Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 153
Filter
Add more filters










Publication year range
1.
Arch Biochem Biophys ; 754: 109962, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38499055

ABSTRACT

Acetohydroxyacid synthase (AHAS) is one of the key enzymes of the biosynthesis of branched-chain amino acids, it is also an effective target for the screening of herbicides and antibiotics. In this study we present a method for preparing Escherichia coli AHAS I holoenzyme (EcAHAS I) with exceptional stability, which provides a solid ground for us to re-investigate the in vitro catalytic properties of the protein. The results show EcAHAS I synthesized in this way exhibits similar function to Bacillus subtilis acetolactate synthase in its catalysis with pyruvate and 2-ketobutyrate (2-KB) as dual-substrate, producing four 2-hydroxy-3-ketoacids including (S)-2-acetolactate, (S)-2-aceto-2-hydroxybutyrate, (S)-2-propionyllactate, and (S)-2-propionyl-2-hydroxybutyrate. Quantification of the reaction indicates that the two substrates almost totally consume, and compound (S)-2-aceto-2- hydroxybutyrate forms in the highest yield among the four major products. Moreover, the protein also condenses two molecules of 2-KB to furnish (S)-2-propionyl-2-hydroxybutyrate. Further exploration manifests that EcAHAS I ligates pyruvate/2-KB and nitrosobenzene to generate two arylhydroxamic acids N-hydroxy-N-phenylacetamide and N-hydroxy-N-phenyl- propionamide. These findings enhance our comprehension of the catalytic characteristics of EcAHAS I. Furthermore, the application of this enzyme as a catalyst in construction of C-N bonds displays promising potential.


Subject(s)
Acetolactate Synthase , Escherichia coli , Acetolactate Synthase/chemistry , Glycogen Synthase , Hydroxybutyrates , Pyruvates , Holoenzymes
2.
J Agric Food Chem ; 71(47): 18227-18238, 2023 Nov 29.
Article in English | MEDLINE | ID: mdl-37567224

ABSTRACT

Herbicides are effective tools to manage weeds and enable food production and sustainable agriculture. Corteva Agriscience R&D has recently discovered new diphenyl-ether compounds displaying excellent postemergent efficacy on important weed species along with corn safety. Here, we describe the chemistry, biology, biochemistry, and computational modeling research that led to the discovery and elucidation of the primary mode of action for these compounds. The target protein was found to be acetolactate synthase (ALS), a key enzyme in the biosynthesis of branched chain amino acids (valine, leucine, and isoleucine). While weed resistance evolution to ALS herbicides is widespread, the molecular interaction of the diphenyl-ether compounds at the active site of the ALS enzyme differs significantly from that of some commercial ALS inhibitors. The unique biochemical profile of these molecules along with their excellent herbicidal activity and corn selectivity make them a noteworthy development in the pursuit of novel, safe, and sustainable weed control solutions.


Subject(s)
Acetolactate Synthase , Herbicides , Herbicides/pharmacology , Herbicides/chemistry , Acetolactate Synthase/chemistry , Herbicide Resistance , Ethers
3.
J Mol Model ; 29(8): 241, 2023 Jul 12.
Article in English | MEDLINE | ID: mdl-37436478

ABSTRACT

CONTEXT: The design and synthesis of safe and highly active sulfonylurea herbicides is still a challenge. Therefore, following some principles of structure-activity relationship (SAR) of sulfonylurea herbicides, this work focuses on evaluating two sulfonylurea derivatives bearing electron-withdrawing substituents, namely, -(CO)OCH3 and -NO2 on the aryl group, on herbicidal activity. To understand the effects caused by the substituent groups, the molecular and electronic structures of the sulfonylureas were evaluated by density functional theory. Likewise, the crystalline supramolecular arrangements of both compounds were analyzed by Hirshfeld surface, QTAIM, and NBO, with the aim of verifying changes in intermolecular interactions caused by substituent groups. Finally, through a toxicophoric analysis, we were able to predict the interacting groups in their biological target, acetolactate synthase, and verify the interactions with the binding site. METHODS: All theoretical calculations were conducted using the highly parameterized empirical exchange-correlation functional M06-2X accompanied by the diffuse and polarized basis set 6-311++G(d,p). The atomic coordinates were obtained directly from the crystalline structures, and from the energies of the frontier molecular orbitals (HOMO and LUMO), chemical descriptors were obtained that indicated the influence of the functional groups in the sulfonylureas on the reactivity of the molecules. The intermolecular interactions in the crystals were analyzed using the Hirshfeld, QTAIM, and NBO surfaces. Toxicophoric modeling was performed by the PharmaGist webserver and molecular docking calculations were performed by the GOLD 2022.1.0 software package so that the ligand was fitted to the binding site in a 10 Å sphere. For this, genetic algorithm parameters were used using the ChemPLP scoring function for docking and ASP for redocking.


