Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.826
Filter
1.
J Appl Microbiol ; 135(9)2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39227165

ABSTRACT

AIMS: This study identifies a unique glutathione S-transferase (GST) in extremophiles using genome, phylogeny, bioinformatics, functional characterization, and RNA sequencing analysis. METHODS AND RESULTS: Five putative GSTs (H0647, H0729, H1478, H3557, and H3594) were identified in Halothece sp. PCC7418. Phylogenetic analysis suggested that H0647, H1478, H0729, H3557, and H3594 are distinct GST classes. Of these, H0729 was classified as an iota-class GST, encoding a high molecular mass GST protein with remarkable features. The protein secondary structure of H0729 revealed the presence of a glutaredoxin (Grx) Cys-Pro-Tyr-Cys (C-P-Y-C) motif that overlaps with the N-terminal domain and harbors a topology similar to the thioredoxin (Trx) fold. Interestingly, recombinant H0729 exhibited a high catalytic efficiency for both glutathione (GSH) and 1-chloro-2, 4-dinitrobenzene (CDNB), with catalytic efficiencies that were 155- and 32-fold higher, respectively, compared to recombinant H3557. Lastly, the Halothece gene expression profiles suggested that antioxidant and phase II detoxification encoding genes are crucial in response to salt stress. CONCLUSION: Iota-class GST was identified in cyanobacteria. This GST exhibited a high catalytic efficiency toward xenobiotic substrates. Our findings shed light on a diversified evolution of GST in cyanobacteria and provide functional dynamics of the genes encoding the enzymatic antioxidant and detoxification systems under abiotic stresses.


Subject(s)
Cyanobacteria , Glutathione Transferase , Phylogeny , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Glutathione Transferase/chemistry , Cyanobacteria/genetics , Cyanobacteria/enzymology , Cyanobacteria/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Glutathione/metabolism , Amino Acid Sequence , Glutaredoxins/genetics , Glutaredoxins/metabolism , Glutaredoxins/chemistry
2.
J Agric Food Chem ; 72(36): 19680-19688, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39225316

ABSTRACT

Spodoptera litura is a significant agricultural pest, and its glutathione S-transferase (GST) plays a crucial role in insecticide resistance. This study aimed to investigate the relationship between the SlGSTe11 gene of S. litura and resistance to cyantraniliprole and nicotine. Transcriptome analysis revealed that SlGSTe11 is highly expressed mainly in fat bodies, with a significant increase in SlGSTe11 gene expression under induction by cyantraniliprole and nicotine. The ectopic expression of the SlGSTe11 gene in transgenic fruit flies resulted in a 5.22-fold increase in the tolerance to cyantraniliprole. Moreover, compared to the UAS-SlGSTe11 line, the Act5C-UAS>SlGSTe11 line laid more eggs and had a lower mortality after nicotine exposure. RNAi-mediated inhibition of SlGSTe11 gene expression led to a significant increase in the mortality of S. litura under cyantraniliprole exposure. In vitro metabolism experiments demonstrated that the recombinant SlGSTe11 protein efficiently metabolizes cyantraniliprole. Molecular docking results indicated that SlGSTe11 has a strong affinity for both cyantraniliprole and nicotine. These findings suggest that SlGSTe11 is involved in the development of resistance to cyantraniliprole and nicotine in S. litura.


Subject(s)
Fat Body , Glutathione Transferase , Insect Proteins , Insecticide Resistance , Insecticides , Nicotine , Pyrazoles , Spodoptera , ortho-Aminobenzoates , Animals , Spodoptera/genetics , Spodoptera/drug effects , Spodoptera/metabolism , Spodoptera/enzymology , Spodoptera/growth & development , Insecticides/pharmacology , Insecticides/metabolism , Insecticides/chemistry , Insect Proteins/genetics , Insect Proteins/metabolism , Insect Proteins/chemistry , ortho-Aminobenzoates/metabolism , ortho-Aminobenzoates/pharmacology , Pyrazoles/pharmacology , Nicotine/metabolism , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Glutathione Transferase/chemistry , Insecticide Resistance/genetics , Fat Body/metabolism , Fat Body/enzymology , Fat Body/drug effects , Molecular Docking Simulation
3.
Ecotoxicol Environ Saf ; 282: 116729, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39024945

