Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 7820, 2024 04 03.
Article in English | MEDLINE | ID: mdl-38570576

ABSTRACT

Anaplasma marginale infection is one of the most common tick-borne diseases, causing a substantial loss in the beef and dairy production industries. Once infected, the pathogen remains in the cattle for life, allowing the parasites to spread to healthy animals. Since clinical manifestations of anaplasmosis occur late in the disease, a sensitive, accurate, and affordable pathogen identification is crucial in preventing and controlling the infection. To this end, we developed an RPA-CRISPR/Cas12a assay specific to A. marginale infection in bovines targeting the msp4 gene. Our assay is performed at one moderately high temperature, producing fluorescent signals or positive readout of a lateral flow dipstick, which is as sensitive as conventional PCR-based DNA amplification. This RPA-CRISPR/Cas12a assay can detect as few as 4 copies/µl of Anaplasma using msp4 marker without cross-reactivity to other common bovine pathogens. Lyophilized components of the assay can be stored at room temperature for an extended period, indicating its potential for field diagnosis and low-resource settings of anaplasmosis in bovines.


Subject(s)
Anaplasma marginale , Anaplasmosis , Cattle Diseases , Tick-Borne Diseases , Cattle , Animals , Anaplasma marginale/genetics , Anaplasmosis/diagnosis , Anaplasmosis/genetics , CRISPR-Cas Systems , Cattle Diseases/genetics , Tick-Borne Diseases/genetics
2.
Protein Expr Purif ; 210: 106320, 2023 10.
Article in English | MEDLINE | ID: mdl-37301245

ABSTRACT

The native Cry4Aa δ-endotoxin produced exclusively in Bacillus thuringiensis during sporulation as a ∼130-kDa inactive protoxin is confined within the parasporal crystalline inclusion that dissolves at alkaline pH in the midgut lumen of mosquito larvae. Here, the recombinant Cry4Aa toxin over-expressed in Escherichia coli at 30 °C as an alkaline-solubilizable inclusion was found inevitably lost during isolation from the cell lysate (pH ∼6.5) of which host cells were pre-suspended in distilled water (pH ∼5.5). When 100 mM KH2PO4 (pH 5.0) was used as host cell-suspending buffer, the cell lysate's pH became more acidic (pH 5.5), allowing the expressed protoxin to be entirely retained in the form of crystalline inclusion rather than a soluble form, and thus high-yield recovery of the partially purified inclusion was obtained. Upon dialysis of the alkaline-solubilized protoxin against the KH2PO4 buffer, the protoxin precipitate was efficiently recovered and still exhibited high toxicity to Aedes aegypti mosquito larvae. Additionally, the precipitated protoxin was completely resolubilized in 50 mM Na2CO3 buffer (pH 9.0) and proteolytically processed by trypsin to produce the 65-kDa activated toxin comprising ∼47- and ∼20-kDa fragments. In silico structural analysis suggested that His154, His388, His536 and His572 were involved in a dissolution of the Cry4Aa inclusion at pH 6.5, conceivably through interchain salt bridge breakage. Altogether, such an optimized protocol described herein was effective for the preparation of alkaline-solubilizable inclusions of the recombinant Cry4Aa toxin in large amounts (>25 mg per liter culture) that would pave the way for further structure-function relationship studies of different Cry toxins.


Subject(s)
Bacillus thuringiensis , Animals , Bacillus thuringiensis/genetics , Bacillus thuringiensis/chemistry , Escherichia coli/genetics , Renal Dialysis , Endotoxins/genetics , Larva , Hemolysin Proteins/genetics , Bacterial Proteins/genetics , Bacterial Proteins/chemistry
3.
Protein Expr Purif ; 210: 106313, 2023 10.
Article in English | MEDLINE | ID: mdl-37276914

ABSTRACT

Many therapeutic proteins are expressed in Escherichia coli bacteria for the low cost and high yield obtained. However, these gram-negative bacteria also generate undesirable endotoxin byproducts such as lipopolysaccharides (LPS). These endotoxins can induce a human immune response and cause severe inflammation. To mitigate this problem, we have employed the ClearColi BL21 (DE3) endotoxin-free cells as an expression host for Cas9 protein production. Cas9 is an endonuclease enzyme that plays a key role in the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) and CRISPR associated protein 9 (CRISPR/Cas9) genome editing technique. This technology is very promising for use in diagnostics as well as treatment of diseases, especially for genetic diseases such as thalassemia. The potential uses for this technology thus generate a considerable interest for Cas9 utilization as a therapeutic protein in clinical treatment. Therefore, special care in protein production should be a major concern. Accordingly, we expressed the Cas9 protein in endotoxin-free bacterial cells achieving 99% purity with activity comparable to commercially available Cas9. Our protocol therefore yields a cost-effective product suitable for invitro experiments with stem cells.