Subject(s)
Acetolactate Synthase , Herbicides , Molecular Docking Simulation , Models, Molecular , Acetolactate Synthase/chemistry , Acetolactate Synthase/metabolism , Herbicides/chemistry , Herbicides/pharmacology , Sulfonylurea Compounds/chemistry , Sulfonylurea Compounds/pharmacology , Pyrimidines
4.
J Agric Food Chem ; 71(13): 5117-5126, 2023 Apr 05.
Article in English | MEDLINE | ID: mdl-36943718

ABSTRACT

Amidosulfuron (AS) is from the commercial sulfonylurea herbicide family. It is highly effective against dicot broad-leaf weeds. This herbicide targets acetohydroxyacid synthase (AHAS), the first enzyme in the branched chain amino acid biosynthesis pathway. Here, we have determined the crystal structure of AS in complex with wildtype Arabidopsis thaliana AHAS (AtAHAS) and with the resistance mutant, S653T. In both structures, the cofactor, ThDP, is modified to a peracetate adduct, consistent with time-dependent accumulative inhibition. Compared to other AHAS-inhibiting herbicides of the sulfonylurea family, AS lacks a second aromatic ring. The replacement is an aryl sulfonyl group with a reduced number of interactions with the enzyme and relatively low affinity (Ki = 4.2 µM vs low nM when two heteroaromatic rings are present). This study shows that effective herbicides can have a relatively high Ki for plant AHAS but can still be a potent herbicide provided accumulative inhibition also occurs.


Subject(s)
Acetolactate Synthase , Arabidopsis , Herbicides , Arabidopsis/metabolism , Acetolactate Synthase/chemistry , Herbicides/chemistry , Sulfonylurea Compounds/chemistry , Herbicide Resistance
5.
Chem Biol Drug Des ; 100(4): 487-501, 2022 10.
Article in English | MEDLINE | ID: mdl-35792871

ABSTRACT

A series of 51 novel sulfonylurea compounds with ortho-alkoxy substituent at phenyl ring were chemically synthesized and spectroscopically characterized. The biological activities of the target compounds were evaluated using the enzyme inhibition against acetohydroxyacid synthase (AHAS; EC 2.2.1.6) from fungal or plant source, as well as cell-based antifungal assay and greenhouse pot herbicidal assay. Among the target compounds, 6e showed desirable antifungal activity against Candida albicans standard isolate sc5314 with minimum inhibition concentration (MIC) of 0.39 mg/L (0.98 µM) after 24 h, and 6a demonstrated promising pre-emergence herbicidal activity against Echinochloacrus-galli at 30 g/ha dosage. Representative compounds 6a, 6e, and 6i showed no cell cytotoxicity even at 40 mg/L concentration. Theoretical DFT calculations indicated HOMO maps should be considered to understand the structure-activity relationships. The present study has hence provided useful information for further discovery of novel antifungal agents or selective herbicides.


Subject(s)
Acetolactate Synthase , Herbicides , Acetolactate Synthase/chemistry , Acetolactate Synthase/metabolism , Alcohols , Antifungal Agents/chemistry , Antifungal Agents/pharmacology , Enzyme Inhibitors/chemistry , Herbicides/chemistry , Herbicides/pharmacology , Structure-Activity Relationship , Sulfonylurea Compounds/pharmacology
6.
J Mol Graph Model ; 116: 108242, 2022 11.
Article in English | MEDLINE | ID: mdl-35671569