ABSTRACT

Global agricultural production is significantly hampered by insect pests, and the demand for natural pragmatic pesticides with environmental concern remains unfulfilled. Ageratina adenophora (Spreng.) also known as Crofton weed, is an invasive perennial herbaceous plant that is known to possess multiple bioactive compounds. In our study, two isomers of ageraphorone metabolites i.e, 10 Hα-9-oxo-ageraphorone (10HA) and 10 Hß-9-oxo-ageraphorone (10HB), were identified from Crofton weed, exhibiting potent antifeedant and larvicidal activities against Plutella xylostella. For antifeedant activity, the median effective concentration (EC50) values for 10HA and 10HB in the choice method were 2279 mg/L and 3233 mg/L, respectively, and for the no choice method, EC50 values were 1721 mg/L and 2394 mg/L, respectively. For larvicidal activity, lethal concentration (LC50) values for 10HA and 10HB were 2421 mg/L and 4109 mg/L at 48 h and 2101 mg/L and 3550 mg/L at 72 h. Furthermore, both in- vivo and in-vitro studies revealed that the isomers 10HA and 10HB exhibited potent detoxifying enzymes inhibition activity such as carboxylesterase and glutathione S-transferases. Molecular docking and MD simulation analysis provide insight into the possible interaction between isomers of ageraphorone metabolites and Carboxylic Ester Hydrolase protein (Gene: pxCCE016b) of P. xylostella, which led to a finding that CarEH protein plays a significant role in the detoxification of the two compounds in P. xylostella. Finally, our findings show that the primary enzymes undergoing inhibition by isomers of ageraphorone metabolites, causing toxicity in insects, are Carboxylesterase and glutathione S-transferase.


Subject(s)
Ageratina , Moths , Sesquiterpenes , Ageratina/chemistry , Moths/drug effects , Animals , Insecticides/chemistry , Insecticides/pharmacology , Plant Extracts/chemistry , Plant Extracts/pharmacology , Larva/drug effects , Molecular Docking Simulation , Binding Sites , Molecular Conformation , Isomerism , Sesquiterpenes/chemistry , Sesquiterpenes/pharmacology , Esterases/chemistry , Esterases/metabolism , Glutathione Transferase/chemistry , Glutathione Transferase/metabolism , Feeding Behavior/drug effects
4.
Biomolecules ; 14(7)2024 Jun 26.
Article in English | MEDLINE | ID: mdl-39062473

ABSTRACT

Glutathione transferase (GST) is a superfamily of ubiquitous enzymes, multigenic in numerous organisms and which generally present homodimeric structures. GSTs are involved in numerous biological functions such as chemical detoxification as well as chemoperception in mammals and insects. GSTs catalyze the conjugation of their cofactor, reduced glutathione (GSH), to xenobiotic electrophilic centers. To achieve this catalytic function, GSTs are comprised of a ligand binding site and a GSH binding site per subunit, which is very specific and highly conserved; the hydrophobic substrate binding site enables the binding of diverse substrates. In this work, we focus our interest in a model organism, the fruit fly Drosophila melanogaster (D. mel), which comprises 42 GST sequences distributed in six classes and composing its GSTome. The goal of this study is to describe the complete structural GSTome of D. mel to determine how changes in the amino acid sequence modify the structural characteristics of GST, particularly in the GSH binding sites and in the dimerization interface. First, we predicted the 3D atomic structures of each GST using the AlphaFold (AF) program and compared them with X-ray crystallography structures, when they exist. We also characterized and compared their global and local folds. Second, we used multiple sequence alignment coupled with AF-predicted structures to characterize the relationship between the conservation of amino acids in the sequence and their structural features. Finally, we applied normal mode analysis to estimate thermal B-factors of all GST structures of D. mel. Particularly, we extracted flexibility profiles of GST and identify key residues and motifs that are systematically involved in the ligand binding/dimerization processes and thus playing a crucial role in the catalytic function. This methodology will be extended to guide the in silico design of synthetic GST with new/optimal catalytic properties for detoxification applications.


Subject(s)
Drosophila melanogaster , Glutathione Transferase , Animals , Drosophila melanogaster/enzymology , Glutathione Transferase/chemistry , Glutathione Transferase/metabolism , Glutathione Transferase/genetics , Binding Sites , Amino Acid Sequence , Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Models, Molecular , Crystallography, X-Ray , Glutathione/metabolism , Glutathione/chemistry , Protein Multimerization
5.
Biomolecules ; 14(7)2024 Jun 26.
Article in English | MEDLINE | ID: mdl-39062472

ABSTRACT

This study presents a comprehensive analysis of the dimerization interfaces of fly GSTs through sequence alignment. Our investigation revealed GSTE1 as a particularly intriguing target, providing valuable insights into the variations within Delta and Epsilon GST interfaces. The X-ray structure of GSTE1 was determined, unveiling remarkable thermal stability and a distinctive dimerization interface. Utilizing circular dichroism, we assessed the thermal stability of GSTE1 and other Drosophila GSTs with resolved X-ray structures. The subsequent examination of GST dimer stability correlated with the dimerization interface supported by findings from X-ray structural analysis and thermal stability measurements. Our discussion extends to the broader context of GST dimer interfaces, offering a generalized perspective on their stability. This research enhances our understanding of the structural and thermodynamic aspects of GST dimerization, contributing valuable insights to the field.