Subject(s)
CRISPR-Associated Protein 9 , CRISPR-Cas Systems , Humans , Endotoxins/genetics , Gene Editing/methods , Repressor Proteins
4.
Biochem Biophys Res Commun ; 668: 111-117, 2023 08 06.
Article in English | MEDLINE | ID: mdl-37245291

ABSTRACT

Lysostaphin endopeptidase cleaves pentaglycine cross-bridges found in staphylococcal cell-wall peptidoglycans and proves very effective in combatting methicillin-resistant Staphylococcus aureus. Here, we revealed the functional importance of two loop residues, Tyr270 in loop 1 and Asn372 in loop 4, which are highly conserved among the M23 endopeptidase family and are found close to the Zn2+-coordinating active site. Detailed analyses of the binding groove architecture together with protein-ligand docking showed that these two loop residues potentially interact with the docked ligand-pentaglycine. Ala-substituted mutants (Y270A and N372A) were generated and over-expressed in Escherichia coli as a soluble form at levels comparable to the wild type. A drastic decrease in staphylolytic activity against S. aureus was observed for both mutants, suggesting an essential role of the two loop residues in lysostaphin function. Further substitutions with an uncharged polar Gln side-chain revealed that only the Y270Q mutation caused a dramatic reduction in bioactivity. In silico predicting the effect of binding site mutations revealed that all mutations displayed a large ΔΔGbind value, signifying requirements of the two loop residues for efficient binding to pentaglycine. Additionally, MD simulations revealed that Y270A and Y270Q mutations induced large flexibility of the loop 1 region, showing markedly increased RMSF values. Further structural analysis suggested that Tyr270 conceivably participated in the oxyanion stabilization of the enzyme catalysis. Altogether, our present study disclosed that two highly conserved loop residues, loop 1-Tyr270 and loop 4-Asn372, located near the lysostaphin active site are crucially involved in staphylolytic activity toward binding and catalysis of pentaglycine cross-links.


Subject(s)
Lysostaphin , Methicillin-Resistant Staphylococcus aureus , Lysostaphin/chemistry , Lysostaphin/metabolism , Lysostaphin/pharmacology , Staphylococcus aureus , Catalytic Domain , Ligands , Endopeptidases/genetics , Endopeptidases/metabolism , Catalysis
5.
Protein Pept Lett ; 30(5): 439-447, 2023.
Article in English | MEDLINE | ID: mdl-37076471

ABSTRACT

BACKGROUND: Glutathionylation is a protein post-translational modification triggered by oxidative stress. The susceptible proteins are modified by the addition of glutathione to specific cysteine residues. Virus infection also induces oxidative stress in the cell, which affects cellular homeostasis. It is not just the cellular proteins but the viral proteins that can also be modified by glutathionylation events, thereby impacting the function of the viral proteins. OBJECTIVES: This study was conducted to identify the effects of modification by glutathionylation on the guanylyltransferase activity of NS5 and identify the cysteine residues modified for the three flavivirus NS5 proteins. METHODS: The capping domain of NS5 proteins from 3 flaviviruses was cloned and expressed as recombinant proteins. A gel-based assay for guanylyltransferase activity was performed using a GTP analog labeled with the fluorescent dye Cy5 as substrate. The protein modification by glutathionylation was induced by GSSG and evaluated by western blot. The reactive cysteine residues were identified by mass spectrometry. RESULTS: It was found that the three flavivirus proteins behaved in a similar fashion with increasing glutathionylation yielding decreased guanylyltransferase activity. The three proteins also possessed conserved cysteines and they appeared to be modified for all three proteins. CONCLUSION: The glutathionylation appeared to induce conformational changes that affect enzyme activity. The conformational changes might also create binding sites for host cell protein interactions at later stages of viral propagation with the glutathionylation event, thereby serving as a switch for function change.


Subject(s)
Dengue Virus , Encephalitis Virus, Japanese , Flavivirus , Viral Nonstructural Proteins , Zika Virus , Cysteine , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism
6.
Toxins (Basel) ; 13(8)2021 08 09.
Article in English | MEDLINE | ID: mdl-34437424

