Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Emerg Microbes Infect ; 10(1): 2291-2299, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34779708

ABSTRACT

The emergence of drug-resistant tuberculosis (TB) constitutes a major challenge to TB control programmes. There is an urgent need to develop effective anti-TB drugs with novel mechanisms of action. Aspartate-semialdehyde dehydrogenase (ASADH) is the second enzyme in the aspartate metabolic pathway. The absence of the pathway in humans and the absolute requirement of aspartate in bacteria make ASADH a highly attractive drug target. In this study, we used ASADH coupled with Escherichia coli type III aspartate kinase (LysC) to establish a high-throughput screening method to find new anti-TB inhibitors. IMB-XMA0038 was identified as an inhibitor of MtASADH with an IC50 value of 0.59 µg/mL through screening. The interaction between IMB-XMA0038 and MtASADH was confirmed by surface plasmon resonance (SPR) assay and molecular docking analysis. Furthermore, IMB-XMA0038 was found to inhibit various drug-resistant MTB strains potently with minimal inhibitory concentrations (MICs) of 0.25-0.5 µg/mL. The conditional mutant strain MTB::asadh cultured with different concentrations of inducer (10-5 or 10-1 µg/mL pristinamycin) resulted in a maximal 16 times difference in MICs. At the same time, IMB-XMA0038 showed low cytotoxicity in vitro and vivo. In mouse model, it encouragingly declined the MTB colony forming units (CFU) in lung by 1.67 log10 dosed at 25 mg/kg for 15 days. In conclusion, our data demonstrate that IMB-XMA0038 is a promising lead compound against drug-resistant tuberculosis.


Subject(s)
Antitubercular Agents/chemistry , Aspartate-Semialdehyde Dehydrogenase/antagonists & inhibitors , Bacterial Proteins/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Tuberculosis/microbiology , Animals , Antitubercular Agents/administration & dosage , Aspartate-Semialdehyde Dehydrogenase/chemistry , Aspartate-Semialdehyde Dehydrogenase/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Enzyme Inhibitors/administration & dosage , Humans , Male , Mice , Molecular Docking Simulation , Mycobacterium tuberculosis/genetics , Tuberculosis/drug therapy
2.
Pak J Pharm Sci ; 34(6): 2219-2226, 2021 Nov.
Article in English | MEDLINE | ID: mdl-35034884

ABSTRACT

This in silico work was carried out to reveal the proposed anti-fungal efficacy of some clove ingredient compounds against aspartate semialdehyde dehydrogenase, 6C8W and 6C85, enzymes from Blastomyces dermatitidis. The molecular docking simulation was implemented utilizing the Auto Dock 4.2. software. A set of 17 compounds were selected for this study, which is known to be active ingredients of Syzygium aromaticum crude and oil. The best docking scores associated with the Blastomyces dermatitidis enzymes 6C85 and 6C8W were for Maslinic acid and Oleanolic acid, followed by Stigmasterol and Campesterol. It was found that these compounds possess inhibitory potential against 6C85 and 6C8W and hence have anti-fungal efficacy. Maslinic acid and Oleanolic acid produced the strongest binding to 6C85 and 6C8W over the remaining bioactive compounds by forming H-bonds with some amino acids in these enzymes.


Subject(s)
Antifungal Agents/pharmacology , Aspartate-Semialdehyde Dehydrogenase/antagonists & inhibitors , Blastomyces/drug effects , Enzyme Inhibitors/pharmacology , Fungal Proteins/antagonists & inhibitors , Molecular Docking Simulation , Plant Extracts/pharmacology , Syzygium , Antifungal Agents/isolation & purification , Aspartate-Semialdehyde Dehydrogenase/metabolism , Blastomyces/enzymology , Catalytic Domain , Enzyme Inhibitors/isolation & purification , Fungal Proteins/metabolism , Hydrogen Bonding , Oleanolic Acid/isolation & purification , Oleanolic Acid/pharmacology , Plant Extracts/isolation & purification , Protein Conformation , Structure-Activity Relationship , Syzygium/chemistry , Triterpenes/isolation & purification , Triterpenes/pharmacology
3.
Drug Dev Res ; 81(6): 736-744, 2020 09.
Article in English | MEDLINE | ID: mdl-32383780