Subject(s)
Glutathione Transferase , Protein Multimerization , Thermodynamics , Animals , Glutathione Transferase/chemistry , Glutathione Transferase/metabolism , Glutathione Transferase/genetics , Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Crystallography, X-Ray , Drosophila melanogaster/enzymology , Models, Molecular , Amino Acid Sequence , Drosophila/enzymology
6.
Biosens Bioelectron ; 261: 116501, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38905858

ABSTRACT

A novel laccase mimic enzyme Cu-Mn with excellent photothermal properties was firstly prepared via a combination of hydrothermal and in situ synthesis. Cu-Mn nanozymes could catalyze the typical laccase substrate 2,4-dichlorophenol (2,4-DP) to generate the red quinone imine. Further, loading the MnO2 nanosheets with photothermal properties, Cu-Mn nanozymes possessed not only excellent laccase catalytic activity, but also high photothermal conversion efficiency. The presence of glutathione S-transferase (GST) recovered the glutathione (GSH)-induced weakness of the laccase activity and photothermal properties of Cu-Mn. Hence, a GST enzyme-regulated dual-mode sensing strategy was established based on Cu-Mn nanozymes. The detection limits of GST monitoring based on colorimetric and photothermal methods were 0.092 and 0.087 U/L with response times of 20 min and 8 min, respectively. Furthermore, the proposed method enabled the measuring of GST levels in human serum and was successfully employed in the primary evaluation of hepatitis patients. Another attraction, the impressive photothermal behavior also endowed the Cu-Mn nanozymes with promising antimicrobial properties, which exhibited significant antimicrobial effects against Escherichia coli (E.coli) and Staphylococcus aureus (S.aureus). Unsurprisingly, multifunctional Cu-Mn nanozymes certainly explore new paths in biochemical analysis and antimicrobial applications.


Subject(s)
Anti-Bacterial Agents , Biosensing Techniques , Copper , Escherichia coli , Glutathione Transferase , Laccase , Staphylococcus aureus , Laccase/chemistry , Humans , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Glutathione Transferase/chemistry , Escherichia coli/drug effects , Staphylococcus aureus/drug effects , Copper/chemistry , Copper/pharmacology , Catalysis , Oxidation-Reduction , Limit of Detection , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Chlorophenols/pharmacology , Chlorophenols/chemistry , Colorimetry/methods , Oxides/chemistry , Manganese Compounds/chemistry , Manganese Compounds/pharmacology , Nanostructures/chemistry
7.
Nanoscale ; 16(31): 14748-14756, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-38921728

ABSTRACT

Targeted drug delivery using metal-organic frameworks (MOFs) has shown significant progress. However, the tumor microenvironment (TME) impedes efficient MOF particle transfer into tumor cells. To tackle this issue, we pre-coated nano-sized MOF-808 particles with multifunctional proteins: glutathione S-transferase (GST)-affibody (Afb) and collagenase, aiming to navigate the TME more effectively. The surface of MOF-808 particles is coated with GST-Afb-a fusion protein of GST and human epidermal growth factor receptor 2 (HER2) Afb or epidermal growth factor receptor (EGFR) Afb which has target affinity. We also added collagenase enzymes capable of breaking down collagen in the extracellular matrix (ECM) through supramolecular conjugation, all without chemical modification. By stabilizing these proteins on the surface, GST-Afb mitigate biomolecule absorption, facilitating specific tumor cell targeting. Simultaneously, collagenase degrades the ECM in the TME, enabling deep tissue penetration of MOF particles. Our resulting system, termed collagenase-GST-Afb-MOF-808 (Col-Afb-M808), minimizes undesired interactions between MOF particles and external biological proteins. It not only induces cell death through Afb-mediated cell-specific targeting, but also showcases advanced cellular internalization in 3D multicellular spheroid cancer models, with effective deep tissue penetration. The therapeutic efficacy of Col-Afb-M808 was further assessed via in vivo imaging and evaluation of tumor inhibition following injection of IR-780 loaded Col-Afb-M808 in 4T1tumor-bearing nude mice. This study offers key insights into the regulation of the multifunctional protein-adhesive surface of MOF particles, paving the way for the designing even more effective targeted drug delivery systems with nano-sized MOF particles.