ABSTRACT

In addition to the receptor-binding domain (DII), the C-terminal domain (DIII) of three-domain Cry insecticidal δ-endotoxins from Bacillus thuringiensis has been implicated in target insect specificity, yet its precise mechanistic role remains unclear. Here, the 21 kDa high-purity isolated DIII fragment derived from the Cry4Ba mosquito-specific toxin was achieved via optimized preparative FPLC, allowing direct rendering analyses for binding characteristics toward its target receptor-Aedes aegypti membrane-bound alkaline phosphatase (Aa-mALP). Binding analysis via dotblotting revealed that the Cry4Ba-DIII truncate was capable of specific binding to nitrocellulose-bound Aa-mALP, with a binding signal comparable to its 65 kDa Cry4Ba-R203Q full-length toxin. Further determination of binding affinity via sandwich ELISA revealed that Cry4Ba-DIII exhibited a rather weak binding to Aa-mALP with a dissociation constant (Kd) of ≈1.1 × 10-7 M as compared with the full-length toxin. Intermolecular docking between the Cry4Ba-R203Q active toxin and Aa-mALP suggested that four Cry4Ba-DIII residues, i.e., Glu522, Asn552, Asn576, and Leu615, are potentially involved in such toxin-receptor interactions. Ala substitutions of each residue (E522A, N552A, N576A and L615A) revealed that only the L615A mutant displayed a drastic decrease in biotoxicity against A. aegypti larvae. Additional binding analysis revealed that the L615A-impaired toxin also exhibited a reduction in binding capability to the surface-immobilized Aa-mALP receptor, while two bio-inactive DII-mutant toxins, Y332A and F364A, which almost entirely lost their biotoxicity, apparently retained a higher degree of binding activity. Altogether, our data disclose a functional importance of the C-terminal domain of Cry4Ba for serving as a potential receptor-binding moiety in which DIII-Leu615 could conceivably be exploited for the binding to Aa-mALP, highlighting its contribution to toxin interactions with such a target receptor in mediating larval toxicity.


Subject(s)
Alkaline Phosphatase/metabolism , Bacillus thuringiensis Toxins/metabolism , Endotoxins/metabolism , Hemolysin Proteins/metabolism , Insect Proteins/metabolism , Aedes , Animals , Bacillus thuringiensis Toxins/genetics , Endotoxins/genetics , Escherichia coli/genetics , Hemolysin Proteins/genetics , Insect Control , Insecticides , Molecular Docking Simulation , Pest Control, Biological , Protein Domains
7.
Protein Pept Lett ; 28(6): 643-650, 2021.
Article in English | MEDLINE | ID: mdl-33183185

ABSTRACT

BACKGROUND: Gastric pathogen Helicobacter pylori secretes VacA cytotoxin displaying a high degree of polymorphic variations of which the highest VacA pathogenicity correlates with m1-type variant followed by VacA-m2. OBJECTIVE: To comparatively evaluate expression in Escherichia coli of the mature VacA variants (m1- and m2-types) and their 33- and 55/59-kDa domains fused with His(6) tag at N- or C-terminus. METHODS: All VacA clones expressed in E. coli TOP10™ were analyzed by SDS-PAGE and Western blotting. VacA inclusions were solubilized under native conditions (~150-rpm shaking at 37°C for 2 h in 20 mM HEPES (pH7.4) and 150 mM NaCl). Membrane-perturbing and cytotoxic activities of solubilized VacA proteins were assessed via liposome-entrapped dye leakage and resazurin- based cell viability assays, respectively. VacA binding to human gastric adenocarcinoma cells was assessed by immunofluorescence microscopy. Side-chain hydrophobicity of VacA was analyzed through modeled structures constructed by homology- and ab initio-based modeling. RESULTS: Both full-length VacA-m1 and 33-kDa domain were efficiently expressed only in the presence of N-terminal extension while its 55-kDa domain was capably expressed with either N- or Cterminal extension. Selectively enhanced expression was also observed for VacA-m2. Protein expression profiles revealed a critical period in IPTG-induced production of the 55-kDa domain with N-terminal extension unlike its C-terminal extension showing relatively stable expression. Both VacA- m1 isolated domains were able to independently bind to cultured gastric cells similar to the full- length toxin, albeit the 33-kDa domain exhibited significantly higher activity of membrane perturbation than others. Membrane-perturbing and cytotoxic activities observed for VacA-m1 appeared to be higher than those of VacA-m2. Homology-based modeling and sequence analysis suggested a potential structural impact of non-polar residues located at the N-terminus of the mature VacA toxin and its 33-kDa domain. CONCLUSION: Our data provide molecular insights into selective influence of the N-terminally added tag on efficient expression of recombinant VacA variants, signifying biochemical and biological implications of the hydrophobic stretch within the N-terminal domain.


Subject(s)
Bacterial Proteins , Recombinant Fusion Proteins , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/pharmacology , Cell Line, Tumor , Cell Membrane/metabolism , Cell Survival/drug effects , Escherichia coli/genetics , Humans , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Stomach/cytology
8.
Biochem Biophys Res Commun ; 514(2): 365-371, 2019 06 25.
Article in English | MEDLINE | ID: mdl-31040022