ABSTRACT

Potent inhibitors of an essential microbial enzyme have been shown to be effective growth inhibitors of Candida albicans, a pathogenic fungus. C. albicans is the main cause of oropharyngeal candidiasis, and also causes invasive fungal infections, including systemic sepsis, leading to serious complications in immunocompromised patients. As the rates of drug-resistant fungal infections continue to rise novel antifungal treatments are desperately needed. The enzyme aspartate semialdehyde dehydrogenase (ASADH) is critical for the functioning of the aspartate biosynthetic pathway in microbes and plants. Because the aspartate pathway is absent in humans, ASADH has the potential to be a promising new target for antifungal research. Deleting the asd gene encoding for ASADH significantly decreases the survival of C. albicans, establishing this enzyme as essential for this organism. Previously developed ASADH inhibitors were tested against several strains of C. albicans to measure their possible therapeutic impact. The more potent inhibitors show a good correlation between enzyme inhibitor potency and fungal growth inhibition. Growth curves generated by incubating different C. albicans strains with varying enzyme inhibitor levels show significant slowing of fungal growth by these inhibitors against each of these strains, similar to the effect observed with a clinical antifungal drug. The most effective inhibitors also demonstrated relatively low cytotoxicity against a human epithelial cell line. Taken together, these results establish that the ASADH enzyme is a promising new target for further development as a novel antifungal treatment against C. albicans and related fungal species.


Subject(s)
Antifungal Agents/pharmacology , Aspartate-Semialdehyde Dehydrogenase/antagonists & inhibitors , Benzoquinones/pharmacology , Candida albicans/drug effects , Naphthoquinones/pharmacology , Aspartate-Semialdehyde Dehydrogenase/genetics , Candida albicans/genetics , Candida albicans/growth & development , Cell Survival/drug effects , Cells, Cultured , Gene Deletion , Humans , Mouth Mucosa/cytology
4.
Molecules ; 24(19)2019 Oct 07.
Article in English | MEDLINE | ID: mdl-31591315

ABSTRACT

Helicobacter pylori infection is a WHO class 1 carcinogenic factor of gastric adenocarcinoma. In the past decades, many studies have demonstrated the increasing trend of antibiotic resistance and pointed out the necessity of new effective treatment. This study was aimed at identifying phytochemicals that can inhibit H. pylori and possibly serve as adjuvant treatments. Here, in silico molecular docking and drug-like properties analyses were performed to identify potential inhibitors of urease, shikimate kinase and aspartate-semialdehyde dehydrogenase. These three enzymes are targets of the treatment of H. pylori. Susceptibility and synergistic testing were performed on the selected phytochemicals and the positive control antibiotic, amoxicillin. The in-silico study revealed that oroxindin, rosmarinic acid and verbascoside are inhibitors of urease, shikimate kinase and aspartate-semialdehyde dehydrogenase, respectively, in which, oroxindin has the highest potency against H. pylori, indicated by a minimum inhibitory concentration (MIC) value of 50 µg/mL. A combination of oroxindin and amoxicillin demonstrated additive effects against H. pylori, as indicated by a fractional inhibitory concentration (FIC) value of 0.75. This study identified phytochemicals that deserve further investigation for the development of adjuvant therapeutic agents to current antibiotics against H. pylori.


Subject(s)
Amoxicillin/pharmacology , Anti-Bacterial Agents/pharmacology , Helicobacter pylori/drug effects , Phytochemicals/pharmacology , Anti-Bacterial Agents/chemistry , Aspartate-Semialdehyde Dehydrogenase/antagonists & inhibitors , Chromones/chemistry , Chromones/pharmacology , Cinnamates/chemistry , Cinnamates/pharmacology , Clarithromycin/pharmacology , Computer Simulation , Depsides/chemistry , Depsides/pharmacology , Drug Resistance, Microbial/drug effects , Glucosides/chemistry , Glucosides/pharmacology , Glucuronates/chemistry , Glucuronates/pharmacology , Molecular Docking Simulation , Phenols/chemistry , Phenols/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phytochemicals/chemistry , Urease/antagonists & inhibitors , Rosmarinic Acid
5.
J Biomol Struct Dyn ; 37(2): 394-410, 2019 Feb.
Article in English | MEDLINE | ID: mdl-29334340