Subject(s)
Metal-Organic Frameworks , Metal-Organic Frameworks/chemistry , Humans , Animals , Mice , Cell Line, Tumor , Glutathione Transferase/metabolism , Glutathione Transferase/chemistry , Collagenases/chemistry , Collagenases/metabolism , Female , Receptor, ErbB-2/metabolism , ErbB Receptors/metabolism , Mice, Nude , Drug Delivery Systems , Tumor Microenvironment/drug effects , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Mice, Inbred BALB C
8.
Parasites Hosts Dis ; 62(2): 205-216, 2024 May.
Article in English | MEDLINE | ID: mdl-38835261

ABSTRACT

Sigma-class glutathione transferase (GST) proteins with dual GST and prostaglandin synthase (PGS) activities play a crucial role in the establishment of Clonorchis sinensis infection. Herein, we analyzed the structural and enzymatic properties of sigma-class GST (CsGST-σ) proteins to obtain insight into their antioxidant and immunomodulatory functions in comparison with mu-class GST (CsGST-µ) proteins. CsGST-σ proteins conserved characteristic structures, which had been described in mammalian hematopoietic prostaglandin D2 synthases. Recombinant forms of these CsGST-σ and CsGST-µ proteins expressed in Escherichia coli exhibited considerable degrees of GST and PGS activities with substantially different specific activities. All recombinant proteins displayed higher affinities toward prostaglandin H2 (PGS substrate; average Km of 30.7 and 3.0 µm for prostaglandin D2 [PGDS] and E2 synthase [PGES], respectively) than those toward CDNB (GST substrate; average Km of 1,205.1 µm). Furthermore, the catalytic efficiency (Kcat/Km) of the PGDS/PGES activity was higher than that of GST activity (average Kcat/Km of 3.1, 0.7, and 7.0×10-3 s-1µm-1 for PGDS, PGES, and GST, respectively). Our data strongly suggest that the C. sinensis sigma- and mu-class GST proteins are deeply involved in regulating host immune responses by generating PGD2 and PGE2 in addition to their roles in general detoxification.


Subject(s)
Clonorchis sinensis , Glutathione Transferase , Intramolecular Oxidoreductases , Glutathione Transferase/metabolism , Glutathione Transferase/chemistry , Glutathione Transferase/genetics , Clonorchis sinensis/enzymology , Clonorchis sinensis/genetics , Animals , Intramolecular Oxidoreductases/metabolism , Intramolecular Oxidoreductases/chemistry , Intramolecular Oxidoreductases/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/chemistry , Lipocalins/metabolism , Lipocalins/genetics , Lipocalins/chemistry , Lipocalins/immunology , Escherichia coli/genetics , Prostaglandin H2/metabolism , Prostaglandin H2/chemistry , Kinetics
9.
J Agric Food Chem ; 72(22): 12489-12497, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38773677

ABSTRACT

The glutathione S-transferases (GSTs) are important detoxifying enzymes in insects. Our previous studies found that the susceptibility of Chilo suppressalis to abamectin was significantly increased when the CsGST activity was inhibited by glutathione (GSH) depletory. In this study, the potential detoxification mechanisms of CsGSTs to abamectin were explored. Six CsGSTs of C. suppressalis were expressed in vitro. Enzymatic kinetic parameters including Km and Vmax of recombinant CsGSTs were determined, and results showed that all of the six CsGSTs were catalytically active and displaying glutathione transferase activity. Insecticide inhibitions revealed that a low concentration of abamectin could effectively inhibit the activities of CsGSTs including CsGSTd1, CsGSTe4, CsGSTo2, CsGSTs3, and CsGSTu1. However, the in vitro metabolism assay found that the six CsGSTs could not metabolize abamectin directly. Additionally, the glutathione transferase activity of CsGSTs in C. suppressalis was significantly increased post-treatment with abamectin. Comprehensive analysis of the results in present and our previous studies demonstrated that CsGSTs play an important role in detoxification of abamectin by catalyzing the conjugation of GSH to abamectin in C. suppressalis, and the high binding affinities of CsGSTd1, CsGSTe4, CsGSTo2, CsGSTs3, and CsGSTu1 with abamectin might also suggest the involvement of CsGSTs in detoxification of abamectin via the noncatalytic passive binding and sequestration instead of direct metabolism. These studies are helpful to better understand the detoxification mechanisms of GSTs in insects.


Subject(s)
Glutathione Transferase , Insect Proteins , Insecticides , Ivermectin , Moths , Glutathione Transferase/metabolism , Glutathione Transferase/genetics , Glutathione Transferase/chemistry , Animals , Insecticides/metabolism , Insecticides/pharmacology , Insecticides/chemistry , Moths/metabolism , Moths/drug effects , Moths/enzymology , Ivermectin/analogs & derivatives , Ivermectin/metabolism , Ivermectin/pharmacology , Ivermectin/chemistry , Insect Proteins/metabolism , Insect Proteins/genetics , Insect Proteins/chemistry , Kinetics , Oryza/metabolism , Oryza/parasitology , Oryza/chemistry , Glutathione/metabolism , Glutathione/chemistry
10.
Protein J ; 43(3): 613-626, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38743189

