Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Carbohydr Polym ; 300: 120255, 2023 Jan 15.
Article in English | MEDLINE | ID: mdl-36372512

ABSTRACT

Hyaluronan (HA) plays important roles in a wide range of biological events. The principal receptor of HA in the human body is the Cluster of Differentiation 44 (CD44). To enhance the binding between HA and CD44, a new approach was designed to take advantage of the four-component Ugi reaction. By modifying the carboxyl group on HA with various amine, aldehyde, and isocyanide moieties through the Ugi reaction, 36 HA like polysaccharides were generated. Two lead compounds were identified with enhanced CD44 binding compared to unmodified HA, which was confirmed by surface plasmon resonance (SPR), cellular studies and an in vivo mouse tumor model. Ski-learn as a machine learning tool was applied to analyze library data and yield predictions with an accuracy over 80 %. In conclusion, modification of HA via the Ugi reaction can be a promising strategy to develop novel binders toward HA receptors such as CD44.


Subject(s)
Hyaluronan Receptors , Hyaluronic Acid , Humans , Mice , Animals , Hyaluronic Acid/chemistry , Hyaluronan Receptors/metabolism , Polysaccharides
2.
Arch Pharm (Weinheim) ; 355(9): e2200095, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35642311

ABSTRACT

Since the discovery of the caspase-2 (Casp2)-mediated ∆tau314 cleavage product and its associated impact on tauopathies such as Alzheimer's disease, the design of selective Casp2 inhibitors has become a focus in medicinal chemistry research. In the search for new lead structures with respect to Casp2 selectivity and drug-likeness, we have taken an approach by looking more closely at the specific sites of Casp2-mediated proteolysis. Using seven selected protein cleavage sequences, we synthesized a peptide series of 53 novel molecules and studied them using in vitro pharmacology, molecular modeling, and crystallography. Regarding Casp2 selectivity, AcITV(Dab)D-CHO (23) and AcITV(Dap)D-CHO (26) demonstrated the best selectivity (1-6-fold), although these trends were only moderate. However, some analogous tetrapeptides, most notably AcDKVD-CHO (45), showed significantly increased Casp3 selectivities (>100-fold). Tetra- and tripeptides display decreased or no Casp2 affinity, supporting the assumption that a motif of five amino acids is required for efficient Casp2 inhibition. Overall, the results provide a reasonable basis for the development of both selective Casp2 and Casp3 inhibitors.


Subject(s)
Caspase 2 , Caspase 2/metabolism , Caspase 3/metabolism , Caspase Inhibitors/pharmacology , Proteolysis , Structure-Activity Relationship
3.
ACS Omega ; 7(1): 207-214, 2022 Jan 11.
Article in English | MEDLINE | ID: mdl-35036692

ABSTRACT

The first crystal structure of the human cytosolic malate dehydrogenase I (MDH1) is described. Structure determination at a high resolution (1.65 Å) followed production, isolation, and purification of human MDH1 using a bacterial expression system. The structure is a binary complex of MDH1 with only a bound malonate molecule in the substrate binding site. Comparisons of this structure with malate dehydrogenase enzymes from other species confirm that the human enzyme adopts similar secondary, tertiary, and quaternary structures and that the enzyme retains a similar conformation even when nicotinamide adenine dinucleotide (NAD+) is not bound. A comparison to the highly homologous porcine (sus scrofa) MDH1 ternary structures leads to the conclusion that only small conformational differences are needed to accommodate binding by NAD+ or other NAD+ mimetics. Conformational differences observed in the second subunit show that the NAD+ binding elements are nevertheless quite flexible. Comparison of hMDH1 to the human mitochondrial malate dehydrogenase (hMDH2) reveals some key differences in the α7-α8 loop, which lies directly beneath the substrate binding pocket. These differences might be exploited in the structure-assisted design of selective small molecule inhibitors of hMDH1, an emerging target for the development of anticancer therapeutics.