ABSTRACT

Lymphatic filariasis is a debilitating vector borne parasitic disease that infects human lymphatic system by nematode Brugia malayi. Currently available anti-filarial drugs are effective only on the larval stages of parasite. So far, no effective drugs are available for humans to treat filarial infections. In this regard, aspartate semialdehyde dehydrogenase (ASDase) in lysine biosynthetic pathway from Wolbachia endosymbiont Brugia malayi represents an attractive therapeutic target for the development of novel anti-filarial agents. In this present study, molecular modeling combined with molecular dynamics simulations and structure-based virtual screening were performed to identify potent lead molecules against ASDase. Based on Glide score, toxicity profile, binding affinity and mode of interactions with the ASDase, five potent lead molecules were selected. The molecular docking and dynamics results revealed that the amino acid residues Arg103, Asn133, Cys134, Gln161, Ser164, Lys218, Arg239, His246, and Asn321 plays a crucial role in effective binding of Top leads into the active site of ASDase. The stability of the ASDase-lead complexes was confirmed by running the 30 ns molecular dynamics simulations. The pharmacokinetic properties of the identified lead molecules are in the acceptable range. Furthermore, density functional theory and binding free energy calculations were performed to rank the lead molecules. Thus, the identified lead molecules can be used for the development of anti-filarial agents to combat the pathogenecity of Brugia malayi.


Subject(s)
Anthelmintics/chemistry , Aspartate-Semialdehyde Dehydrogenase/chemistry , Brugia malayi/enzymology , Enzyme Inhibitors/chemistry , Molecular Docking Simulation , Molecular Dynamics Simulation , Amino Acid Sequence , Animals , Anthelmintics/pharmacology , Aspartate-Semialdehyde Dehydrogenase/antagonists & inhibitors , Binding Sites , Catalytic Domain , Chemical Phenomena , Drug Discovery/methods , Enzyme Inhibitors/pharmacology , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Protein Binding
6.
SLAS Discov ; 23(6): 520-531, 2018 07.
Article in English | MEDLINE | ID: mdl-29608391

ABSTRACT

Pathogenic fungi represent a growing threat to human health, with an increase in the frequency of drug-resistant fungal infections. Identifying targets from among the selected metabolic pathways that are unique to microbial species presents an opportunity to develop new antifungal agents against new and untested targets to combat this growth threat. Aspartate semialdehyde dehydrogenase (ASADH) catalyzes a key step in a uniquely microbial amino acid biosynthetic pathway and is essential for microbial viability. This enzyme, purified from four pathogenic fungal organisms ( Candida albicans, Aspergillus fumigatus, Cryptococcus neoformans, and Blastomyces dermatitidis), has been screened against fragment libraries to identify initial enzyme inhibitors. The binding of structural analogs of the most promising lead compounds was measured against these fungal ASADHs to establish important structure-activity relationships among these different inhibitor classes. The most potent of these inhibitors have been docked into structures of this fungal enzyme target to identify important structural elements that serve as critical binding determinants. Several inhibitors with low micromolar inhibition constants have been identified that showed selectivity against these related enzymes from different fungal species. Subsequent screening against a library of drugs and drug candidates identified some additional inhibitors containing a consistent set of functional groups required for fungal ASADH inhibition. Additional elaboration of these core structures will likely lead to more potent and selective inhibitors.


Subject(s)
Antifungal Agents/pharmacology , Aspartate-Semialdehyde Dehydrogenase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Fungi/drug effects , Small Molecule Libraries/pharmacology , Fungi/metabolism , Structure-Activity Relationship
7.
J Cell Biochem ; 119(3): 2832-2842, 2018 03.
Article in English | MEDLINE | ID: mdl-29068470

ABSTRACT

The emergence of multi-drug resistant strains and co-occurrence of tuberculosis with HIV creates a major burden to the human health globally. Failure of primary antibacterial therapy necessitates the identification of new mycobacterial drugs. In this study, a comprehensive analysis involving bottom-up systems biology approach was applied wherein we have identified potential therapeutic targets of Mycobacterium tuberculosis infections. Our study prioritized M. tuberculosis therapeutic targets (aspartate-ß-semialdeyhde dehydrogenase [ASD], dihydrodipicolinate reductase and diaminopimelate decarboxylase) based on flux and elementary mode analysis using direct mathematical modeling of the relevant metabolic pathways. Molecular docking and simulation studies of the priority target (ie, ASD) revealed the therapeutic potential of the selected natural products (Huperzine A, Rosmarinic acid, and Curcumin) based ASD inhibitors. The study highlights the crucial role of systems biology in conjunction with molecular interaction (docking) for probing novel leads against an increasingly resistant pathogen, M. tuberculousis.