ABSTRACT

Pyrimidine compounds comprise a class of acetohydroxyacid synthase (AHAS) inhibitors, thus possessing herbicidal activity. Due to the ongoing development of resistance by weeds to current herbicides, the design of new agrochemical candidates is often required. This work reports the proposition of unprecedented pyrimidines as herbicides guided by quantitative structure-activity relationship (QSAR) modeling. Multivariate image analysis (MIA) descriptors for 66 pyrimidine derivatives obtained from different sources were regressed against inhibitory activity data, and the resulting QSAR models were found to be reliable and predictive (r2 = 0.88 ± 0.07, q2 = 0.53 ± 0.06, and r2pred = 0.51 ± 0.10 in a bootstrapping experiment using electronegativity-based descriptors). The chemical features responsible for the herbicidal activities were analyzed through MIA contour maps that describe the substituent effects on the response variables, whereas the interaction between the proposed compounds and AHAS was analyzed through docking studies. From the proposed compounds, at least five pyrimidine derivatives exhibited promising performance as AHAS inhibitors compared to the known analogs.


Subject(s)
Acetolactate Synthase , Herbicides , Acetolactate Synthase/chemistry , Acetolactate Synthase/metabolism , Computer Simulation , Herbicides/chemistry , Herbicides/pharmacology , Pyrimidines/pharmacology , Quantitative Structure-Activity Relationship
7.
Genes (Basel) ; 12(11)2021 11 22.
Article in English | MEDLINE | ID: mdl-34828447

ABSTRACT

The sustainability of rice cropping systems is jeopardized by the large number and variety of populations of polyploid Echinochloa spp. resistant to ALS inhibitors. Better knowledge of the Echinochloa species present in Italian rice fields and the study of ALS genes involved in target-site resistance could significantly contribute to a better understanding of resistance evolution and management. Using a CAPS-rbcL molecular marker, two species, E. crus-galli (L.) P. Beauv. and E. oryzicola (Vasinger) Vasing., were identified as the most common species in rice in Italy. Mutations involved in ALS inhibitor resistance in the different species were identified and associated with the ALS homoeologs. The relative expression of the ALS gene copies was evaluated. Molecular characterization led to the identification of three ALS genes in E. crus-galli and two in E. oryzicola. The two species also carried different point mutations conferring resistance: Ala122Asn in E. crus-galli and Trp574Leu in E. oryzicola. Mutations were carried in the same gene copy (ALS1), which was significantly more expressed than the other copies (ALS2 and ALS3) in both species. These results explain the high resistance level of these populations and why mutations in the other ALS copies are not involved in herbicide resistance.


Subject(s)
Acetolactate Synthase/genetics , Echinochloa/genetics , Mutation , Plant Proteins/genetics , Acetolactate Synthase/antagonists & inhibitors , Acetolactate Synthase/chemistry , Acetolactate Synthase/metabolism , Binding Sites , Drug Resistance , Echinochloa/classification , Echinochloa/drug effects , Enzyme Inhibitors/pharmacology , Gene Dosage , Plant Proteins/metabolism , Protein Binding
8.
Sci Rep ; 11(1): 21055, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34702838

ABSTRACT

Aspergillus fumigatus is a fungal pathogen whose effects can be debilitating and potentially fatal in immunocompromised patients. Current drug treatment options for this infectious disease are limited to just a few choices (e.g. voriconazole and amphotericin B) and these themselves have limitations due to potentially adverse side effects. Furthermore, the likelihood of the development of resistance to these current drugs is ever present. Thus, new treatment options are needed for this infection. A new potential antifungal drug target is acetohydroxyacid synthase (AHAS; EC 2.2.1.6), the first enzyme in the branched chain amino acid biosynthesis pathway, and a target for many commercial herbicides. In this study, we have expressed, purified and characterised the catalytic subunit of AHAS from A. fumigatus and determined the inhibition constants for several known herbicides. The most potent of these, penoxsulam and metosulam, have Ki values of 1.8 ± 0.9 nM and 1.4 ± 0.2 nM, respectively. Molecular modelling shows that these compounds are likely to bind into the herbicide binding pocket in a mode similar to Candida albicans AHAS. We have also shown that these two compounds inhibit A. fumigatus growth at a concentration of 25 µg/mL. Thus, AHAS inhibitors are promising leads for the development of new anti-aspergillosis therapeutics.