4.
ACS Pharmacol Transl Sci ; 5(1): 20-40, 2022 Jan 14.
Article in English | MEDLINE | ID: mdl-35059567

ABSTRACT

Alzheimer's disease (AD) was first described by Alois Alzheimer over 100 years ago, but there is still no overarching theory that can explain its cause in detail. There are also no effective therapies to treat either the cause or the associated symptoms of this devastating disease. A potential approach to better understand the pathogenesis of AD could be the development of selective caspase-2 (Casp2) probes, as we have shown that a Casp2-mediated cleavage product of tau (Δtau314) reversibly impairs cognitive and synaptic function in animal models of tauopathies. In this article, we map out the Casp2 binding site through the preparation and assay of a series of 35 pentapeptide inhibitors with the goal of gaining selectivity against caspase-3 (Casp3). We also employed computational docking methods to understand the key interactions in the binding pocket of Casp2 and the differences predicted for binding at Casp3. Moreover, we crystallographically characterized the binding of selected pentapeptides with Casp3. Furthermore, we engineered and expressed a series of recombinant tau mutants and investigated them in an in vitro cleavage assay. These studies resulted in simple peptidic inhibitors with nanomolar affinity, for example, AcVDV(Dab)D-CHO (24) with up to 27.7-fold selectivity against Casp3. Our findings provide a good basis for the future development of selective Casp2 probes and inhibitors that can serve as pharmacological tools in planned in vivo studies and as lead compounds for the design of bioavailable and more drug-like small molecules.

5.
J Mol Biol ; 430(17): 2709-2721, 2018 08 17.
Article in English | MEDLINE | ID: mdl-29787766

ABSTRACT

Inherited peripheral neuropathies are a group of neurodegenerative disorders that clinically affect 1 in 2500 individuals. Recently, genetic mutations in human histidine nucleotide-binding protein 1 (hHint1) have been strongly and most frequently associated with patients suffering from axonal neuropathy with neuromyotonia. However, the correlation between the impact of these mutations on the hHint1 structure, enzymatic activity and in vivo function has remained ambiguous. Here, we provide detailed biochemical characterization of a set of these hHint1 mutations. Our findings indicate that half of the mutations (R37P, G93D and W123*) resulted in a destabilization of the dimeric state and a significant decrease in catalytic activity and HINT1 inhibitor binding affinity. The H112N mutant was found to be dimeric, but devoid of catalytic activity, due to the loss of the catalytically essential histidine; nevertheless, it exhibited high affinity to AMP and a HINT1 inhibitor. In contrast to the active-site mutants, the catalytic activity and dimeric structure of the surface mutants, C84R and G89V, were found to be similar to the wild-type enzyme. Taken together, our results suggest that the pathophysiology of inherited axonal neuropathy with neuromyotonia can be induced by conversion of HINT1 from a homodimer to monomer, by modification of select surface residues or by a significant reduction of the enzyme's catalytic efficiency.


Subject(s)
Isaacs Syndrome/genetics , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Mutation , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Peripheral Nervous System Diseases/genetics , Amino Acid Sequence , Crystallography, X-Ray , Histidine/metabolism , Humans , Mutant Proteins/genetics , Nerve Tissue Proteins/genetics , Protein Conformation , Sequence Homology
6.
Mol Pharm ; 14(11): 3987-3997, 2017 11 06.
Article in English | MEDLINE | ID: mdl-28968488

ABSTRACT

Nucleotide analogues that incorporate a metabolically labile nucleoside phosphoramidate (a ProTide) have found utility as prodrugs. In humans, ProTides can be cleaved by human histidine triad nucleotide binding protein 1 (hHint1) to expose the nucleotide monophosphate. Activation by this route circumvents highly selective nucleoside kinases that limit the use of nucleosides as prodrugs. To better understand the diversity of potential substrates of hHint1, we created and studied a series of phosphoramidate nucleosides. Using a combination of enzyme kinetics, X-ray crystallography, and isothermal titration calorimetry with both wild-type and inactive mutant enzymes, we have been able to explore the energetics of substrate binding and establish a structural basis for catalytic efficiency. Diverse nucleobases are well tolerated, but portions of the ribose are needed to position substrates for catalysis. Beneficial characteristics of the amine leaving group are also revealed. Structural principles revealed by these results may be exploited to tune the rate of substrate hydrolysis to strategically alter the intracellular release of the product nucleoside monophosphate from the ProTide.