ABSTRACT

Glutathione-S-transferase enzymes (GSTs) are essential components of the phase II detoxification system and protect organisms from oxidative stress induced by xenobiotics and harmful toxins such as 1-chloro-2,4-dinitrobenzene (CDNB). In Tetrahymena thermophila, the TtGSTm34 gene was previously reported to be one of the most responsive GST genes to CDNB treatment (LD50 = 0.079 mM). This study aimed to determine the kinetic features of recombinantly expressed and purified TtGSTm34 with CDNB and glutathione (GSH). TtGSTm34-8xHis was recombinantly produced in T. thermophila as a 25-kDa protein after the cloning of the 660-bp full-length ORF of TtGSTm34 into the pIGF-1 vector. A three-dimensional model of the TtGSTm34 protein constructed by the AlphaFold and PyMOL programs confirmed that it has structurally conserved and folded GST domains. The recombinant production of TtGSTm34-8xHis was confirmed by SDS‒PAGE and Western blot analysis. A dual-affinity chromatography strategy helped to purify TtGSTm34-8xHis approximately 3166-fold. The purified recombinant TtGSTm34-8xHis exhibited significantly high enzyme activity with CDNB (190 µmol/min/mg) as substrate. Enzyme kinetic analysis revealed Km values of 0.68 mM with GSH and 0.40 mM with CDNB as substrates, confirming its expected high affinity for CDNB. The optimum pH and temperature were determined to be 7.0 and 25 °C, respectively. Ethacrynic acid inhibited fully TtGSTm34-8xHis enzyme activity. These results imply that TtGSTm34 of T. thermophila plays a major role in the detoxification of xenobiotics, such as CDNB, as a first line of defense in aquatic protists against oxidative damage.


Subject(s)
Cloning, Molecular , Glutathione Transferase , Protozoan Proteins , Recombinant Proteins , Tetrahymena thermophila , Glutathione Transferase/genetics , Glutathione Transferase/chemistry , Glutathione Transferase/metabolism , Tetrahymena thermophila/enzymology , Tetrahymena thermophila/genetics , Recombinant Proteins/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Kinetics , Dinitrochlorobenzene/chemistry , Dinitrochlorobenzene/metabolism , Gene Expression , Glutathione/metabolism , Glutathione/chemistry
11.
Biochem Biophys Res Commun ; 715: 150008, 2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38685186

ABSTRACT

In the last decade, much attention was given to the study of physiological amyloid fibrils. These structures include A-bodies, which are the nucleolar fibrillar formations that appear in the response to acidosis and heat shock, and disassemble after the end of stress. One of the proteins involved in the biogenesis of A-bodies, regardless of the type of stress, is Von-Hippel Lindau protein (VHL). Known also as a tumor suppressor, VHL is capable to form amyloid fibrils both in vitro and in vivo in response to the environment acidification. As with most amyloidogenic proteins fusion with various tags is used to increase the solubility of VHL. Here, we first performed AFM-study of fibrils formed by VHL protein and by VHL fused with GST-tag (GST-VHL) at acidic conditions. It was shown that formed by full-length VHL fibrils are short heterogenic structures with persistent length of 2400 nm and average contour length of 409 nm. GST-tag catalyzes VHL amyloid fibril formation, superimpose chirality, increases length and level of hierarchy, but decreases rigidity of amyloid fibrils. The obtained data indicate that tagging can significantly affect the fibrillogenesis of the target protein.


Subject(s)
Amyloid , Glutathione Transferase , Von Hippel-Lindau Tumor Suppressor Protein , Amyloid/metabolism , Amyloid/chemistry , Glutathione Transferase/metabolism , Glutathione Transferase/chemistry , Humans , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/chemistry , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Microscopy, Atomic Force
12.
Mikrochim Acta ; 191(5): 282, 2024 04 23.
Article in English | MEDLINE | ID: mdl-38652326

ABSTRACT

A novel dual-mode fluorometric and colorimetric sensing platform is reported for determining glutathione S-transferase (GST) by utilizing polyethyleneimine-capped silver nanoclusters (PEI-AgNCs) and cobalt-manganese oxide nanosheets (CoMn-ONSs) with oxidase-like activity. Abundant active oxygen species (O2•-) can be produced through the CoMn-ONSs interacting with dissolved oxygen. Afterward, the pink oxDPD was generated through the oxidation of colorless N,N-diethyl-p-phenylenediamine (DPD) by O2•-, and two absorption peaks at 510 and 551 nm could be observed. Simultaneously, oxDPD could quench the fluorescence of PEI-AgNCs at 504 nm via the inner filter effect (IFE). However, in the presence of glutathione (GSH), GSH prevents the oxidation of DPD due to the reducibility of GSH, leading to the absorbance decrease at 510 and 551 nm. Furthermore, the fluorescence at 504 nm was restored due to the quenching effect of oxDPD on decreased PEI-AgNCs. Under the catalysis of GST, GSH and1-chloro-2,4-dinitrobenzo (CDNB) conjugate to generate an adduct, initiating the occurrence of the oxidation of the chromogenic substrate DPD, thereby inducing a distinct colorimetric response again and the significant quenching of PEI-AgNCs. The detection limits for GST determination were 0.04 and 0.21 U/L for fluorometric and colorimetric modes, respectively. The sensing platform illustrated reliable applicability in detecting GST in real samples.