Subject(s)
Acetolactate Synthase , Antifungal Agents/chemistry , Aspergillus fumigatus/enzymology , Fungal Proteins , Herbicides/chemistry , Pyrimidines/chemistry , Sulfonamides/chemistry , Triazoles/chemistry , Uridine/analogs & derivatives , Acetolactate Synthase/antagonists & inhibitors , Acetolactate Synthase/chemistry , Candida albicans/enzymology , Fungal Proteins/antagonists & inhibitors , Fungal Proteins/chemistry , Uridine/chemistry
9.
Nucleic Acids Res ; 49(W1): W578-W588, 2021 07 02.
Article in English | MEDLINE | ID: mdl-33999189

ABSTRACT

ProteoVision is a web server designed to explore protein structure and evolution through simultaneous visualization of multiple sequence alignments, topology diagrams and 3D structures. Starting with a multiple sequence alignment, ProteoVision computes conservation scores and a variety of physicochemical properties and simultaneously maps and visualizes alignments and other data on multiple levels of representation. The web server calculates and displays frequencies of amino acids. ProteoVision is optimized for ribosomal proteins but is applicable to analysis of any protein. ProteoVision handles internally generated and user uploaded alignments and connects them with a selected structure, found in the PDB or uploaded by the user. It can generate de novo topology diagrams from three-dimensional structures. All displayed data is interactive and can be saved in various formats as publication quality images or external datasets or PyMol Scripts. ProteoVision enables detailed study of protein fragments defined by Evolutionary Classification of protein Domains (ECOD) classification. ProteoVision is available at http://proteovision.chemistry.gatech.edu/.


Subject(s)
Ribosomal Proteins/chemistry , Software , Acetolactate Synthase/chemistry , Bacterial Proteins/chemistry , Internet , Models, Molecular , Peptide Elongation Factor Tu/chemistry , Protein Conformation , Sequence Alignment
10.
Arch Biochem Biophys ; 701: 108807, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33587902

ABSTRACT

The biosynthesis of R-phenylacetylcarbinol (R-PAC) by the acetohydroxy acid synthase, (AHAS) is addressed by molecular dynamics simulations (MD), hybrid quantum mechanics/molecular mechanics (QM/MM), and QM/MM free energy calculations. The results show the reaction starts with the nucleophilic attack of the C2α atom of the HEThDP intermediate on the Cß atom of the carbonyl group of benzaldehyde substrate via the formation of a transition state (TS1) with the HEThDP intermediate under 4'-aminopyrimidium (APH+) form. The calculated activation free energy for this step is 17.4kcal mol-1 at 27 °C. From this point, the reaction continues with the abstraction of Hß atom of the HEThDP intermediate by the Oß atom of benzaldehyde to form the intermediate I. The reaction is completed with the cleavage of the bond C2α-C2 to form the product R-PAC and to regenerate the ylide intermediate under the APH+ form, allowing in this way to reinitiate to the catalytic cycle once more. The calculated activation barrier for this last step is 15.9kcal mol-1 at 27 °C.


Subject(s)
Acetolactate Synthase/chemistry , Benzyl Alcohols/chemical synthesis , Molecular Dynamics Simulation , Benzyl Alcohols/chemistry , Quantum Theory
11.
J Agric Food Chem ; 69(1): 45-54, 2021 Jan 13.
Article in English | MEDLINE | ID: mdl-33372787

ABSTRACT

Herbicide safeners enhance herbicide detoxification in crops without reducing their herbicidal efficacy against target weeds. To alleviate maize injury caused by the sulfonylurea herbicide nicosulfuron, a series of 1,3-disubstituted imidazolidine or hexahydropyrimidine derivatives were rationally designed via bioisosterism and active subunit combinations. Thirty novel compounds were synthesized using an efficient one-pot method and low-cost raw materials and characterized by IR, 1H NMR, 13C NMR, and high-resolution mass spectrometer (HRMS). Bioactivity and structure-activity relationship (SAR) were evaluated for herbicide safeners tested against nicosulfuron injury. Most of the compounds effectively protected sensitive maize against nicosulfuron damage. The parent skeletons and substituents of the target compounds both substantially influenced their safener activity. Compound I-3 exhibited superior bioactivity compared to the safener isoxadifen-ethyl. Molecular docking simulations disclosed that compound I-3 competed with nicosulfuron for the acetolactate synthase active site and demonstrated that this is the protective mechanism of safeners. The target compound I-3 presented with strong herbicide safener activity in maize and is, therefore, a potential candidate for the development of a novel herbicide safener.