Subject(s)
Nerve Tissue Proteins/chemistry , Nucleotides/chemistry , Amides/chemistry , Crystallography, X-Ray , Humans , Phosphoric Acids/chemistry , Substrate Specificity
7.
Biochem Biophys Res Commun ; 491(3): 760-766, 2017 09 23.
Article in English | MEDLINE | ID: mdl-28739258

ABSTRACT

Human histidine triad nucleotide binding protein 1 (hHint1) is a purine nucleoside phosphoramidase and adenylate hydrolase that has emerged as a potential therapeutic target for the management of pain. However, the molecular mechanism of Hint1 in the signaling pathway has remained less clear. The role of metal ions in regulating postsynaptic transmission is well known, and the active site of hHint1 contains multiple histidines. Here we have investigated the effect of divalent metal ions (Cd2+, Cu2+, Mg2+, Mn2+, Ni2+, and Zn2+) on the structural integrity and catalytic activity of hHint1. With the exception of Mg2+, all the divalent ions inhibited hHint1, the rank of order was found to be Cu2+ >Zn2+ >Cd2+ ≥Ni2+ >Mn2+ based on their IC50 and kin/KI values. A crystal structure of hHint1 with bound Cu2+ is described to explain the competitive reversible inactivation of hHint1 by divalent cations. All the metal ions exhibited time- and concentration- dependent inhibition, with the rate of inactivation highly dependent on alterations of the C-terminus. With the exception of Cu2+; restoration of inhibition was observed for all the metal ions after treatment with EDTA. Our studies reveal a loss in secondary structure and aggregation of hHint1 upon incubation with 10-fold excess of copper. Thus, hHint1 appears to be structurally sensitive to irreversible inactivation by copper, which may be of neurotoxicological and pharmacological significance.


Subject(s)
Metals/chemistry , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/ultrastructure , Analgesics, Opioid/chemistry , Animals , Binding Sites , Catalysis , Cations, Divalent/chemistry , Copper/chemistry , Humans , Ions , Neuralgia/metabolism , Protein Binding , Protein Unfolding
8.
Biochemistry ; 56(28): 3559-3570, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28691797

ABSTRACT

Human histidine triad nucleotide binding protein 1 (hHint1) is classified as an efficient nucleoside phosphoramidase and acyl-adenosine monophosphate hydrolase. Human Hint1 has been shown to be essential for the metabolic activation of nucleotide antiviral pronucleotides (i.e., proTides), such as the FDA approved hepatitis C drug, sofosbuvir. The active site of hHint1 comprises an ensemble of strictly conserved histidines, including nucleophilic His112. To structurally investigate the mechanism of hHint1 catalysis, we have designed and prepared nucleoside thiophosphoramidate substrates that are able to capture the transiently formed nucleotidylated-His112 intermediate (E*) using time-dependent crystallography. Utilizing a catalytically inactive hHint1 His112Asn enzyme variant and wild-type enzyme, the enzyme-substrate (ES1) and product (EP2) complexes were also cocrystallized, respectively, thus providing a structural map of the reaction trajectory. On the basis of these observations and the mechanistic necessity of proton transfers, proton inventory studies were carried out. Although we cannot completely exclude the possibility of more than one proton in flight, the results of these studies were consistent with the transfer of a single proton during the formation of the intermediate. Interestingly, structural analysis revealed that the critical proton transfers required for intermediate formation and hydrolysis may be mediated by a conserved active site water channel. Taken together, our results provide mechanistic insights underpinning histidine nucleophilic catalysis in general and hHint1 catalysis, in particular, thus aiding the design of future proTides and the elucidation of the natural function of the Hint family of enzymes.


Subject(s)
Antiviral Agents/metabolism , Nerve Tissue Proteins/metabolism , Sofosbuvir/metabolism , Activation, Metabolic , Catalytic Domain , Crystallography, X-Ray , Humans , Kinetics , Models, Molecular , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Point Mutation , Substrate Specificity
9.
J Med Chem ; 60(13): 5507-5520, 2017 07 13.
Article in English | MEDLINE | ID: mdl-28594172