Subject(s)
Cobalt , Colorimetry , Glutathione Transferase , Manganese Compounds , Metal Nanoparticles , Oxides , Polyethyleneimine , Silver , Polyethyleneimine/chemistry , Silver/chemistry , Cobalt/chemistry , Oxides/chemistry , Manganese Compounds/chemistry , Metal Nanoparticles/chemistry , Colorimetry/methods , Glutathione Transferase/metabolism , Glutathione Transferase/chemistry , Limit of Detection , Oxidoreductases/chemistry , Oxidoreductases/metabolism , Humans , Glutathione/chemistry , Oxidation-Reduction , Biosensing Techniques/methods , Phenylenediamines/chemistry , Nanostructures/chemistry
13.
J Biochem ; 176(1): 69-80, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38471515

ABSTRACT

Schistosoma japonicum glutathione-S-transferase (SjGST), the so-called GST-tag, is one of the most widely used protein tags for the purification of recombinant proteins by affinity chromatography. Attachment of SjGST enables the purification of a protein of interest (POI) using commercially available glutathione-immobilizing resins. Here we produced an SjGST mutant pair that forms heterodimers by adjusting the salt bridge pairs in the homodimer interface of SjGST. An MD study confirmed that the SjGST mutant pair did not disrupt the heterodimer formation. The modified SjGST protein pair coexpressed in Escherichia coli was purified by glutathione-immobilized resin. The stability of the heterodimeric form of the SjGST mutant pair was further confirmed by size exclusion chromatography. Surface plasmon resonance measurements unveiled the selective formation of heterodimers within the pair, accompanied by a significant suppression of homodimerization. The heterodimeric SjGST exhibited enzymatic activity in assays employing a commercially available fluorescent substrate. By fusing one member of the heterodimeric SjGST pair with a fluorescent protein and the other with the POI, we were able to conveniently and sensitively detect protein-protein interactions using fluorescence spectroscopy in the pull-down assays. Thus, utilization of the heterodimeric SjGST would be a useful tag for protein science.


Subject(s)
Chromatography, Affinity , Glutathione Transferase , Schistosoma japonicum , Schistosoma japonicum/enzymology , Glutathione Transferase/metabolism , Glutathione Transferase/chemistry , Glutathione Transferase/isolation & purification , Glutathione Transferase/genetics , Animals , Chromatography, Affinity/methods , Protein Multimerization , Helminth Proteins/metabolism , Helminth Proteins/chemistry , Helminth Proteins/genetics , Helminth Proteins/isolation & purification , Recombinant Proteins/metabolism , Recombinant Proteins/isolation & purification , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Models, Molecular
14.
FEBS Open Bio ; 14(5): 726-739, 2024 May.
Article in English | MEDLINE | ID: mdl-38514457

ABSTRACT

Taenia solium can cause human taeniasis and/or cysticercosis. The latter can in some instances cause human neurocysticercosis which is considered a priority in disease-control strategies and the prevention of mental health problems. Glutathione transferases are crucial for the establishment and long-term survival of T. solium; therefore, we structurally analyzed the 24-kDa glutathione transferase gene (Ts24gst) of T. solium and biochemically characterized its product. The gene promoter showed potential binding sites for transcription factors and xenobiotic regulatory elements. The gene consists of a transcription start site, four exons split by three introns, and a polyadenylation site. The gene architecture is conserved in cestodes. Recombinant Ts24GST (rTs24GST) was active and dimeric. Anti-rTs24GST serum showed slight cross-reactivity with human sigma-class GST. A 3D model of Ts24GST enabled identification of putative residues involved in interactions of the G-site with GSH and of the H-site with CDNB and prostaglandin D2. Furthermore, rTs24GST showed optimal activity at 45 °C and pH 9, as well as high structural stability in a wide range of temperatures and pHs. These results contribute to the better understanding of this parasite and the efforts directed to fight taeniasis/cysticercosis.