Subject(s)
Herbicides/toxicity , Protective Agents/chemical synthesis , Protective Agents/pharmacology , Acetolactate Synthase/chemistry , Acetolactate Synthase/metabolism , Drug Design , Herbicides/chemistry , Imidazolidines/chemistry , Molecular Docking Simulation , Plant Proteins/chemistry , Plant Proteins/metabolism , Protective Agents/chemistry , Pyridines/chemistry , Pyridines/toxicity , Pyrimidines/chemistry , Structure-Activity Relationship , Sulfonylurea Compounds/chemistry , Sulfonylurea Compounds/toxicity , Zea mays/chemistry , Zea mays/drug effects , Zea mays/enzymology
12.
Nat Commun ; 11(1): 4292, 2020 08 27.
Article in English | MEDLINE | ID: mdl-32855421

ABSTRACT

Cost competitive conversion of biomass-derived sugars into biofuel will require high yields, high volumetric productivities and high titers. Suitable production parameters are hard to achieve in cell-based systems because of the need to maintain life processes. As a result, next-generation biofuel production in engineered microbes has yet to match the stringent cost targets set by petroleum fuels. Removing the constraints imposed by having to maintain cell viability might facilitate improved production metrics. Here, we report a cell-free system in a bioreactor with continuous product removal that produces isobutanol from glucose at a maximum productivity of 4 g L-1 h-1, a titer of 275 g L-1 and 95% yield over the course of nearly 5 days. These production metrics exceed even the highly developed ethanol fermentation process. Our results suggest that moving beyond cells has the potential to expand what is possible for bio-based chemical production.


Subject(s)
Biochemistry/methods , Butanols/metabolism , Enzymes/metabolism , Acetolactate Synthase/chemistry , Acetolactate Synthase/metabolism , Adenosine Triphosphate , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Biochemistry/instrumentation , Bioreactors , Cell-Free System , Directed Molecular Evolution , Enzymes/chemistry , Enzymes/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Glucose/metabolism , Temperature , Thermodynamics
13.
Nature ; 586(7828): 317-321, 2020 10.
Article in English | MEDLINE | ID: mdl-32640464

ABSTRACT

Acetohydroxyacid synthase (AHAS), also known as acetolactate synthase, is a flavin adenine dinucleotide-, thiamine diphosphate- and magnesium-dependent enzyme that catalyses the first step in the biosynthesis of branched-chain amino acids1. It is the target for more than 50 commercial herbicides2. AHAS requires both catalytic and regulatory subunits for maximal activity and functionality. Here we describe structures of the hexadecameric AHAS complexes of Saccharomyces cerevisiae and dodecameric AHAS complexes of Arabidopsis thaliana. We found that the regulatory subunits of these AHAS complexes form a core to which the catalytic subunit dimers are attached, adopting the shape of a Maltese cross. The structures show how the catalytic and regulatory subunits communicate with each other to provide a pathway for activation and for feedback inhibition by branched-chain amino acids. We also show that the AHAS complex of Mycobacterium tuberculosis adopts a similar structure, thus demonstrating that the overall AHAS architecture is conserved across kingdoms.


Subject(s)
Acetolactate Synthase/chemistry , Arabidopsis/enzymology , Saccharomyces cerevisiae/enzymology , Acetolactate Synthase/metabolism , Adenosine Triphosphate/metabolism , Amino Acids, Branched-Chain/biosynthesis , Catalytic Domain , Enzyme Activation , Evolution, Molecular , Feedback, Physiological , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Mycobacterium tuberculosis/enzymology , Protein Binding , Protein Conformation , Protein Subunits/chemistry , Protein Subunits/metabolism , Valine/metabolism
14.
Biochem J ; 477(13): 2439-2449, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32538427

ABSTRACT

The acetohydroxyacid synthase (AHAS) holoenzyme catalyzes the first step of branch-chain amino acid biosynthesis and is essential for plants and bacteria. It consists of a regulatory subunit (RSU) and a catalytic subunit (CSU). The allosteric mechanism of the AHAS holoenzyme has remained elusive for decades. Here, we determined the crystal structure of the AHAS holoenzyme, revealing the association between the RSU and CSU in an A2B2 mode. Structural analysis in combination with mutational studies demonstrated that the RSU dimer forms extensive interactions with the CSU dimer, in which a conserved salt bridge between R32 and D120 may act as a trigger to open the activation loop of the CSU, resulting in the activation of the CSU by the RSU. Our study reveals the activation mechanism of the AHAS holoenzyme.