Subject(s)
Antitubercular Agents/chemistry , Aspartate-Semialdehyde Dehydrogenase , Enzyme Inhibitors/chemistry , Molecular Docking Simulation , Mycobacterium tuberculosis/enzymology , Aspartate-Semialdehyde Dehydrogenase/antagonists & inhibitors , Aspartate-Semialdehyde Dehydrogenase/chemistry , Computer Simulation , Tuberculosis/drug therapy , Tuberculosis/enzymology
8.
Bioorg Med Chem ; 23(20): 6622-31, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26404410

ABSTRACT

Aspartate-ß-semialdehyde dehydrogenase (ASADH) lies at the first branch point in the aspartate metabolic pathway which leads to the biosynthesis of several essential amino acids and some important metabolites. This pathway is crucial for many metabolic processes in plants and microbes like bacteria and fungi, but is absent in mammals. Therefore, the key microbial enzymes involved in this pathway are attractive potential targets for development of new antibiotics with novel modes of action. The ASADH enzyme family shares the same substrate binding and active site catalytic groups; however, the enzymes from representative bacterial and fungal species show different inhibition patterns when previously screened against low molecular weight inhibitors identified from fragment library screening. In the present study several approaches, including fragment based drug discovery (FBDD), inhibitor docking, kinetic, and structure-activity relationship (SAR) studies have been used to guide ASADH inhibitor development. Elaboration of a core structure identified by FBDD has led to the synthesis of low micromolar inhibitors of the target enzyme, with high selectivity introduced between the Gram-negative and Gram-positive orthologs of ASADH. This new set of structures open a novel direction for the development of inhibitors against this validated drug-target enzyme.


Subject(s)
Aspartate-Semialdehyde Dehydrogenase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays , Aspartate-Semialdehyde Dehydrogenase/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Molecular Structure , Streptococcus pneumoniae/enzymology , Structure-Activity Relationship
9.
J Biomol Struct Dyn ; 33(5): 1082-93, 2015.
Article in English | MEDLINE | ID: mdl-24875451

ABSTRACT

Aspartate ß-semialdehyde dehydrogenase (ASADH) is a key enzyme for the biosynthesis of essential amino acids and several important metabolites in microbes. Inhibition of ASADH enzyme is a promising drug target strategy against Mycobacterium tuberculosis (Mtb). In this work, in silico approach was used to identify potent inhibitors of Mtb-ASADH. Aspartyl ß-difluorophosphonate (ß-AFP), a known lead compound, was used to understand the molecular recognition interactions (using molecular docking and molecular dynamics analysis). This analysis helped in validating the computational protocol and established the participation of Arg99, Glu224, Cys130, Arg249, and His256 amino acids as the key amino acids in stabilizing ligand-enzyme interactions for effective binding, an essential feature is H-bonding interactions with the two arginyl residues at the two ends of the ligand. Best binding conformation of ß-AFP was selected as a template for shape-based virtual screening (ZINC and NCI databases) to identify compounds that competitively inhibit the Mtb-ASADH. The top rank hits were further subjected to ADME and toxicity filters. Final filter was based on molecular docking analysis. Each screened molecule carries the characteristics of the highly electronegative groups on both sides separated by an average distance of 6 Å. Finally, the best predicted 20 compounds exhibited minimum three H-bonding interactions with Arg99 and Arg249. These identified hits can be further used for designing the more potent inhibitors against ASADH family. MD simulations were also performed on two selected compounds (NSC4862 and ZINC02534243) for further validation. During the MD simulations, both compounds showed same H-bonding interactions and remained bound to key active residues of Mtb-ASADH.


Subject(s)
Aspartate-Semialdehyde Dehydrogenase/chemistry , Bacterial Proteins/chemistry , Enzyme Inhibitors/chemistry , Molecular Docking Simulation , Molecular Dynamics Simulation , Aspartate-Semialdehyde Dehydrogenase/antagonists & inhibitors , Aspartate-Semialdehyde Dehydrogenase/metabolism , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Binding Sites , Computer Simulation , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Hydrogen Bonding , Ligands , Molecular Structure , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Organophosphonates/chemistry , Organophosphonates/metabolism , Organophosphonates/pharmacology , Protein Binding , Protein Structure, Tertiary , Structure-Activity Relationship
10.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 12): 3244-52, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25478842