ABSTRACT

The pyridoxal 5'-phosphate (PLP)-dependent transaminase BioA catalyzes the second step in the biosynthesis of biotin in Mycobacterium tuberculosis (Mtb) and is an essential enzyme for bacterial survival and persistence in vivo. A promising BioA inhibitor 6 containing an N-aryl, N'-benzoylpiperazine scaffold was previously identified by target-based whole-cell screening. Here, we explore the structure-activity relationships (SAR) through the design, synthesis, and biological evaluation of a systematic series of analogues of the original hit using a structure-based drug design strategy, which was enabled by cocrystallization of several analogues with BioA. To confirm target engagement and discern analogues with off-target activity, each compound was evaluated against wild-type (WT) Mtb in biotin-free and -containing medium as well as BioA under- and overexpressing Mtb strains. Conformationally constrained derivative 36 emerged as the most potent analogue with a KD of 76 nM against BioA and a minimum inhibitory concentration of 1.7 µM (0.6 µg/mL) against Mtb in biotin-free medium.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Biotin/biosynthesis , Mycobacterium tuberculosis/drug effects , Piperazines/pharmacology , Pyridoxal Phosphate/metabolism , Transaminases/antagonists & inhibitors , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Bacterial Proteins/metabolism , Biocatalysis , Dose-Response Relationship, Drug , Microbial Sensitivity Tests , Molecular Structure , Mycobacterium tuberculosis/metabolism , Piperazines/chemical synthesis , Piperazines/chemistry , Structure-Activity Relationship , Transaminases/metabolism
10.
J Am Chem Soc ; 139(21): 7132-7135, 2017 05 31.
Article in English | MEDLINE | ID: mdl-28510452

ABSTRACT

Mechanism-based inhibitors (MBIs) are widely employed in chemistry, biology, and medicine because of their exquisite specificity and sustained duration of inhibition. Optimization of MBIs is complicated because of time-dependent inhibition resulting from multistep inactivation mechanisms. The global kinetic parameters kinact and KI have been used to characterize MBIs, but they provide far less information than is commonly assumed, as shown by derivation and simulation of these parameters. We illustrate an alternative and more rigorous approach for MBI characterization through determination of the individual microscopic rate constants. Kinetic analysis revealed the rate-limiting step of inactivation of the PLP-dependent enzyme BioA by dihydro-(1,4)-pyridone 1. This knowledge was subsequently applied to rationally design a second-generation inhibitor scaffold with a nearly optimal maximum inactivation rate (0.48 min-1).


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Mycobacterium tuberculosis/enzymology , Pyridones/pharmacology , Transaminases/antagonists & inhibitors , Bacterial Proteins/metabolism , Enzyme Inhibitors/chemistry , Kinetics , Molecular Structure , Pyridones/chemistry , Transaminases/metabolism
11.
ACS Med Chem Lett ; 7(8): 780-4, 2016 Aug 11.
Article in English | MEDLINE | ID: mdl-27563403

ABSTRACT

Hint1 has recently emerged to be an important target of interest due to its involvement in the regulation of a broad range of CNS functions including opioid signaling, tolerance, neuropathic pain, and nicotine dependence. A series of inhibitors were rationally designed, synthesized, and tested for their inhibitory activity against hHint1 using isothermal titration calorimetry (ITC). The studies resulted in the development of the first small-molecule inhibitors of hHint1 with submicromolar binding affinities. A combination of thermodynamic and high-resolution X-ray crystallographic studies provides an insight into the biomolecular recognition of ligands by hHint1. These novel inhibitors have potential utility as molecular probes to better understand the role and function of hHint1 in the CNS.

12.
FEBS Lett ; 589(24 Pt B): 3836-41, 2015 Dec 21.
Article in English | MEDLINE | ID: mdl-26578066

ABSTRACT

The Bacillus anthracis lethal factor (LF) is one component of a tripartite exotoxin partly responsible for persistent anthrax cytotoxicity after initial bacterial infection. Inhibitors of the zinc metalloproteinase have been investigated as potential therapeutic agents, but LF is a challenging target because inhibitors lack sufficient selectivity or possess poor pharmaceutical properties. These structural studies reveal an alternate conformation of the enzyme, induced upon binding of specific inhibitors, that opens a previously unobserved deep pocket termed S1'(∗) which might afford new opportunities to design selective inhibitors that target this subsite.


