicity and the ease of isotope labeling for NMR studies, heterologous expression of functional GPCRs in E. coli has proven particularly challenging due to the absence of the more evolved protein expression and folding machinery of higher eukaryotic hosts. Here we first give an overview on the previous strategies for GPCR E. coli expression and then describe the development of an optimized robust protocol for the E. coli expression and purification of two mutants of the turkey ß1-adrenergic receptor (ß1AR) uniformly or selectively labeled in 15N or 2H,15N. These mutants had been previously optimized for thermal stability using insect cell expression and used successfully in crystallographic and NMR studies. The same sequences were then used for E. coli expression. Optimization of E. coli expression was achieved by a quantitative analysis of losses of receptor material at each step of the solubilization and purification procedure. Final yields are 0.2-0.3 mg receptor per liter culture. Whereas both expressed mutants are well folded and competent for orthosteric ligand binding, the less stable YY-ß1AR mutant also comprises the two native tyrosines Y5.58 and Y7.53, which enable G protein binding. High-quality 1H-15N TROSY spectra were obtained for E. coli-expressed YY-ß1AR in three different functional states (antagonist, agonist, and agonist + G protein-mimicking nanobody-bound), which are identical to spectra obtained of the same forms of the receptor expressed in insect cells. NdeI and AgeI restriction sites introduced into the expression plasmid allow for the easy replacement of the receptor gene by other GPCR genes of interest, and the provided quantitative workflow analysis may guide the respective adaptation of the purification protocol.


Subject(s)
Escherichia coli Proteins/biosynthesis , Escherichia coli Proteins/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Receptors, G-Protein-Coupled/biosynthesis , Receptors, G-Protein-Coupled/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/isolation & purification , Gene Expression , Genetic Vectors/genetics , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular/methods , Protein Binding , Protein Stability , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/isolation & purification , Recombinant Proteins
18.
Comb Chem High Throughput Screen ; 24(5): 737-742, 2021.
Article in English | MEDLINE | ID: mdl-32928079

ABSTRACT

BACKGROUND: The presence of plasmid mediated mcr-1 gene in multidrug resistant Gram-negative bacteria poses a serious public health concern in today's world. OBJECTIVE: The present study was aimed to detect the presence of plasmid mediated mcr-1 encoding resistance to colistin in multiple drug resistant (MDR) E. coli and K. pneumoniae isolates. METHODS: A total of 180 clinical isolates of E. coli (n=120) and K. pneumoniae (n=60) were isolated from different clinical specimens, i.e., urine, blood, stool and pus, from diagnostic labs of two major public sector tertiary care hospitals in Peshawar, Pakistan. MDR profile of these isolates was assessed through Kirby-Baur disc diffusion method. All isolates were screened for colistin resistance by dilution methods. Colistin resistant isolates were subjected to PCR for mcr-1 detection and confirmation was done by Sanger sequencing method. RESULTS: Overall, 83.3% (100/120) E. coli and 93.3% (56/60) K. pneumoniae were detected as MDR. Colistin resistance was found in 23.3% (28/120) E. coli and 40% (24/60) K. pneumoniae isolates, whereas mcr-1 gene was detected in 10 out of 52 colistin resistant isolates, including six E. coli and four K. pneumoniae isolates. Minimum inhibitory concentrations (MICs) of colistin in these ten mcr-1 positive isolates ranged from 4µg/ml to 16µg/ml. All mcr-1 positive isolates showed 99% sequence similarity when compared with other present sequences in GenBank. CONCLUSION: Hence, our study confirms the presence of mcr-1 mediated colistin resistance in the studied area. Therefore, urgently larger scale surveillance studies are recommended to investigate prevalence of mcr-1 mediated colistin resistance and to prevent its further spread in the area.


Subject(s)
Drug Resistance, Multiple, Bacterial/genetics , Escherichia coli Proteins/genetics , Escherichia coli/genetics , Klebsiella pneumoniae/genetics , Anti-Bacterial Agents/pharmacology , Colistin/pharmacology , Dose-Response Relationship, Drug , Escherichia coli/drug effects , Escherichia coli Proteins/isolation & purification , Escherichia coli Proteins/metabolism , Humans , Klebsiella pneumoniae/drug effects , Pakistan
19.
Methods Mol Biol ; 2186: 11-18, 2021.
Article in English | MEDLINE | ID: mdl-32918726

ABSTRACT

The ionic currents passing through nanopores can be used to sequence DNA and identify molecules at the single-molecule level. Recently, researchers have started using nanopores for the detection and analysis of proteins, providing a new platform for single-molecule enzymology studies and more efficient biomolecular sensing applications. For this approach, the homo-oligomeric Cytolysin A (ClyA) nanopore has been demonstrated as a powerful tool. Here, we describe a simple protocol allowing the production of ClyA nanopores. Monomers of ClyA are expressed in Escherichia coli and oligomerized in the presence of detergent. Subsequently, different oligomer variants are electrophoretically resolved and stored in a gel matrix for long-term use.


Subject(s)
Cytotoxins/isolation & purification , Escherichia coli Proteins/isolation & purification , Escherichia coli/metabolism , Nanopores , Perforin/isolation & purification , Cytotoxins/chemistry , Cytotoxins/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Perforin/chemistry , Perforin/metabolism
20.
Protein Pept Lett ; 28(1): 108-114, 2021.
Article in English | MEDLINE | ID: mdl-32520670

ABSTRACT

BACKGROUND: The heterologous production of antimicrobial peptides in bacterial models can produce insoluble proteins due to the lack of proper folding. Fusion proteins have been used to increase the expression and solubility of these types of proteins with varying degrees of success. OBJECTIVES: Here, we demonstrate the use of the small metal-binding proteins CusF3H+ (9.9kDa) and SmbP (9.9kDa) as fusion partners for the soluble expression of the bioactive antimicrobial peptide VpDef(6.9 kDa) in Escherichia coli. METHODS: The recombinant VpDef (rVpDef) peptide was expressed as a translational fusion with CusF3H+ and SmbP in Escherichia coli SHuffle under different small-scale culture conditions. The best conditions were applied to 1-liter cultures, with subsequent purification of the recombinant protein through IMAC chromatography. The recombinant protein was digested using enterokinase to liberate the peptide from the fusion protein, and a second IMAC chromatography step removed the fusion protein. The purified peptide was tested against two Gram-positive and two Gram-negative bacteria. RESULTS: The use either of CusF3H+ or of SmbP results in recombinant proteins that are found in the soluble fraction of the bacterial lysate; these recombinant proteins are easily purified through IMAC chromatography, and rVpDef is readily separated following enterokinase treatment. The purified rVpDef peptide exhibits antimicrobial properties against both Gram-positive and Gram-negative. CONCLUSION: Use of the fusion proteins CusF3H+ and SmbP results in production of a soluble recombinant protein containing the antimicrobial peptide rVpDef that is correctly folded and that retains its antimicrobial properties once purified.


Subject(s)
Copper Transport Proteins , Defensins , Escherichia coli Proteins , Escherichia coli/metabolism , Recombinant Fusion Proteins , Copper Transport Proteins/biosynthesis , Copper Transport Proteins/chemistry , Copper Transport Proteins/genetics , Copper Transport Proteins/isolation & purification , Defensins/biosynthesis , Defensins/chemistry , Defensins/genetics , Defensins/isolation & purification , Escherichia coli/genetics , Escherichia coli Proteins/biosynthesis , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/isolation & purification , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL
...