ABSTRACT

The aspartate pathway is essential for the production of the amino acids required for protein synthesis and of the metabolites needed in bacterial development. This pathway also leads to the production of several classes of quorum-sensing molecules that can trigger virulence in certain microorganisms. The second enzyme in this pathway, aspartate ß-semialdehyde dehydrogenase (ASADH), is absolutely required for bacterial survival and has been targeted for the design of selective inhibitors. Fragment-library screening has identified a new set of inhibitors that, while they do not resemble the substrates for this reaction, have been shown to bind at the active site of ASADH. Structure-guided development of these lead compounds has produced moderate inhibitors of the target enzyme, with some selectivity observed between the Gram-negative and Gram-positive orthologs of ASADH. However, many of these inhibitor analogs and derivatives have not yet achieved the expected enhanced affinity. Structural characterization of these enzyme-inhibitor complexes has provided detailed explanations for the barriers that interfere with optimal binding. Despite binding in the same active-site region, significant changes are observed in the orientation of these bound inhibitors that are caused by relatively modest structural alterations. Taken together, these studies present a cautionary tale for issues that can arise in the systematic approach to the modification of lead compounds that are being used to develop potent inhibitors.


Subject(s)
Aspartate-Semialdehyde Dehydrogenase/antagonists & inhibitors , Aspartate-Semialdehyde Dehydrogenase/chemistry , Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Streptococcus pneumoniae/enzymology , Vibrio cholerae/enzymology , Aspartate-Semialdehyde Dehydrogenase/metabolism , Aspartic Acid/metabolism , Biosynthetic Pathways , Catalytic Domain , Cholera/microbiology , Crystallography, X-Ray , Humans , Models, Molecular , Pneumococcal Infections/microbiology , Protein Conformation , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Streptococcus pneumoniae/chemistry , Streptococcus pneumoniae/metabolism , Vibrio cholerae/chemistry , Vibrio cholerae/metabolism
11.
Bioorg Med Chem ; 20(9): 2950-6, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22464683

ABSTRACT

Microbes that have gained resistance against antibiotics pose a major emerging threat to human health. New targets must be identified that will guide the development of new classes of antibiotics. The selective inhibition of key microbial enzymes that are responsible for the biosynthesis of essential metabolites can be an effective way to counter this growing threat. Aspartate semialdehyde dehydrogenases (ASADHs) produce an early branch point metabolite in a microbial biosynthetic pathway for essential amino acids and for quorum sensing molecules. In this study, molecular modeling and docking studies were performed to achieve two key objectives that are important for the identification of new selective inhibitors of ASADH. First, virtual screening of a small library of compounds was used to identify new core structures that could serve as potential inhibitors of the ASADHs. Compounds have been identified from diverse chemical classes that are predicted to bind to ASADH with high affinity. Next, molecular docking studies were used to prioritize analogs within each class for synthesis and testing against representative bacterial forms of ASADH from Streptococcus pneumoniae and Vibrio cholerae. These studies have led to new micromolar inhibitors of ASADH, demonstrating the utility of this molecular modeling and docking approach for the identification of new classes of potential enzyme inhibitors.


Subject(s)
Aspartate-Semialdehyde Dehydrogenase/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Aspartate-Semialdehyde Dehydrogenase/metabolism , Binding Sites , Enzyme Inhibitors/chemical synthesis , Kinetics , Molecular Dynamics Simulation , Protein Structure, Tertiary , Streptococcus pneumoniae/enzymology , Vibrio cholerae/enzymology
12.
J Biomol Screen ; 17(5): 673-82, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22460173

ABSTRACT

The rise in organisms resistant to existing drugs has added urgency to the search for new antimicrobial agents. Aspartate ß-semialdehyde dehydrogenase (ASADH) catalyzes a critical step in an essential microbial pathway that is absent in mammals. Our laboratory is using fragment library screening to identify efficient and selective ASADH inhibitors. These preliminary agents are then tested to identify compounds with desired antimicrobial properties for further refinement. Toward this end, we have established a microplate-based, dual-assay approach using a single reagent to evaluate antibiotic activity and mammalian cell toxicity during early stage development. The bacterial assay uses nonpathogenic bacteria to allow efficacy testing without a dedicated microbial laboratory. Toxicity assays are performed with a panel of mammalian cells derived from representative susceptible tissues. These assays can be adapted to target other microbial systems, such as fungi and biofilms, and additional mammalian cell lines can be added as needed. Application of this screening approach to antibiotic standards demonstrates the ability of these assays to identify bacterial selectivity and potential toxicity issues. Tests with selected agents from the ASADH inhibitor fragment library show some compounds with antibiotic activity, but as expected, most of these early agents display higher than desired mammalian cell toxicity.