Subject(s)
Antigens, Bacterial/chemistry , Antigens, Bacterial/metabolism , Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , Matrix Metalloproteinase Inhibitors/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Bacterial Toxins/antagonists & inhibitors , Binding Sites , Ligands , Matrix Metalloproteinase Inhibitors/chemistry , Models, Molecular , Protein Conformation , Structure-Activity Relationship , Tyrosine/metabolism
13.
J Med Chem ; 58(21): 8723-33, 2015 Nov 12.
Article in English | MEDLINE | ID: mdl-26492514

ABSTRACT

The lethal factor (LF) enzyme secreted by Bacillus anthracis is a zinc hydrolase that is chiefly responsible for anthrax-related cell death. Although many studies of the design of small molecule LF inhibitors have been conducted, no LF inhibitor is yet available as a therapeutic agent. Inhibitors with considerable chemical diversity have been developed and investigated; however, the LF S2' subsite has not yet been systematically explored as a potential target for lead optimization. Here we present synthesis, experimental evaluation, modeling, and structural biology for a novel series of sulfonamide hydroxamate LF inhibitor analogues specifically designed to extend into, and probe chemical preferences of, this S2' subsite. We discovered that this region accommodates a wide variety of chemical functionalities and that a broad selection of ligand structural modifications directed to this area can be incorporated without significant deleterious alterations in biological activity. We also identified key residues in this subsite that can potentially be targeted to improve inhibitor binding.


Subject(s)
Anthrax/microbiology , Antigens, Bacterial/chemistry , Bacillus anthracis/enzymology , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/chemistry , Enzyme Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Alkylation , Anthrax/drug therapy , Antigens, Bacterial/metabolism , Bacillus anthracis/chemistry , Bacillus anthracis/metabolism , Bacterial Toxins/metabolism , Enzyme Inhibitors/chemistry , Humans , Hydroxamic Acids/chemistry , Models, Molecular , Structure-Activity Relationship
14.
J Med Chem ; 58(18): 7349-7369, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26299766

ABSTRACT

Mycobacterium tuberculosis (Mtb), responsible for both latent and symptomatic tuberculosis (TB), remains the second leading cause of mortality among infectious diseases worldwide. Mycobacterial biotin protein ligase (MtBPL) is an essential enzyme in Mtb and regulates lipid metabolism through the post-translational biotinylation of acyl coenzyme A carboxylases. We report the synthesis and evaluation of a systematic series of potent nucleoside-based inhibitors of MtBPL that contain modifications to the ribofuranosyl ring of the nucleoside. All compounds were characterized by isothermal titration calorimetry (ITC) and shown to bind potently with K(D)s ≤ 2 nM. Additionally, we obtained high-resolution cocrystal structures for a majority of the compounds. Despite fairly uniform biochemical potency, the whole-cell Mtb activity varied greatly with minimum inhibitory concentrations (MIC) ranging from 0.78 to >100 µM. Cellular accumulation studies showed a nearly 10-fold enhancement in accumulation of a C-2'-α analogue over the corresponding C-2'-ß analogue, consistent with their differential whole-cell activity.


Subject(s)
Antitubercular Agents/chemistry , Bacterial Proteins/antagonists & inhibitors , Carbon-Nitrogen Ligases/antagonists & inhibitors , Mycobacterium tuberculosis/enzymology , Nucleosides/chemistry , Antitubercular Agents/chemical synthesis , Antitubercular Agents/pharmacology , Bacterial Proteins/metabolism , Biotinylation , Carbon-Nitrogen Ligases/metabolism , Crystallography, X-Ray , Microbial Sensitivity Tests , Models, Molecular , Mycobacterium tuberculosis/drug effects , Nucleosides/chemical synthesis , Nucleosides/pharmacology , Protein Conformation , Stereoisomerism , Structure-Activity Relationship , Thermodynamics
15.
J Med Chem ; 58(13): 5208-17, 2015 Jul 09.
Article in English | MEDLINE | ID: mdl-26068403