Subject(s)
Acetolactate Synthase/chemistry , Holoenzymes/chemistry , Allosteric Regulation/physiology , Crystallography
15.
J Chem Inf Model ; 60(2): 915-922, 2020 02 24.
Article in English | MEDLINE | ID: mdl-31841000

ABSTRACT

Acetohydroxyacid synthase (AHAS) is a thiamin diphosphate-dependent enzyme involved in the biosynthesis of valine, leucine, isoleucine, and lysine. Experimental evidence has shown that mutation of the Gln202 residue results in a decrease in the enzymatic activity, thus suggesting the main role of the carboligation catalyzed by AHAS. It has been postulated that this residue acts as an acid/base group, protonating the carbonyl oxygen from the 2-ketoacid substrate, during the carboligation reaction. However, previous studies have revealed that 2-ketoacid is not engaged in catalytically relevant interactions with ionizable groups that can act as an acid/base group during the catalysis. Therefore, it has been proposed that the carboligation reaction could occur through an intramolecular proton transfer without the assistance of an amino acid residue with acid-base properties. To decipher the role of Gln202, in this work, we studied the last two catalytic steps of the AHAS through quantum mechanics/molecular mechanics calculations using a full enzyme model of the wild-type AHAS and the Gln202Ala mutant. Our results indicate that the carboligation mechanism occurs through an intramolecular proton transfer that does not require the action of an additional acid-base group. The mechanism is composed of two steps in which the last one is rate-limiting. Our findings reveal that Gln202 stabilizes a catalytic water molecule in the reactive site through electrostatic contributions that are mostly relevant during the carboligation step, in agreement with experimental evidence. The catalytic water engages in intermolecular hydrogen bonds with the reacting species and makes a strong electronic contribution to the stabilization of the reaction intermediate (AL-ThDP).


Subject(s)
Acetolactate Synthase/chemistry , Acetolactate Synthase/metabolism , Biocatalysis , Glutamine , Yeasts/enzymology , Hydrogen Bonding , Models, Molecular , Protein Conformation , Quantum Theory
16.
J Agric Food Chem ; 67(42): 11568-11576, 2019 Oct 23.
Article in English | MEDLINE | ID: mdl-31584809

ABSTRACT

Tribenuron-methyl (TM), as one of the sulfonylurea (SU) herbicides, has been widely and effectively applied for many kinds of plants. SUs inhibit plant growth by restraining the biosynthetic pathway of branched-chain amino acids (BCAAs) catalyzed by acetolactate synthase (ALS). Safeners are agrochemicals that protect crops from herbicide injuries. To improve the crop tolerance under TM toxicity stress, this paper evaluated the protective effect of N-tosyloxazolidine-3-carboxamide. It turned out that most of the tested compounds showed significant protection against TM via enhancing the glutathione (GSH) content and glutathione S-transferase (GST) activity. Among all of the tested compounds, compound 16 exhibited more excellent protection than the contrast safener R-28725 and other target compounds. A positive correlation between the growth level, endogenous GSH content, and GST activity was observed in this research. The GST kinetic parameter Vmax of the maize was increased by 29.6% after treatment with compound 16, while Km was decreased by 51.9% compared to the untreated control. The molecular docking model indicated that compound 16 could compete with TM in the active site of ALS, which could interpret the protective effects of safeners. The present work demonstrated that N-tosyloxazolidine-3-carboxamide derivatives could be considered as potential candidates for developing new safeners in the future.