Subject(s)
Anti-Bacterial Agents/pharmacology , Aspartate-Semialdehyde Dehydrogenase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Microbial Sensitivity Tests/methods , Anti-Bacterial Agents/toxicity , Cell Line , Enzyme Inhibitors/toxicity , Humans , Inhibitory Concentration 50 , Reproducibility of Results
13.
Chem Biol Drug Des ; 79(1): 128-36, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22039970

ABSTRACT

The aspartate biosynthetic pathway provides essential metabolites for many important biological functions, including the production of four essential amino acids. As this critical pathway is only present in plants and microbes, any disruptions will be fatal to these organisms. An early pathway enzyme, l-aspartate-ß-semialdehyde dehydrogenase, produces a key intermediate at the first branch point of this pathway. Developing potent and selective inhibitors against several orthologs in the l-aspartate-ß-semialdehyde dehydrogenase family can serve as lead compounds for antibiotic development. Kinetic studies of two small molecule fragment libraries have identified inhibitors that show good selectivity against l-aspartate-ß-semialdehyde dehydrogenases from two different bacterial species, Streptococcus pneumoniae and Vibrio cholerae, despite the presence of an identical constellation of active site amino acids in this homologous enzyme family. Structural characterization of enzyme-inhibitor complexes have elucidated different modes of binding between these structurally related enzymes. This information provides the basis for a structure-guided approach to the development of more potent and more selective inhibitors.


Subject(s)
Anti-Bacterial Agents/chemistry , Aspartate-Semialdehyde Dehydrogenase/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Aspartate-Semialdehyde Dehydrogenase/metabolism , Binding Sites , Catalytic Domain , Computer Simulation , Crystallography, X-Ray , Drug Design , Enzyme Activation/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Propionates/chemistry , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/enzymology , Substrate Specificity , Vibrio cholerae/drug effects , Vibrio cholerae/enzymology
14.
Mol Biosyst ; 7(5): 1564-75, 2011 May.
Article in English | MEDLINE | ID: mdl-21369577

ABSTRACT

Inhibitors of the enzyme aspartate semialdehyde dehydrogenase, a key biological target for the generation of a new class of antibiotic compounds, have been developed. To investigate improvements to binding within an inhibitor series, the lowering of the entropic barrier to binding through conformational restriction was investigated. A library of linear and cyclic substrate analogues was generated and computational docking used to aid in structure selection. The cyclic phosphonate inhibitor 18 was thus identified as complimentary to the enzyme active-site. Synthesis and in vitro inhibition assay revealed a K(i) of 3.8 mM against natural substrate, where the linear analogue of 18, compound 15, had previously shown no inhibitory activity. Two further inhibitors, phosphate analogue diastereoisomers 17a and 17b, were synthesised and also found to have low millimolar K(i) values. As a result of the computational docking investigations, a novel substrate binding interaction was discovered: hydrogen bonding between the substrate (phosphate hydroxy-group as the hydrogen bond donor) and the NADPH cofactor (2'-oxygen as the hydrogen bond acceptor).


Subject(s)
Aspartate-Semialdehyde Dehydrogenase/antagonists & inhibitors , Bacterial Proteins/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Amino Acid Sequence , Aspartate-Semialdehyde Dehydrogenase/genetics , Aspartate-Semialdehyde Dehydrogenase/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Computer Simulation , Enzyme Inhibitors/chemical synthesis , Escherichia coli/enzymology , Escherichia coli/genetics , Hydrogen Bonding , Kinetics , Models, Molecular , Molecular Conformation , Molecular Sequence Data , Molecular Structure , NADP/chemistry , NADP/metabolism , Protein Binding , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Streptococcus pneumoniae/enzymology , Streptococcus pneumoniae/genetics , Structure-Activity Relationship , Substrate Specificity
15.
J Biomol Screen ; 15(9): 1042-50, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20855558