ABSTRACT

Although the TlyA hemolysin from Helicobacter pylori has been implicated as a potential virulence factor involved in mediating host cell colonization and hence disease progression, its structural determinants underlying the biological activity are still largely uncertain. In this study, an important role of the formation of a particular disulfide bond for functional oligomeric assembly of the H. pylori TlyA toxin was evidently elucidated. The 27-kDa TlyA recombinant protein was overexpressed in Escherichia coli, subsequently purified to near homogeneity by cation exchange chromatography, and proven to be hemolytically active against sheep erythrocytes. Additionally, TlyA-induced hemolytic activity was significantly diminished under conditions of disulfide bond reduction with a thiol-reducing agent, dithiothreitol. When the purified TlyA protein was subjected to modified SDS-PAGE under non-reducing conditions, the presence of an oligomeric state of this protein was clearly revealed by its apparent molecular mass of ∼48 kDa. Recombinant E. coli cells expressing TlyA also displayed contact-dependent hemolysis of erythrocytes, suggesting TlyA localization at the bacterial outer membrane and thus supporting the formation of disulfide-bonded TlyA. Homology-based modeling and in silico structural assembly analysis of TlyA signified potential intermolecular, rather than intramolecular, disulfide bonding through Cys124 and Cys128. Subsequently, single substitution of either of these Cys residues with Ser severely affected the oligomeric assembly of both TlyA mutants and hence abolished their hemolytic activity. Altogether, our present data provide pivotal evidence that the formation of intermolecular disulfide bonding between Cys124 and Cys128 plays a critical role in structural assembly of a biologically active-TlyA oligomer.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cysteine/metabolism , Disulfides/metabolism , Helicobacter pylori , Hemolysis , Virulence Factors/chemistry , Virulence Factors/metabolism , Animals , Bacterial Proteins/analysis , Bacterial Proteins/genetics , Computer Simulation , Cysteine/chemistry , Disulfides/chemistry , Electrophoresis, Polyacrylamide Gel , Erythrocytes/pathology , Escherichia coli , Helicobacter pylori/chemistry , Helicobacter pylori/metabolism , Mutation , Oxidation-Reduction , Sheep , Virulence Factors/analysis , Virulence Factors/genetics
9.
Toxins (Basel) ; 11(2)2019 01 23.
Article in English | MEDLINE | ID: mdl-30678087

ABSTRACT

Although the C-terminal domain (DIII) of three-domain Cry insecticidal toxins from Bacillus thuringiensis has been implicated in various biological functions, its exact role still remains to be elucidated. Here, the 21-kDa isolated DIII fragment of the 65-kDa Cry4Ba mosquito-specific toxin was analyzed for its binding characteristics toward lipid-bilayer membranes. When the highly-purified Cry4Ba-DIII protein was structurally verified by attenuated total reflection Fourier transform infrared (ATR-FTIR) spectroscopy, it revealed the presence of a distinct ß-sheet structure, corresponding to its structure embodied in the Cry4Ba crystal structure. Binding analysis via surface plasmon resonance (SPR) spectroscopy revealed that the 21-kDa Cry4Ba-DIII truncate displayed tight binding to immobilized liposome membranes in a two-step manner, exhibiting a dissociation rate constant (kd) comparable to the 65-kDa full-length toxin. Also similar to the Cry4Ba full-length toxin, its isolated DIII truncate was able to anchor a part of its molecule into the immobilized membrane as the SPR signal was still detected after prolonged treatment with proteinase K. However, unlike the full-length active toxin, the DIII truncate was unable to induce membrane permeability of calcein-loaded liposomes or ion-channel formation in planar lipid bilayers. Together, our present data have disclosed a pivotal role of C-terminal DIII in serving as a membrane anchor rather than a pore-forming moiety of the Cry4Ba mosquito-active toxin, highlighting its potential mechanistic contribution to the interaction of the full-length toxin with lipid membranes in mediating toxicity.


Subject(s)
Bacterial Proteins , Endotoxins , Hemolysin Proteins , Animals , Bacillus thuringiensis Toxins , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Membrane/metabolism , Culicidae , Endotoxins/chemistry , Endotoxins/genetics , Endotoxins/metabolism , Hemolysin Proteins/chemistry , Hemolysin Proteins/genetics , Hemolysin Proteins/metabolism , Lipid Bilayers/metabolism , Protein Domains
10.
Parasit Vectors ; 11(1): 515, 2018 Sep 20.
Article in English | MEDLINE | ID: mdl-30236155