Subject(s)
Herbicides/toxicity , Plant Proteins/metabolism , Protective Agents/pharmacology , Zea mays/drug effects , Zea mays/enzymology , Acetolactate Synthase/chemistry , Acetolactate Synthase/metabolism , Glutathione Transferase/chemistry , Glutathione Transferase/metabolism , Kinetics , Molecular Docking Simulation , Plant Proteins/chemistry , Sulfonylurea Compounds/toxicity , Zea mays/chemistry
17.
Molecules ; 24(17)2019 Aug 22.
Article in English | MEDLINE | ID: mdl-31443550

ABSTRACT

Herbicide safeners selectively protect crops from herbicide injury while maintaining the herbicidal effect on the target weed. To some extent, the detoxification of herbicides is related to the effect of herbicide safeners on the level and activity of herbicide target enzymes. In this work, the expression of the detoxifying enzyme glutathione S-transferase (GST) and antioxidant enzyme activities in maize seedlings were studied in the presence of three potential herbicide safeners: 3-dichloroacetyl oxazolidine and its two optical isomers. Further, the protective effect of chiral herbicide safeners on detoxifying chlorsulfuron in maize was evaluated. All safeners increased the expression levels of herbicide detoxifying enzymes, including GST, catalase (CAT), and peroxidase (POD) to reduce sulfonylurea herbicide phytotoxicity in maize seedlings. Our results indicate that the R-isomer of 3-(dichloroacetyl)-2,2,5-trimethyl-1,3-oxazolidine can induce glutathione (GSH) production, GST activity, and the ability of GST to react with the substrate 1-chloro-2,4-dinitrobenzene (CDNB) in maize, meaning that the R-isomer can protect maize from damage by chlorsulfuron. Information about antioxidative enzyme activity was obtained to determine the role of chiral safeners in overcoming the oxidative stress in maize attributed to herbicides. The interaction of safeners and active target sites of acetolactate synthase (ALS) was demonstrated by molecular docking modeling, which indicated that both isomers could form a good interaction with ALS. Our findings suggest that the detoxification mechanism of chiral safeners might involve the induction of the activity of herbicide detoxifying enzymes as well as the completion of the target active site between the safener and chlorsulfuron.


Subject(s)
Inactivation, Metabolic/drug effects , Oxazoles/chemistry , Oxazoles/pharmacology , Protective Agents/chemistry , Protective Agents/pharmacology , Zea mays/drug effects , Zea mays/metabolism , Acetolactate Synthase/chemistry , Acetolactate Synthase/metabolism , Binding Sites , Catalase/metabolism , Catalytic Domain , Glutathione/metabolism , Molecular Conformation , Molecular Docking Simulation , Molecular Dynamics Simulation , Peroxidase/metabolism , Protein Binding , Structure-Activity Relationship , Zea mays/chemistry
18.
Enzyme Microb Technol ; 129: 109357, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31307581

ABSTRACT

Corynebacterium glutamicum acetohydroxyacid synthase (AHAS), composed of two subunits IlvB and IlvN, catalyzes the first reaction in the biosynthetic pathway of branched-chain amino acids. It either condenses two pyruvates to yield acetolactate, leading to the biosynthesis of L-valine and L-leucine, or condenses pyruvate with 2-ketobutyrate to yield acetohydroxybutyrate, leading to L-isoleucine biosynthesis. However, the mechanism for the substrate specificity of C. glutamicum AHAS remains unknown. In this study, AHASs from an L-valine-producing C. glutamicum VWB-1 and an L-isoleucine-producing C. glutamicum IWJ001 were analyzed. The amino acid sequence of IlvN from both strains are the same, but the 138th and 404th residues of IlvB from the two strains are different; they are alanine and valine in IWJ001 (IlvB138A404V), but valine and alanine in VWB-1 (IlvB138V404A). When IlvB138A404V and IlvB138V404A were overexpressed in wild type C. glutamicum ATCC14067 and its △alr△aceE△ilvA△leuA mutant YTW-104, the latter led to much more L-valine production than the former. AHAS activity studies also showed that the 138th valine was important for binding the 2nd substrate pyruvate but not the 404th alanine. YTW-104/pJYW4-ilvB138V404A-ilvNCE could produce 25.93 g/L L-valine. The results indicate that the 138th valine of IlvB in AHAS could play an important role, leading to the increased L-valine biosynthesis in C. glutamicum.