ABSTRACT

The microbial threat to human health is growing due to the dramatic increase in the number of multidrug-resistant organisms. The decline in effective antibiotics available to treat these growing threats has provided greater urgency to the search for new antibiotics. Clearly, new approaches must be developed against novel targets to control these resistant infectious organisms. The screening of low molecular weight compounds against new protein targets provides an opportunity to identify novel inhibitors as starting points for the development of new antibiotics. Custom fragment libraries have been assembled and screened against 3 representative forms of a key enzyme in an essential microbial biosynthetic pathway. Although each of these aspartate semialdehyde dehydrogenases (ASADHs) catalyzes the same reaction and each shares identical active site functional groups, subtle differences in enzyme structures have led to different binding selectivity among the initial hits from these fragment libraries. Amino acid analogues have been identified that show selectivity for either the gram-negative or gram-positive bacterial enzyme forms. A series of benzophenone analogues selectively inhibit the gram-negative ASADH, whereas some haloacids and substituted aromatic acids have been found to inhibit only the fungal form of ASADH. Each of these low molecular weight compounds possesses high ligand binding efficiency for their target enzyme forms. These results support the goal of designing lead compounds that will selectively target ASADHs from different microbial species.


Subject(s)
Aspartate-Semialdehyde Dehydrogenase/antagonists & inhibitors , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/analysis , Enzyme Inhibitors/pharmacology , Small Molecule Libraries/analysis , Small Molecule Libraries/pharmacology , Amino Acids/analysis , Amino Acids/pharmacology , Benzophenones/analysis , Benzophenones/pharmacology , Biocatalysis/drug effects , Candida albicans/drug effects , Microbial Sensitivity Tests , Streptococcus pneumoniae/drug effects , Substrate Specificity , Vibrio cholerae/drug effects
16.
Chembiochem ; 6(12): 2255-60, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16261551

ABSTRACT

Unsaturated and fluorinated analogues of aspartyl-beta-phosphate were synthesised as potential inhibitors of the bacterial enzyme aspartate semialdehyde dehydrogenase (ASA-DH). Acetylenic and Z-olefinic analogues showed competitive inhibition, but an E-olefinic analogue was inactive. A monofluoromethylene phosphonate competed poorly, but showed time-dependent inhibition of ASA-DH in the absence of phosphate. Simulated docking procedures were used to rationalise the results. These studies showed that substrate and inhibitor binding are mediated by interaction with two active-site arginine residues, and for likely covalent attachment to the active-site thiol group, electrophilic carbon atoms should be located 4.5 A, or less, from the thiol.


Subject(s)
Aspartate-Semialdehyde Dehydrogenase/antagonists & inhibitors , Enzyme Inhibitors/chemical synthesis , Arginine , Aspartic Acid/analogs & derivatives , Aspartic Acid/chemical synthesis , Aspartic Acid/pharmacology , Bacterial Proteins/antagonists & inhibitors , Binding Sites , Binding, Competitive , Drug Design , Enzyme Inhibitors/pharmacology , Structure-Activity Relationship , Substrate Specificity
17.
Res Microbiol ; 155(7): 525-34, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15313252

ABSTRACT

Carbon flow through the lysine branch of the aspartate biosynthetic pathway is a rate-limiting step in the formation of cephamycin C, a broad spectrum beta-lactam antibiotic produced by Streptomyces clavuligerus. In this study, genes which encode the enzymes catalyzing the first two steps of the aspartate pathway, ask (aspartokinase) and asd (aspartate semialdehyde dehydrogenase), in S. clavuligerus NRRL 3585 were cloned and sequenced. Nucleotide sequencing and codon preference analysis revealed three complete open reading frames (ORFs). ORF2 starts within ORF1 and terminates by utilizing the same stop codon as ORF1, an arrangement typical of many ask genes. ORF3 is located 2 nucleotides downstream of ORF1,2. Database comparisons with these proteins identified ORF1 as the large (alpha) subunit of aspartokinase, ORF2 as the small (beta) subunit of aspartokinase and ORF3 as the aspartate semialdehyde dehydrogenase. The cloned genes were functionally expressed in auxotrophic Escherichia coli strains, CGSC5074 (ask(-)) and E. coli CGSC5080 (asd(-)), the two enzymes were partially purified from E. coli cell extracts and their kinetic parameters were determined. The effects of end product amino acids and diaminopimelic acid on the activity of Ask and Asd enzymes were also described.