ABSTRACT

The PLP-dependent transaminase (BioA) of Mycobacterium tuberculosis and other pathogens that catalyzes the second step of biotin biosynthesis is a now well-validated target for antibacterial development. Fragment screening by differential scanning fluorimetry has been performed to discover new chemical scaffolds and promote optimization of existing inhibitors. Calorimetry confirms binding of six molecules with high ligand efficiency. Thermodynamic data identifies which molecules bind with the enthalpy driven stabilization preferred in compounds that represent attractive starting points for future optimization. Crystallographic characterization of complexes with these molecules reveals the dynamic nature of the BioA active site. Different side chain conformational states are stabilized in response to binding by different molecules. A detailed analysis of conformational diversity in available BioA structures is presented, resulting in the identification of two states that might be targeted with molecular scaffolds incorporating well-defined conformational attributes. This new structural data can be used as part of a scaffold hopping strategy to further optimize existing inhibitors or create new small molecules with improved therapeutic potential.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Small Molecule Libraries/pharmacology , Transaminases/chemistry , Transaminases/metabolism , Tuberculosis/drug therapy , Binding Sites , Calorimetry, Differential Scanning , Catalytic Domain , Crystallography, X-Ray , Models, Molecular , Molecular Conformation , Mycobacterium tuberculosis/drug effects , Thermodynamics
16.
Chem Biol ; 22(1): 76-86, 2015 Jan 22.
Article in English | MEDLINE | ID: mdl-25556942

ABSTRACT

Biotin biosynthesis is essential for survival and persistence of Mycobacterium tuberculosis (Mtb) in vivo. The aminotransferase BioA, which catalyzes the antepenultimate step in the biotin pathway, has been established as a promising target due to its vulnerability to chemical inhibition. We performed high-throughput screening (HTS) employing a fluorescence displacement assay and identified a diverse set of potent inhibitors including many diversity-oriented synthesis (DOS) scaffolds. To efficiently select only hits targeting biotin biosynthesis, we then deployed a whole-cell counterscreen in biotin-free and biotin-containing medium against wild-type Mtb and in parallel with isogenic bioA Mtb strains that possess differential levels of BioA expression. This counterscreen proved crucial to filter out compounds whose whole-cell activity was off target as well as identify hits with weak, but measurable whole-cell activity in BioA-depleted strains. Several of the most promising hits were cocrystallized with BioA to provide a framework for future structure-based drug design efforts.


Subject(s)
Biotin/biosynthesis , Mycobacterium tuberculosis/metabolism , Antitubercular Agents/chemistry , Antitubercular Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Binding Sites , Biotin/antagonists & inhibitors , Calorimetry , Crystallography, X-Ray , Drug Design , High-Throughput Screening Assays , Hydrogen Bonding , Microbial Sensitivity Tests , Molecular Dynamics Simulation , Mycobacterium tuberculosis/drug effects , Protein Structure, Tertiary , Structure-Activity Relationship , Transaminases/antagonists & inhibitors , Transaminases/metabolism
17.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 11): 2813-22, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25372673

ABSTRACT

The secreted anthrax toxin consists of three components: the protective antigen (PA), edema factor (EF) and lethal factor (LF). LF, a zinc metalloproteinase, compromises the host immune system primarily by targeting mitogen-activated protein kinase kinases in macrophages. Peptide substrates and small-molecule inhibitors bind LF in the space between domains 3 and 4 of the hydrolase. Domain 3 is attached on a hinge to domain 2 via residues Ile300 and Pro385, and can move through an angular arc of greater than 35° in response to the binding of different ligands. Here, multiple LF structures including five new complexes with co-crystallized inhibitors are compared and three frequently populated LF conformational states termed `bioactive', `open' and `tight' are identified. The bioactive position is observed with large substrate peptides and leaves all peptide-recognition subsites open and accessible. The tight state is seen in unliganded and small-molecule complex structures. In this state, domain 3 is clamped over certain substrate subsites, blocking access. The open position appears to be an intermediate state between these extremes and is observed owing to steric constraints imposed by specific bound ligands. The tight conformation may be the lowest-energy conformation among the reported structures, as it is the position observed with no bound ligand, while the open and bioactive conformations are likely to be ligand-induced.


Subject(s)
Antigens, Bacterial/chemistry , Bacillus anthracis/chemistry , Bacterial Toxins/chemistry , Metalloendopeptidases/chemistry , Anthrax/microbiology , Antigens, Bacterial/metabolism , Bacillus anthracis/metabolism , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/metabolism , Binding Sites , Crystallography, X-Ray , Drug Design , Humans , Ligands , Metalloendopeptidases/metabolism , Models, Molecular , Peptides , Protein Conformation/drug effects , Protein Structure, Tertiary/drug effects , Sulfonamides/chemistry , Sulfonamides/pharmacology
18.
J Med Chem ; 57(6): 2714-25, 2014 Mar 27.
Article in English | MEDLINE | ID: mdl-24606063