Subject(s)
Acetolactate Synthase/chemistry , Bacterial Proteins/chemistry , Corynebacterium glutamicum/enzymology , Acetolactate Synthase/genetics , Acetolactate Synthase/metabolism , Amino Acid Motifs , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Butyrates/metabolism , Corynebacterium glutamicum/chemistry , Corynebacterium glutamicum/genetics , Isoleucine/metabolism , Pyruvic Acid/metabolism , Substrate Specificity , Valine/metabolism
19.
Biochemistry ; 58(15): 1992-2008, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30887800

ABSTRACT

Conformational factors that predicate selectivity for valine or isoleucine binding to IlvN leading to the regulation of aceto hydroxy acid synthase I (AHAS I) of Escherichia coli have been determined for the first time from high-resolution (1.9-2.43 Å) crystal structures of IlvN·Val and IlvN·Ile complexes. The valine and isoleucine ligand binding pockets are located at the dimer interface. In the IlvN·Ile complex, among residues in the binding pocket, the side chain of Cys43 is 2-fold disordered (χ1 angles of gauche- and trans). Only one conformation can be observed for the identical residue in the IlvN·Val complexes. In a reversal, the side chain of His53, located at the surface of the protein, exhibits two conformations in the IlvN·Val complex. The concerted conformational switch in the side chains of Cys43 and His53 may play an important role in the regulation of the AHAS I holoenzyme activity. A significant result is the establishment of the subunit composition in the AHAS I holoenzyme by analytical ultracentrifugation. Solution nuclear magnetic resonance and analytical ultracentrifugation experiments have also provided important insights into the hydrodynamic properties of IlvN in the ligand-free and -bound states. The structural and biophysical data unequivocally establish the molecular basis for differential binding of the ligands to IlvN and a rationale for the resistance of IlvM to feedback inhibition by the branched-chain amino acids.


Subject(s)
Isoleucine/chemistry , Protein Conformation , Valine/chemistry , Acetolactate Synthase/chemistry , Acetolactate Synthase/genetics , Acetolactate Synthase/metabolism , Amino Acid Sequence , Binding Sites/genetics , Crystallography, X-Ray , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Feedback, Physiological , Hydrogen Bonding , Isoleucine/genetics , Isoleucine/metabolism , Models, Molecular , Protein Binding , Valine/genetics , Valine/metabolism
20.
Proc Natl Acad Sci U S A ; 115(41): E9649-E9658, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30249642

ABSTRACT

The increased prevalence of drug-resistant human pathogenic fungal diseases poses a major threat to global human health. Thus, new drugs are urgently required to combat these infections. Here, we demonstrate that acetohydroxyacid synthase (AHAS), the first enzyme in the branched-chain amino acid biosynthesis pathway, is a promising new target for antifungal drug discovery. First, we show that several AHAS inhibitors developed as commercial herbicides are powerful accumulative inhibitors of Candida albicans AHAS (Ki values as low as 800 pM) and have determined high-resolution crystal structures of this enzyme in complex with several of these herbicides. In addition, we have demonstrated that chlorimuron ethyl (CE), a member of the sulfonylurea herbicide family, has potent antifungal activity against five different Candida species and Cryptococcus neoformans (with minimum inhibitory concentration, 50% values as low as 7 nM). Furthermore, in these assays, we have shown CE and itraconazole (a P450 inhibitor) can act synergistically to further improve potency. Finally, we show in Candida albicans-infected mice that CE is highly effective in clearing pathogenic fungal burden in the lungs, liver, and spleen, thus reducing overall mortality rates. Therefore, in view of their low toxicity to human cells, AHAS inhibitors represent a new class of antifungal drug candidates.


Subject(s)
Acetolactate Synthase , Antifungal Agents , Candida albicans/enzymology , Candidiasis , Cryptococcosis , Cryptococcus neoformans/enzymology , Fungal Proteins , Acetolactate Synthase/antagonists & inhibitors , Acetolactate Synthase/chemistry , Acetolactate Synthase/metabolism , Animals , Antifungal Agents/chemistry , Antifungal Agents/pharmacology , Candidiasis/drug therapy , Candidiasis/enzymology , Cryptococcosis/drug therapy , Cryptococcosis/enzymology , Fungal Proteins/antagonists & inhibitors , Fungal Proteins/chemistry , Herbicides/chemistry , Herbicides/pharmacology , Humans , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...