ABSTRACT

BACKGROUND: Mosquitoes transmit many vector-borne infectious diseases including malaria, dengue, chikungunya, yellow fever, filariasis, and Japanese encephalitis. The insecticidal δ-endotoxins Cry4, Cry11, and Cyt produced from Bacillus thuringiensis have been used for bio-control of mosquito larvae. Cry δ-endotoxins are synthesised as inactive protoxins in the form of crystalline inclusions in which they are processed to active toxins in larval midgut lumen. Previously, we demonstrated that the activated Cry4Ba toxin has to alter the permeability of the peritrophic membrane (PM), allowing toxin passage across PM to reach specific receptors on microvilli of larval midgut epithelial cells, where the toxin undergoes conformational changes, followed by membrane insertion and pore formation, resulting in larval death. A peritrophic membrane (PM)-binding calcofluor has been proposed to inhibit chitin formation and enhance baculovirus infection of lepidopteran Trichoplusia ni. METHODS: In this study, Aedes aegypti larvae were fed with the calcofluor and Cry4Ba toxin to investigate the effect of this agent on the toxicity of the Cry4Ba toxin. RESULTS: Calcofluor displayed an enhancing effect when co-fed with the Cry4Ba wild-type toxin. The agent could restore the killing activity of the partially active Cry4Ba mutant E417A/Y455A toward Ae. aegypti larvae. PM destruction was observed after larval challenge with calcofluor together with the toxin. Interestingly, calcofluor increased Cry4Ba toxin susceptibility toward semi-susceptible Culex quinquefasciatus larvae. However, calcofluor alone or in combination with the toxin showed no mortality effect on non-susceptible fresh-water fleas, Moina macrocopa. CONCLUSIONS: Our results suggest that PM may contribute to the resistance of the mosquito larvae to Cry4Ba toxin. The PM-permeability alternating calcofluor might be a promising candidate for enhancing insect susceptibility, which will consequently improve Cry4Ba efficacy in field settings in the future.


Subject(s)
Aedes/drug effects , Bacillus thuringiensis/genetics , Bacterial Proteins/pharmacology , Benzenesulfonates/pharmacology , Endotoxins/pharmacology , Hemolysin Proteins/pharmacology , Insecticides/pharmacology , Aedes/ultrastructure , Animals , Bacillus thuringiensis Toxins , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Endotoxins/genetics , Endotoxins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Hemolysin Proteins/genetics , Hemolysin Proteins/metabolism , Insecticides/metabolism , Larva/drug effects , Larva/ultrastructure , Membranes/drug effects , Membranes/metabolism , Membranes/ultrastructure , Mosquito Vectors/drug effects , Mutation , Permeability/drug effects , Pest Control, Biological , Siphonaptera/drug effects
11.
Protein Expr Purif ; 151: 106-112, 2018 11.
Article in English | MEDLINE | ID: mdl-29944958

ABSTRACT

Lysostaphin, a bacteriolytic toxin from Staphylococcus simulans, is a Zn2+-dependent endopeptidase that cleaves pentaglycine cross-bridges found in peptidoglycan of certain Staphylococci. Here, we have investigated a critical influence of Zn2+ ions on lysostaphin-induced bioactivity. Initially, we succeeded in producing a large amount with high purity of the 28-kDa His-tagged mature lysostaphin via soluble expression in Escherichia coli and subsequent purification via immobilized-Ni2+ affinity chromatography (IMAC). The purified monomeric bacteriocin exhibited concentration-dependent bioactivity against S. aureus and its methicillin-resistant strain through cell-wall hydrolysis rather than membrane perturbation. Following pre-incubation of the purified lysostaphin with exogenous Zn2+, a marked inhibition in staphylolytic activity was observed. When the pre-mixture was exposed to 1,10-phenanthroline (PNT, a Zn2+-chelator), the adverse effect of the exogenous Zn2+ on bioactivity was greatly decreased. Conversely, lysostaphin pre-treated with excess PNT retained relatively high bioactivity, indicating ineffective chelation of PNT to detach the catalytic Zn2+ from the active-site pocket. Structural analysis of the lysostaphin-catalytic domain together with amino acid sequence alignments of lysostaphin-like endopeptidases revealed a potential extraneous Zn2+-binding site found in close proximity to the Zn2+-coordinating active site. Overall our results provide more insights into an adverse influence of exogenous Zn2+ ions on staphylolytic activity of the purified Zn2+-dependent endopeptidase lysostaphin, implicating the presence of an extraneous inhibitory metal-binding site.


Subject(s)
Anti-Bacterial Agents/isolation & purification , Lysostaphin/isolation & purification , Staphylococcus/enzymology , Anti-Bacterial Agents/biosynthesis , Anti-Bacterial Agents/pharmacology , Biocatalysis , Cations, Divalent , Cell Wall/metabolism , Chelating Agents/chemistry , Chelating Agents/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Drug Resistance, Bacterial , Escherichia coli/genetics , Escherichia coli/metabolism , Lysostaphin/biosynthesis , Lysostaphin/pharmacology , Phenanthrolines/chemistry , Phenanthrolines/pharmacology , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , Staphylococcus aureus/drug effects , Zinc/chemistry , Zinc/pharmacology
12.
Curr Microbiol ; 75(2): 223-230, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29032467

ABSTRACT

The membrane perturbing action of the VacA toxin from Helicobacter pylori is responsible for vacuole formation in intracellular compartments and the induction of apoptosis. The VacA toxin contains 2 major domains, p33 and p55, which are involved in receptor binding and membrane pore formation, respectively. Improved methodologies for VacA purification and assays are urgently needed for further detailed investigations on the mechanism of action of this significant virulence factor. We found that by fusing mouse DHFR with the N-terminus of the full-length (p88) VacA toxin, expression levels in recombinant E. coli were substantially increased when compared to the conventional (His)6-tagged protein. The DHFR-VacA fusion protein was active in sulforhodamine dye-release assays using liposomes at acidic pH in a concentration-dependent manner. Enzymatic activity of DHFR in the fusion protein was comparable to a commercial reference sample of purified DHFR; however, activity was insensitive to inhibition by methotrexate. Our findings suggest that the VacA p88 toxin with a modified N-terminus still maintains its capability for membrane insertion and that pH-dependent conformational changes occur during interaction of VacA with membranes.


Subject(s)
Bacterial Proteins/metabolism , Coloring Agents/metabolism , Liposomes/metabolism , Recombinant Fusion Proteins/metabolism , Tetrahydrofolate Dehydrogenase/metabolism , Animals , Bacterial Proteins/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Membranes/metabolism , Mice , Recombinant Fusion Proteins/genetics , Tetrahydrofolate Dehydrogenase/genetics
13.
Biochem Biophys Res Commun ; 485(4): 720-724, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28238785

ABSTRACT

Proteolytic degradation of the ∼100-kDa isolated RTX (Repeat-in-ToXin) subdomain (CyaA-RTX) of the Bordetella pertussis CyaA-hemolysin (CyaA-Hly) was evidently detected upon solely-prolonged incubation. Here, a truncated CyaA-Hly fragment (CyaA-HP/BI) containing hydrophobic and acylation regions connected with the first RTX block (BI1015-1088) was constructed as a putative precursor for investigating its potential autocatalysis. The 70-kDa His-tagged CyaA-HP/BI fragment which was over-expressed in Escherichia coli as insoluble aggregate was entirely solubilized with 4 M urea. After re-naturation in a Ni2+-NTA affinity column, the purified-refolded CyaA-HP/BI fragment in HEPES buffer (pH 7.4) supplemented with 2 mM CaCl2 was completely degraded upon incubation at 37 °C for 3 h. Addition of 1,10-phenanthroline‒an inhibitor of Zn2+-dependent metalloproteases markedly reduced the extent of degradation for CyaA-HP/BI and CyaA-RTX, but the degradative effect was clearly enhanced by addition of 100 mM ZnCl2. Structural analysis of a plausible CyaA-HP/BI model revealed a potential Zn2+-binding His-Asp cluster located between the acylation region and RTX-BI1015-1088. Moreover, Arg997‒one of the identified cleavage sites of the CyaA-RTX fragment was located in close proximity to the Zn2+-binding catalytic site. Overall results demonstrated for the first time that the observed proteolysis of CyaA-HP/BI and CyaA-RTX fragments is conceivably due to their Zn2+-dependent autocatalytic activity.


Subject(s)
Adenylate Cyclase Toxin/metabolism , Bacterial Proteins/metabolism , Bordetella pertussis/metabolism , Hemolysin Proteins/metabolism , Zinc/metabolism , Adenylate Cyclase Toxin/chemistry , Adenylate Cyclase Toxin/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Binding Sites/genetics , Biocatalysis/drug effects , Blotting, Western , Bordetella pertussis/genetics , Escherichia coli/genetics , Hemolysin Proteins/chemistry , Hemolysin Proteins/genetics , Models, Molecular , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phenanthrolines/pharmacology , Protein Domains , Protein Precursors/chemistry , Protein Precursors/genetics , Protein Precursors/metabolism , Proteolysis/drug effects , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Zinc/chemistry , Zinc/pharmacology
14.
J Biol Chem ; 290(34): 20793-20803, 2015 Aug 21.
Article in English | MEDLINE | ID: mdl-26112409

ABSTRACT

The insecticidal feature of the three-domain Cry δ-endotoxins from Bacillus thuringiensis is generally attributed to their capability to form oligomeric pores, causing lysis of target larval midgut cells. However, the molecular description of their oligomerization process has not been clearly defined. Here a stable prepore of the 65-kDa trypsin-activated Cry4Ba mosquito-specific toxin was established through membrane-mimetic environments by forming an ∼200-kDa octyl-ß-D-glucoside micelle-induced trimer. The SDS-resistant trimer caused cytolysis to Sf9 insect cells expressing Aedes-mALP (a Cry4Ba receptor) and was more effective than a toxin monomer in membrane perturbation of calcein-loaded liposomes. A three-dimensional model of toxin trimer obtained by negative-stain EM in combination with single-particle reconstruction at ∼5 nm resolution showed a propeller-shaped structure with 3-fold symmetry. Fitting the three-dimensional reconstructed EM map with a 100-ns molecular dynamics-simulated Cry4Ba structure interacting with an octyl-ß-D-glucoside micelle showed relative positioning of individual domains in the context of the trimeric complex with a major protrusion from the pore-forming domain. Moreover, high-speed atomic force microscopy imaging at nanometer resolution and a subsecond frame rate demonstrated conformational transitions from a propeller-like to a globularly shaped trimer upon lipid membrane interactions, implying prepore-to-pore conversion. Real-time trimeric arrangement of monomers associated with L-α-dimyristoylphosphatidylcholine/3-[(3-cholamidopropyl)dimethylammonio]-2-hydroxy-1-propanesulfonic acid bicelle membranes was also envisaged by successive high-speed atomic force microscopy imaging, depicting interactions among three individual subunits toward trimer formation. Together, our data provide the first pivotal insights into the structural requirement of membrane-induced conformational changes of Cry4Ba toxin monomers for the molecular assembly of a prepore trimer capable of inserting into target membranes to generate a lytic pore.


Subject(s)
Bacillus thuringiensis/chemistry , Bacterial Proteins/chemistry , Bacterial Toxins/chemistry , Endotoxins/chemistry , Hemolysin Proteins/chemistry , Insecticides/chemistry , Sf9 Cells/drug effects , Aedes/cytology , Aedes/drug effects , Animals , Bacillus thuringiensis/metabolism , Bacillus thuringiensis Toxins , Bacterial Proteins/genetics , Bacterial Proteins/toxicity , Bacterial Toxins/genetics , Bacterial Toxins/toxicity , Dimyristoylphosphatidylcholine/chemistry , Endotoxins/genetics , Endotoxins/toxicity , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Glucosides/chemistry , Hemolysin Proteins/genetics , Hemolysin Proteins/toxicity , Insect Proteins/genetics , Insect Proteins/metabolism , Insecticides/metabolism , Insecticides/toxicity , Liposomes/chemistry , Micelles , Molecular Dynamics Simulation , Pest Control, Biological , Protein Conformation , Protein Multimerization , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/toxicity , Sf9 Cells/cytology , Spodoptera/cytology , Spodoptera/drug effects , Sulfonic Acids/chemistry
15.
Biochem Biophys Res Commun ; 450(2): 948-52, 2014 Jul 25.
Article in English | MEDLINE | ID: mdl-24971536

ABSTRACT

Bacillus thuringiensis Cry4Aa toxin was previously shown to be much more toxic to Culex mosquito-larvae than its closely related toxin - Cry4Ba, conceivably due to their sequence differences within the ß10-ß11 receptor-binding loop. Here, single-Ala substitutions of five residues (Pro(510), Thr(512), Tyr(513), Lys(514) and Thr(515)) within the Cry4Aa ß10-ß11 loop revealed that only Lys(514) corresponding to the relative position of Cry4Ba-Asp(454) is crucial for toxicity against Culex quinquefasciatus larvae. Interestingly, charge-reversal mutations at Cry4Ba-Asp(454) (D454R and D454K) revealed a marked increase in toxicity against such less-susceptible larvae. In situ binding analyses revealed that both Cry4Ba-D454R and D454K mutants exhibited a significant increase in binding to apical microvilli of Culex larval midguts, albeit at lower-binding activity when compared with Cry4Aa. Altogether, our present data suggest that a positively charged side-chain near the tip of the ß10-ß11 loop plays a critical role in determining target specificity of Cry4Aa against Culex spp., and hence a great increase in the Culex larval toxicity of Cry4Ba was obtained toward an opposite-charge conversion of the corresponding Asp(454).


Subject(s)
Bacillus thuringiensis/metabolism , Bacterial Proteins/genetics , Culex/microbiology , Endotoxins/genetics , Hemolysin Proteins/genetics , Amino Acid Sequence , Animals , Bacillus thuringiensis Toxins , Bacterial Proteins/metabolism , Bacterial Proteins/pharmacology , Culex/drug effects , Endotoxins/metabolism , Endotoxins/pharmacology , Hemolysin Proteins/metabolism , Hemolysin Proteins/pharmacology , Larva/drug effects , Larva/microbiology , Molecular Sequence Data , Mosquito Control , Mutation , Protein Binding , Protein Structure, Secondary , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology
16.
Biochem Biophys Res Commun ; 407(4): 708-13, 2011 Apr 22.
Article in English | MEDLINE | ID: mdl-21439264

ABSTRACT

Bacillus thuringiensis Cry4Ba toxin selectively kills Aedes aegypti mosquito larvae as it is in part due to the presence of specific membrane-bound protein receptors. In this study, using data mining approach, we initially identified three potential glycosylphosphatidylinositol-linked aminopeptidase N (GPI-APN) isoforms, APN2778, APN2783 and APN5808, which are believed to act as Cry4Ba toxin receptors. These three isoforms that are functionally expressed in the larval midgut can be sequence-specific knocked down (ranging from ∼80 % to 95 %) by soaking the Aedes aegypti larvae in buffer of long double-stranded GPI-APN RNAs (∼300-680 bp). Finally, to see the physiological effect of APN knockdowns, the larvae were fed with Escherichia coli expressing Cry4Ba toxin. The results revealed that all the three identified GPI-APN isoforms may possibly function as a Cry4Ba receptor, particularly for APN2783 as those larvae with this transcript knockdown showed a dramatic increase in resistance to Cry4Ba toxicity.


Subject(s)
Aedes/drug effects , Aedes/genetics , Bacterial Proteins/toxicity , CD13 Antigens/genetics , Drug Resistance/genetics , Endotoxins/toxicity , Hemolysin Proteins/toxicity , Insecticides/toxicity , Aedes/enzymology , Amino Acid Sequence , Animals , Bacillus thuringiensis Toxins , Gene Knockdown Techniques , Glycosylphosphatidylinositols/metabolism , Isoenzymes/genetics , Larva/drug effects , Larva/enzymology , Larva/genetics , Molecular Sequence Data , RNA Interference , RNA, Messenger/antagonists & inhibitors , Sequence Analysis, Protein , Transcription, Genetic
17.
Article in English | MEDLINE | ID: mdl-20516610

ABSTRACT

To obtain a complete structure of the Bacillus thuringiensis Cry4Ba mosquito-larvicidal protein, a 65 kDa functional form of the Cry4Ba-R203Q mutant toxin was generated for crystallization by eliminating the tryptic cleavage site at Arg203. The 65 kDa trypsin-resistant fragment was purified and crystallized using the sitting-drop vapour-diffusion method. The crystals belonged to the rhombohedral space group R32, with unit-cell parameters a = b = 184.62, c = 187.36 A. Diffraction data were collected to at least 2.07 A resolution using synchrotron radiation and gave a data set with an overall R(merge) of 9.1% and a completeness of 99.9%. Preliminary analysis indicated that the asymmetric unit contained one molecule of the active full-length mutant, with a V(M) coefficient and solvent content of 4.33 A(3) Da(-1) and 71%, respectively.


Subject(s)
Bacillus thuringiensis/chemistry , Bacterial Proteins/chemistry , Endotoxins/chemistry , Hemolysin Proteins/chemistry , Bacillus thuringiensis Toxins , Bacterial Proteins/genetics , Crystallization , Crystallography, X-Ray , Endotoxins/genetics , Hemolysin Proteins/genetics , Molecular Weight , Mutation
18.
J Biochem Mol Biol ; 40(1): 58-64, 2007 Jan 31.
Article in English | MEDLINE | ID: mdl-17244483

ABSTRACT

Similar to the other known structures of Bacillus thuringiensis Cry delta-endotoxins, the crystal structure of the 65-kDa activated Cry4Ba toxin comprises three domains which are, from the N- to C-terminus, a bundle of alpha-helices, a three-beta-sheet domain, and a beta-sandwich. To investigate the properties of the C-terminal domain III in isolation from the rest of the toxin, the cloned Cry4Ba-domain III was over-expressed as a 21-kDa soluble protein in Escherichia coli, which cross-reacted with anti-Cry4Ba domain III monoclonal antibody. A highly-purified domain III was obtained in a monomeric form by ion-exchange and size-exclusion FPLC. Circular dichroism spectroscopy indicated that the isolated domain III fragment distinctly exists as a beta-sheet structure, corresponding to the domain III structure embodied in the Cry4Ba crystal structure. In vitro binding analysis via immuno-histochemical assay revealed that the Cry4Ba-domain III protein was able to bind to the apical microvilli of the susceptible Stegomyia aegypti larval midguts, albeit at lower-binding activity when compared with the full-length active toxin. These results demonstrate for the first time that the C-terminal domain III of the Cry4Ba mosquito-larvicidal protein, which can be isolated as a native folded monomer, conceivably participates in toxin-receptor recognition.


Subject(s)
Bacillus thuringiensis/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Cloning, Molecular , Endotoxins/genetics , Endotoxins/metabolism , Escherichia coli/metabolism , Hemolysin Proteins/genetics , Hemolysin Proteins/metabolism , Aedes , Animals , Bacillus thuringiensis Toxins , Bacterial Proteins/chemistry , Bacterial Proteins/isolation & purification , Bacterial Toxins/chemistry , Bacterial Toxins/isolation & purification , Crystallography, X-Ray , Endotoxins/chemistry , Endotoxins/isolation & purification , Hemolysin Proteins/chemistry , Hemolysin Proteins/isolation & purification , Models, Molecular , Protein Structure, Tertiary , Recombinant Proteins/metabolism , Solubility
SELECTION OF CITATIONS
SEARCH DETAIL
...