ABSTRACT

Selective inhibitors of hyaluronan (HA) binding to the cell surface receptor CD44 will have value as probes of CD44-mediated signaling and have potential as therapeutic agents in chronic inflammation, cardiovascular disease, and cancer. Using biophysical binding assays, fragment screening, and crystallographic characterization of complexes with the CD44 HA binding domain, we have discovered an inducible pocket adjacent to the HA binding groove into which small molecules may bind. Iterations of fragment combination and structure-driven design have allowed identification of a series of 1,2,3,4-tetrahydroisoquinolines as the first nonglycosidic inhibitors of the CD44-HA interaction. The affinity of these molecules for the CD44 HA binding domain parallels their ability to interfere with CD44 binding to polymeric HA in vitro. X-ray crystallographic complexes of lead compounds are described and compared to a new complex with a short HA tetrasaccharide, to establish the tetrahydroisoquinoline pharmacophore as an attractive starting point for lead optimization.


Subject(s)
Carbohydrates , Hyaluronan Receptors/drug effects , Proteins/chemistry , Animals , Binding Sites , Crystallography, X-Ray , Drug Design , Drug Evaluation, Preclinical , Humans , Indicators and Reagents , Mice , Models, Molecular , Molecular Conformation , Polysaccharides/chemistry , Protein Binding/drug effects , Structure-Activity Relationship
19.
Chembiochem ; 15(4): 575-86, 2014 Mar 03.
Article in English | MEDLINE | ID: mdl-24482078

ABSTRACT

7,8-Diaminopelargonic acid synthase (BioA) of Mycobacterium tuberculosis is a recently validated target for therapeutic intervention in the treatment of tuberculosis (TB). Using biophysical fragment screening and structural characterization of compounds, we have identified a potent aryl hydrazine inhibitor of BioA that reversibly modifies the pyridoxal-5'-phosphate (PLP) cofactor, forming a stable quinonoid. Analogous hydrazides also form covalent adducts that can be observed crystallographically but are incapable of inactivating the enzyme. In the X-ray crystal structures, small molecules induce unexpected conformational remodeling in the substrate binding site. We compared these conformational changes to those induced upon binding of the substrate (7-keto-8-aminopelargonic acid), and characterized the inhibition kinetics and the X-ray crystal structures of BioA with the hydrazine compound and analogues to unveil the mechanism of this reversible covalent modification.


Subject(s)
Bacterial Proteins/metabolism , Hydrazines/metabolism , Mycobacterium tuberculosis/enzymology , Transaminases/metabolism , Amino Acids/chemistry , Amino Acids/metabolism , Bacterial Proteins/antagonists & inhibitors , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Hydrazines/chemistry , Kinetics , Molecular Docking Simulation , Transaminases/antagonists & inhibitors
20.
J Chem Inf Model ; 53(10): 2634-47, 2013 Oct 28.
Article in English | MEDLINE | ID: mdl-24047445

ABSTRACT

A software tool and workflow based on distance geometry is presented that can be used to search for local similarity in substructures in a comprehensive database of experimentally derived macromolecular structure. The method does not rely on fold annotation, specific secondary structure assignments, or sequence homology and may be used to locate compound substructures of multiple segments spanning different macromolecules that share a queried backbone geometry. This generalized substructure searching capability is intended to allow users to play an active part in exploring the role specific substructures play in larger protein domains, quaternary assemblies of proteins, and macromolecular complexes of proteins and polynucleotides. The user may select any portion or portions of an existing structure or complex to serve as a template for searching, and other structures that share the same structural features are identified, retrieved and overlaid to emphasize substructural likeness. Matching structures may be compared using a variety of integrated tools including molecular graphics for structure visualization and matching substructure sequence logos. A number of examples are provided that illustrate how generalized substructure searching may be used to understand both the similarity, and individuality of specific macromolecular structures. Web-based access to our substructure searching services is freely available at https://drugsite.msi.umn.edu.


Subject(s)
Algorithms , Ankyrins/chemistry , Cyclin-Dependent Kinase Inhibitor p16/chemistry , DNA/chemistry , Polynucleotides/chemistry , Software , Amino Acid Sequence , Databases, Chemical , Databases, Protein , Humans , Internet , Models, Molecular , Molecular Sequence Data , Protein Structure, Secondary , Protein Structure, Tertiary , Structural Homology, Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...