Subject(s)
Aspartate Kinase/genetics , Aspartate-Semialdehyde Dehydrogenase/genetics , Cephamycins/biosynthesis , Operon , Streptomyces/genetics , Aspartate Kinase/antagonists & inhibitors , Aspartate Kinase/isolation & purification , Aspartate Kinase/metabolism , Aspartate-Semialdehyde Dehydrogenase/antagonists & inhibitors , Aspartate-Semialdehyde Dehydrogenase/isolation & purification , Aspartate-Semialdehyde Dehydrogenase/metabolism , Cloning, Molecular , Codon, Initiator , Codon, Terminator , Open Reading Frames/genetics , Rifamycins/biosynthesis , Sequence Analysis, DNA , Sequence Homology , Streptomyces/enzymology , Streptomyces/metabolism
18.
Biochim Biophys Acta ; 1696(1): 23-9, 2004 Jan 14.
Article in English | MEDLINE | ID: mdl-14726201

ABSTRACT

Aspartate-beta-semialdehyde dehydrogenase (ASADH) from Escherichia coli is inhibited by L- and D-cystine, and by other cystine derivatives. Enzyme inhibition is quantitatively reversed by addition of dithiothreitol (DTT), dithioerythrytol, beta-mercaptoethanol, di-mercaptopropanol or glutathione to the cystine-inactivated enzyme. Cystine labeling of the enzyme is a pH dependent process and is optimal at pH values ranging from 7.0 to 7.5. Both the cysteine incorporation profile and the inactivation curve of the enzyme as a function of pH suggest that a group(s) with pK(a) of 8.5 could be involved in cystine binding. Stoichiometry of the inactivation reaction indicates that one cysteine residue from the enzyme subunit is reactive against cystine, as found by direct incorporation of radioactive cystine into the enzyme and by free-thiol titration of the enzyme with 5,5'-dithiobis-2-nitrobenzoic acid (DTNB) before and after the cystine treatment. One mole of cysteine is released from each mol of cystine after reaction with the enzyme. ASA, NADP and NADPH did not prevent cystine inhibition. The [35S]cysteine-labelled enzyme can be visualized after electrophoresis in polyacrylamide gels and further detection by autoradiography. After pepsin treatment of the [35S]cysteine-inactivated enzyme, a main radioactive peptide was isolated by HPLC. The amino acid sequence of this peptide was determined as FVGGN(Cys)(2)TVSL, thus demonstrating that the essential 135Cys is the amino acid residue modified by the treatment with cystine.


Subject(s)
Aspartate-Semialdehyde Dehydrogenase/antagonists & inhibitors , Cystine/pharmacology , Escherichia coli Proteins/antagonists & inhibitors , Aspartate-Semialdehyde Dehydrogenase/analysis , Aspartate-Semialdehyde Dehydrogenase/chemistry , Autoradiography , Cystine/analogs & derivatives , Cystine/chemistry , Electrophoresis, Polyacrylamide Gel , Escherichia coli Proteins/chemistry , Hydrogen-Ion Concentration , Mass Spectrometry , Pepsin A , Peptide Fragments/analysis , Sulfur Radioisotopes
19.
C R Biol ; 326(5): 501-8, 2003 May.
Article in English | MEDLINE | ID: mdl-12886877

ABSTRACT

The enzymatic activities of threonine pathway in Escherichia coli are sensitive to pollutants such as cadmium, copper and mercury, which, even at low concentration, can substantially decrease or even block the pathway at several steps. Our aim was to investigate the complex effects on a metabolic pathway of such general enzyme inhibitors with several sites of action, using a previously developed computer simulation of the pathway. For this purpose, the inhibition parameters were experimentally determined and incorporated in the model. The calculation of the flux control coefficient distribution between the five steps of the threonine pathway showed that control remains shared between the three first steps under most inhibition conditions. Response coefficient analysis shows that the inhibition of aspartate semialdehyde dehydrogenase is quantitatively dominant in most circumstances.


Subject(s)
Environmental Pollutants/pharmacology , Escherichia coli/drug effects , Escherichia coli/metabolism , Models, Biological , Threonine/metabolism , Aspartate-Semialdehyde Dehydrogenase/antagonists & inhibitors , Cadmium/pharmacology , Computer Simulation , Copper/pharmacology , Enzyme Inhibitors/pharmacology , Kinetics , Mathematics , Mercury/pharmacology
20.
Mini Rev Med Chem ; 3(2): 115-27, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12570844
SELECTION OF CITATIONS
SEARCH DETAIL
...