Subject(s)
Glutathione Transferase , Taenia solium , Taenia solium/genetics , Taenia solium/enzymology , Animals , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Glutathione Transferase/chemistry , Humans , Models, Molecular , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/chemistry , Promoter Regions, Genetic/genetics
15.
J Struct Biol ; 216(2): 108089, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38537893

ABSTRACT

Fusion proteins (FPs) are frequently utilized as a biotechnological tool in the determination of macromolecular structures using X-ray methods. Here, we explore the use of different protein tags in various FP, to obtain initial phases by using them in a partial molecular replacement (MR) and constructing the remaining FP structure with ARP/wARP. Usually, the tag is removed prior to crystallization, however leaving the tag on may facilitate crystal formation, and structural determination by expanding phases from known to unknown segments of the complex. In this study, the Protein Data Bank was mined for an up-to-date list of FPs with the most used protein tags, Maltose Binding Protein (MBP), Green Fluorescent Protein (GFP), Thioredoxin (TRX), Glutathione transferase (GST) and the Small Ubiquitin-like Modifier Protein (SUMO). Partial MR using the protein tag, followed by automatic model building, was tested on a subset of 116 FP. The efficiency of this method was analyzed and factors that influence the coordinate construction of a substantial portions of the fused protein were identified. Using MBP, GFP, and SUMO as phase generators it was possible to build at least 75 % of the protein of interest in 36 of the 116 cases tested. Our results reveal that tag selection has a significant impact; tags with greater structural stability, such as GFP, increase the success rate. Further statistical analysis identifies that resolution, Wilson B factor, solvent percentage, completeness, multiplicity, protein tag percentage in the FP (considering amino acids), and the linker length play pivotal roles using our approach. In cases where a structural homologous is absent, this method merits inclusion in the toolkit of protein crystallographers.


Subject(s)
Green Fluorescent Proteins , Maltose-Binding Proteins , Recombinant Fusion Proteins , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Green Fluorescent Proteins/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/chemistry , Maltose-Binding Proteins/genetics , Maltose-Binding Proteins/chemistry , Maltose-Binding Proteins/metabolism , Crystallography, X-Ray/methods , Glutathione Transferase/genetics , Glutathione Transferase/chemistry , Glutathione Transferase/metabolism , Thioredoxins/chemistry , Thioredoxins/genetics , Thioredoxins/metabolism , Models, Molecular , Databases, Protein , Crystallization/methods , Protein Conformation
16.
J Biol Chem ; 300(4): 107123, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38417796

ABSTRACT

Thiram is a toxic fungicide extensively used for the management of pathogens in fruits. Although it is known that thiram degrades in plant tissues, the key enzymes involved in this process remain unexplored. In this study, we report that a tau class glutathione S-transferase (GST) from Carica papaya can degrade thiram. This enzyme was easily obtained by heterologous expression in Escherichia coli, showed low promiscuity toward other thiuram disulfides, and catalyzed thiram degradation under physiological reaction conditions. Site-directed mutagenesis indicated that G-site residue S67 shows a key influence for the enzymatic activity toward thiram, while mutation of residue S13, which reduced the GSH oxidase activity, did not significantly affect the thiram-degrading activity. The formation of dimethyl dithiocarbamate, which was subsequently converted into carbon disulfide, and dimethyl dithiocarbamoylsulfenic acid as the thiram degradation products suggested that thiram undergoes an alkaline hydrolysis that involves the rupture of the disulfide bond. Application of the GST selective inhibitor 4-chloro-7-nitro-2,1,3-benzoxadiazole reduced papaya peel thiram-degrading activity by 95%, indicating that this is the main degradation route of thiram in papaya. GST from Carica papaya also catalyzed the degradation of the fungicides chlorothalonil and thiabendazole, with residue S67 showing again a key influence for the enzymatic activity. These results fill an important knowledge gap in understanding the catalytic promiscuity of plant GSTs and reveal new insights into the fate and degradation products of thiram in fruits.


Subject(s)
Carica , Glutathione Transferase , Thiram , Carica/enzymology , Carica/genetics , Fungicides, Industrial/metabolism , Glutathione Transferase/metabolism , Glutathione Transferase/genetics , Glutathione Transferase/chemistry , Mutagenesis, Site-Directed , Plant Proteins/chemistry , Plant Proteins/genetics , Plant Proteins/metabolism , Thiram/metabolism , Escherichia coli/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
17.
Biosci Biotechnol Biochem ; 87(10): 1183-1192, 2023 Sep 21.
Article in English | MEDLINE | ID: mdl-37403406

ABSTRACT

Phenylpropanone monomers, including guaiacyl hydroxypropanone, are important precursors for the synthesis of various chemicals. The monomers are obtained in a three-step cascade reaction catalyzed by a group of enzymes in the ß-etherase system that cleaves the ß-O-4 bond, the major bond in lignin. In this study, one of the ß-etherase of the glutathione-S-transferase superfamily, AbLigF2, was discovered in genus Altererythrobacter, and the recombinant etherase was characterized. The enzyme showed maximal activity at 45 °C, maintained 30% of its activity after 2 h at 50 °C, and was the most thermostable among the previously reported enzymes. Moreover, N13, S14, and S115, located near the thiol group of glutathione, had a significant effect on the maximum reaction rate of enzyme activity. This study suggests that AbLigF2 has the potential to serve as a thermostable enzyme for lignin utilization and provides insights into its catalytic mechanism.


Subject(s)
Lignin , Oxidoreductases , Lignin/chemistry , Catalysis , Glutathione Transferase/chemistry , Glutathione
18.
Article in English | MEDLINE | ID: mdl-37150092

ABSTRACT

Glutathione S-transferases (GSTs) are major enzymes in detoxification phase II, and have been functioned in resistance to various insecticides or oxidative stress. Herein, we selected the non-biting midge, Propsilocerus akamusi, widespread in Asian aquatic ecosystems, to uncover the gene location, structure, and phylogenetics relationship of GSTs at genome scale first time. Thirty-three cytosolic and four microsomal GST genes were identified and located on the four chromosomes. The cytosolic GSTs involved in the eight subclasses and five GST genes were unclassified. The expansion of GST genes in P. akamusi experienced duplication events on the delta, theta, xi, iota, and unclassified subclasses. The RNA-Seq analyses and RT-qPCR validation showed that the expression of PaGSTt2 gene is significantly elevated, with deltamethrin concentration increasing. The tertiary structure of PaGSTt2 enzyme was reconstructed, which was different from the other theta gene in the active site. In addition, the GST genes of six chironomids were first described based on the assembled genomes to explore the difference of those in the adaptation to kinds of environments. The GST frame for P. akmusi and its expression profiles provide valuable resources to understand their role in insecticide resistance of this species, as well as those of other biting midges.


Subject(s)
Ceratopogonidae , Chironomidae , Animals , Glutathione Transferase/chemistry , Chironomidae/genetics , Chironomidae/metabolism , Ceratopogonidae/genetics , Ceratopogonidae/metabolism , Ecosystem , Genome-Wide Association Study , Phylogeny , Gene Expression Profiling
19.
J Mol Graph Model ; 122: 108457, 2023 07.
Article in English | MEDLINE | ID: mdl-37004419

ABSTRACT

Schistosoma glutathione transferases (GSTs) have been identified as attractive drug targets for the design of novel antischistosomals. Here, we used in silico methods to validate the discriminative inhibitory properties of bromosulfophthalein (BSP) against the 26-kDa GST from S. japonicum (Sj26GST), and the 28-kDa GST from S. haematobium (Sh28GST), versus human GST (hGST) isoforms alpha (hGSTA), mu (hGSTM) and pi (hGSTP). The use of BSP as an archetypal selective inhibitor was harnessed to produce molecular dynamics-derived pharmacophores of the two targets. Pharmacophore-based screening using a large dataset of experimental and approved drug compounds was performed to produce a shortlist of candidates. The top candidate for each target was prioritised via molecular docking, yielding guanosine-3'-monophosphate-5'-diphosphate (G3D) for Sj26GST, and quercetin-3'-O-phosphate (Q3P) for Sh28GST. Comparative molecular dynamics studies of both candidates compared to BSP showed similar characteristics of binding stability and strength, suggesting their potential to emulate the inhibitory effects of BSP.


Subject(s)
Molecular Dynamics Simulation , Sulfobromophthalein , Animals , Humans , Molecular Docking Simulation , Pharmacophore , Schistosoma/metabolism , Glutathione Transferase/chemistry , Glutathione Transferase/metabolism , Glutathione/metabolism
20.
Mol Biochem Parasitol ; 254: 111559, 2023 06.
Article in English | MEDLINE | ID: mdl-37024059

ABSTRACT

Glutathione transferase enzymes (GSTs) are believed to be a major detoxification system in helminth parasites and have been associated with immunomodulation of the host response. Echinococcus granulosus sensu lato (s.l.) is a cestode parasite known to express at least five different GSTs, but no Omega-class enzymes have been reported in this parasite or in any other cestode. Herein we report the identification of a new member of the GST superfamily in E. granulosus s.l., which is phylogenetically related to the Omega-class: EgrGSTO. Through mass spectrometry, we showed that the 237 amino acids protein EgrGSTO is expressed by the parasite. Moreover, we identified homologues of EgrGSTO in other eight members of the Taeniidae family, including E. canadensis, E. multilocularis, E. oligarthrus, Hydatigera taeniaeformis, Taenia asiatica, T. multiceps, T. saginata and T. solium. A manual sequence inspection and rational modification yielded eight Taeniidae's GSTO sequences, each one encoding for a 237 aa polypeptide showing 80.2% overall identity. To the best of our knowledge, this is the first description of genes encoding for Omega-class GSTs in worms belonging to the Taeniidae family -that at least in E. granulosus s.l. is expressed as a protein- suggesting the gene encodes for a functional protein.


Subject(s)
Echinococcus granulosus , Parasites , Taenia , Animals , Glutathione Transferase/chemistry , Echinococcus granulosus/genetics , Parasites/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL