Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 4065, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38744895

ABSTRACT

Proteolysis-targeting chimeras (PROTACs) represent a new therapeutic modality involving selectively directing disease-causing proteins for degradation through proteolytic systems. Our ability to exploit targeted protein degradation (TPD) for antibiotic development remains nascent due to our limited understanding of which bacterial proteins are amenable to a TPD strategy. Here, we use a genetic system to model chemically-induced proximity and degradation to screen essential proteins in Mycobacterium smegmatis (Msm), a model for the human pathogen M. tuberculosis (Mtb). By integrating experimental screening of 72 protein candidates and machine learning, we find that drug-induced proximity to the bacterial ClpC1P1P2 proteolytic complex leads to the degradation of many endogenous proteins, especially those with disordered termini. Additionally, TPD of essential Msm proteins inhibits bacterial growth and potentiates the effects of existing antimicrobial compounds. Together, our results provide biological principles to select and evaluate attractive targets for future Mtb PROTAC development, as both standalone antibiotics and potentiators of existing antibiotic efficacy.


Subject(s)
Anti-Bacterial Agents , Bacterial Proteins , Mycobacterium smegmatis , Mycobacterium tuberculosis , Proteolysis , Proteolysis/drug effects , Mycobacterium smegmatis/drug effects , Mycobacterium smegmatis/metabolism , Mycobacterium smegmatis/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Anti-Bacterial Agents/pharmacology , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/metabolism , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/growth & development , Humans , Microbial Sensitivity Tests , Machine Learning
3.
J Biol Chem ; 298(11): 102553, 2022 11.
Article in English | MEDLINE | ID: mdl-36208775

ABSTRACT

The unfoldase ClpC1 is one of the most exciting drug targets against tuberculosis. This AAA+ unfoldase works in cooperation with the ClpP1P2 protease and is the target of at least four natural product antibiotics: cyclomarin, ecumicin, lassomycin, and rufomycin. Although these molecules are promising starting points for drug development, their mechanisms of action remain largely unknown. Taking advantage of a middle domain mutant, we determined the first structure of Mycobacterium tuberculosis ClpC1 in its apo, cyclomarin-, and ecumicin-bound states via cryo-EM. The obtained structure displays features observed in other members of the AAA+ family and provides a map for further drug development. While the apo and cyclomarin-bound structures are indistinguishable and have N-terminal domains that are invisible in their respective EM maps, around half of the ecumicin-bound ClpC1 particles display three of their six N-terminal domains in an extended conformation. Our structural observations suggest a mechanism where ecumicin functions by mimicking substrate binding, leading to ATPase activation and changes in protein degradation profile.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Humans , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Bacterial Proteins/metabolism , Mycobacterium tuberculosis/metabolism , Molecular Chaperones/metabolism
4.
J Bacteriol ; 203(4)2021 01 25.
Article in English | MEDLINE | ID: mdl-33229461

ABSTRACT

The ClpP1P2 proteolytic complex is essential in Mycobacterium tuberculosis Proteolysis by ClpP1P2 requires an associated ATPase, either ClpX or ClpC1. Here, we sought to define the unique contributions of the ClpX ATPase to mycobacterial growth. We formally demonstrated that ClpX is essential for mycobacterial growth, and to understand its essential functions, we identified ClpX-His-interacting proteins by pulldown and tandem mass spectrometry. We found an unexpected association between ClpX and proteins involved in DNA replication, and we confirm a physical association between ClpX and the essential DNA maintenance protein single-stranded-DNA binding protein (SSB). Purified SSB is not degraded by ClpXP1P2; instead, SSB enhances ATP hydrolysis by ClpX and degradation of the model substrate GFP-SsrA by ClpXP1P2. This activation of ClpX is mediated by the C-terminal tail of SSB, which had been implicated in the activation of other ATPases associated with DNA replication. Consistent with the predicted interactions, depletion of clpX transcript perturbs DNA replication. These data reveal that ClpX participates in DNA replication and identify the first activator of ClpX in mycobacteria.IMPORTANCE Tuberculosis, caused by Mycobacterium tuberculosis, imposes a major global health burden, surpassing HIV and malaria in annual deaths. The ClpP1P2 proteolytic complex and its cofactor ClpX are attractive drug targets, but their precise cellular functions are unclear. This work confirms ClpX's essentiality and describes a novel interaction between ClpX and SSB, a component of the DNA replication machinery. Further, we demonstrate that a loss of ClpX is sufficient to interrupt DNA replication, suggesting that the ClpX-SSB complex may play a role in DNA replication in mycobacteria.


Subject(s)
Bacterial Proteins/metabolism , Endopeptidase Clp/metabolism , Gene Expression Regulation, Bacterial/physiology , Gene Expression Regulation, Enzymologic/physiology , Mycobacterium tuberculosis/enzymology , Adenosine Triphosphatases/metabolism , Binding Sites , DNA Replication , DNA, Bacterial , DNA-Binding Proteins , Endopeptidase Clp/genetics , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Protein Binding
5.
Nat Commun ; 11(1): 1661, 2020 04 03.
Article in English | MEDLINE | ID: mdl-32245967

ABSTRACT

Pyrazinamide is a sterilizing first-line tuberculosis drug. Genetic, metabolomic and biophysical analyses previously demonstrated that pyrazinoic acid, the bioactive form of the prodrug pyrazinamide (PZA), interrupts biosynthesis of coenzyme A in Mycobacterium tuberculosis by binding to aspartate decarboxylase PanD. While most drugs act by inhibiting protein function upon target binding, we find here that pyrazinoic acid is only a weak enzyme inhibitor. We show that binding of pyrazinoic acid to PanD triggers degradation of the protein by the caseinolytic protease ClpC1-ClpP. Thus, the old tuberculosis drug pyrazinamide exerts antibacterial activity by acting as a target degrader, a mechanism of action that has recently emerged as a successful strategy in drug discovery across disease indications. Our findings provide the basis for the rational discovery of next generation PZA.


Subject(s)
Antitubercular Agents/pharmacology , Carboxy-Lyases/antagonists & inhibitors , Mycobacterium tuberculosis/drug effects , Proteolysis/drug effects , Pyrazinamide/analogs & derivatives , Antitubercular Agents/therapeutic use , Bacterial Proteins/metabolism , Carboxy-Lyases/genetics , Carboxy-Lyases/metabolism , Drug Resistance, Bacterial/genetics , Endopeptidase Clp/metabolism , Heat-Shock Proteins/metabolism , Humans , Microbial Sensitivity Tests , Mutation , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Pyrazinamide/pharmacology , Pyrazinamide/therapeutic use , Tuberculosis/drug therapy , Tuberculosis/microbiology
6.
Anal Biochem ; 567: 30-37, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30543804

ABSTRACT

Tuberculosis affects about 100 million people worldwide and causes nearly 2 million deaths annually. It has been estimated that one third of all humans is infected with latent Mycobacterium tuberculosis (Mtb). Moreover, Mtb has become increasingly resistant to available antibiotics. Consequently, it is important to identify and characterize new therapeutic targets in Mtb and to synthesize selective inhibitors. ClpP1, ClpP2 and their associated regulatory ATPases, ClpX and ClpC1 are required for the growth of Mtb and for its virulence during murine infection and are highly attractive drug targets, especially since they are not present in the cytosol of mammalian cells, and they differ markedly from the mitochondrial ClpP complex. The importance of these proteins in Mtb is emphasized by the existence of several natural antibiotics targeting this system. In order to find new inhibitors of ClpC1P1P2 system, we developed an assay based on the ATP-dependent degradation of a fluorescent protein substrate. The hits obtained were further characterized with a set of secondary assays to identify precise targets within a complex. A large library of compounds was screened and led to the identification of a ClpC1 ATPase inhibitor demonstrating that this approach can be used in future searches for anti-TB agents.


Subject(s)
Antitubercular Agents/chemistry , Bacterial Proteins/antagonists & inhibitors , Heat-Shock Proteins/antagonists & inhibitors , Mycobacterium tuberculosis/metabolism , Serine Proteinase Inhibitors/chemistry , Antitubercular Agents/metabolism , Antitubercular Agents/pharmacology , Bacterial Proteins/metabolism , Cell Survival/drug effects , Heat-Shock Proteins/metabolism , Hep G2 Cells , High-Throughput Screening Assays , Humans , Mycobacterium tuberculosis/drug effects , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/metabolism , Serine Proteinase Inhibitors/pharmacology
7.
Angew Chem Int Ed Engl ; 57(1): 348-353, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29067779

ABSTRACT

The spread of antibiotic resistance is a major challenge for the treatment of Mycobacterium tuberculosis infections. In addition, the efficacy of drugs is often limited by the restricted permeability of the mycomembrane. Frontline antibiotics inhibit mycomembrane biosynthesis, leading to rapid cell death. Inspired by this mechanism, we exploited ß-lactones as putative mycolic acid mimics to block serine hydrolases involved in their biosynthesis. Among a collection of ß-lactones, we found one hit with potent anti-mycobacterial and bactericidal activity. Chemical proteomics using an alkynylated probe identified Pks13 and Ag85 serine hydrolases as major targets. Validation through enzyme assays and customized 13 C metabolite profiling showed that both targets are functionally impaired by the ß-lactone. Co-administration with front-line antibiotics enhanced the potency against M. tuberculosis by more than 100-fold, thus demonstrating the therapeutic potential of targeting mycomembrane biosynthesis serine hydrolases.


Subject(s)
Antitubercular Agents/pharmacology , Lactones/pharmacology , Mycobacterium tuberculosis/drug effects , Mycolic Acids/antagonists & inhibitors , Acyltransferases/drug effects , Antigens, Bacterial/drug effects , Bacterial Proteins/drug effects , Cell Membrane Permeability/drug effects , Microbial Sensitivity Tests , Mycobacterium tuberculosis/metabolism , Mycolic Acids/metabolism , Polyketide Synthases/drug effects
8.
Mol Microbiol ; 101(2): 194-209, 2016 07.
Article in English | MEDLINE | ID: mdl-26919556

ABSTRACT

The Clp protease complex in Mycobacterium tuberculosis is unusual in its composition, functional importance and activation mechanism. Whilst most bacterial species contain a single ClpP protein that is dispensable for normal growth, mycobacteria have two ClpPs, ClpP1 and ClpP2, which are essential for viability and together form the ClpP1P2 tetradecamer. Acyldepsipeptide antibiotics of the ADEP class inhibit the growth of Gram-positive firmicutes by activating ClpP and causing unregulated protein degradation. Here we show that, in contrast, mycobacteria are killed by ADEP through inhibition of ClpP function. Although ADEPs can stimulate purified M. tuberculosis ClpP1P2 to degrade larger peptides and unstructured proteins, this effect is weaker than for ClpP from other bacteria and depends on the presence of an additional activating factor (e.g. the dipeptide benzyloxycarbonyl-leucyl-leucine in vitro) to form the active ClpP1P2 tetradecamer. The cell division protein FtsZ, which is a particularly sensitive target for ADEP-activated ClpP in firmicutes, is not degraded in mycobacteria. Depletion of the ClpP1P2 level in a conditional Mycobacterium bovis BCG mutant enhanced killing by ADEP unlike in other bacteria. In summary, ADEPs kill mycobacteria by preventing interaction of ClpP1P2 with the regulatory ATPases, ClpX or ClpC1, thus inhibiting essential ATP-dependent protein degradation.


Subject(s)
Depsipeptides/therapeutic use , Endopeptidase Clp/drug effects , Endopeptidase Clp/metabolism , Adenosine Triphosphatases/metabolism , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/drug effects , Bacterial Proteins/metabolism , Depsipeptides/chemistry , Depsipeptides/pharmacology , Endopeptidase Clp/physiology , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/metabolism , Peptide Hydrolases/metabolism , Proteolysis , Serine Endopeptidases/metabolism
9.
J Biol Chem ; 291(14): 7465-76, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26858247

ABSTRACT

The ClpP protease complex and its regulatory ATPases, ClpC1 and ClpX, inMycobacterium tuberculosis(Mtb) are essential and, therefore, promising drug targets. TheMtbClpP protease consists of two heptameric rings, one composed of ClpP1 and the other of ClpP2 subunits. Formation of the enzymatically active ClpP1P2 complex requires binding of N-blocked dipeptide activators. We have found a new potent activator, benzoyl-leucine-leucine (Bz-LL), that binds with higher affinity and promotes 3-4-fold higher peptidase activity than previous activators. Bz-LL-activated ClpP1P2 specifically stimulates the ATPase activity ofMtbClpC1 and ClpX. The ClpC1P1P2 and ClpXP1P2 complexes exhibit 2-3-fold enhanced ATPase activity, peptide cleavage, and ATP-dependent protein degradation. The crystal structure of ClpP1P2 with bound Bz-LL was determined at a resolution of 3.07 Å and with benzyloxycarbonyl-Leu-Leu (Z-LL) bound at 2.9 Å. Bz-LL was present in all 14 active sites, whereas Z-LL density was not resolved. Surprisingly, Bz-LL adopts opposite orientations in ClpP1 and ClpP2. In ClpP1, Bz-LL binds with the C-terminal leucine side chain in the S1 pocket. One C-terminal oxygen is close to the catalytic serine, whereas the other contacts backbone amides in the oxyanion hole. In ClpP2, Bz-LL binds with the benzoyl group in the S1 pocket, and the peptide hydrogen bonded between parallel ß-strands. The ClpP2 axial loops are extended, forming an open axial channel as has been observed with bound ADEP antibiotics. Thus occupancy of the active sites of ClpP allosterically alters sites on the surfaces thereby affecting the association of ClpP1 and ClpP2 rings, interactions with regulatory ATPases, and entry of protein substrates.


Subject(s)
Bacterial Proteins/chemistry , Dipeptides/chemistry , Multienzyme Complexes/chemistry , Mycobacterium tuberculosis/enzymology , Serine Endopeptidases/chemistry , Allosteric Regulation , Bacterial Proteins/metabolism , Binding Sites , Dipeptides/metabolism , Multienzyme Complexes/metabolism , Protein Binding , Protein Structure, Quaternary , Protein Structure, Secondary , Serine Endopeptidases/metabolism
10.
J Biol Chem ; 290(17): 11008-20, 2015 Apr 24.
Article in English | MEDLINE | ID: mdl-25759383

ABSTRACT

The ClpP1P2 protease complex is essential for viability in Mycobacteria tuberculosis and is an attractive drug target. Using a fluorogenic tripeptide library (Ac-X3X2X1-aminomethylcoumarin) and by determining specificity constants (kcat/Km), we show that ClpP1P2 prefers Met ≫ Leu > Phe > Ala in the X1 position, basic residues or Trp in the X2 position, and Pro ≫ Ala > Trp in the X3 position. We identified peptide substrates that are hydrolyzed up to 1000 times faster than the standard ClpP substrate. These positional preferences were consistent with cleavage sites in the protein GFPssrA by ClpXP1P2. Studies of ClpP1P2 with inactive ClpP1 or ClpP2 indicated that ClpP1 was responsible for nearly all the peptidase activity, whereas both ClpP1 and ClpP2 contributed to protein degradation. Substrate-based peptide boronates were synthesized that inhibit ClpP1P2 peptidase activity in the submicromolar range. Some of them inhibited the growth of Mtb cells in the low micromolar range indicating that cleavage specificity of Mtb ClpP1P2 can be used to design novel anti-bacterial agents.


Subject(s)
Anti-Bacterial Agents/chemistry , Bacterial Proteins/antagonists & inhibitors , Boronic Acids/chemistry , Multienzyme Complexes/antagonists & inhibitors , Mycobacterium tuberculosis/enzymology , Oligopeptides/chemistry , Peptide Library , Serine Proteinase Inhibitors/chemistry , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Boronic Acids/pharmacology , Dose-Response Relationship, Drug , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , Mycobacterium tuberculosis/growth & development , Oligopeptides/pharmacology , Serine Endopeptidases/chemistry , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/pharmacology
11.
Antimicrob Agents Chemother ; 59(2): 880-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25421483

ABSTRACT

Drug-resistant tuberculosis (TB) has lent urgency to finding new drug leads with novel modes of action. A high-throughput screening campaign of >65,000 actinomycete extracts for inhibition of Mycobacterium tuberculosis viability identified ecumicin, a macrocyclic tridecapeptide that exerts potent, selective bactericidal activity against M. tuberculosis in vitro, including nonreplicating cells. Ecumicin retains activity against isolated multiple-drug-resistant (MDR) and extensively drug-resistant (XDR) strains of M. tuberculosis. The subcutaneous administration to mice of ecumicin in a micellar formulation at 20 mg/kg body weight resulted in plasma and lung exposures exceeding the MIC. Complete inhibition of M. tuberculosis growth in the lungs of mice was achieved following 12 doses at 20 or 32 mg/kg. Genome mining of lab-generated, spontaneous ecumicin-resistant M. tuberculosis strains identified the ClpC1 ATPase complex as the putative target, and this was confirmed by a drug affinity response test. ClpC1 functions in protein breakdown with the ClpP1P2 protease complex. Ecumicin markedly enhanced the ATPase activity of wild-type (WT) ClpC1 but prevented activation of proteolysis by ClpC1. Less stimulation was observed with ClpC1 from ecumicin-resistant mutants. Thus, ClpC1 is a valid drug target against M. tuberculosis, and ecumicin may serve as a lead compound for anti-TB drug development.


Subject(s)
Antitubercular Agents/therapeutic use , Mycobacterium tuberculosis/drug effects , Peptides, Cyclic/therapeutic use , Tuberculosis/drug therapy , Animals , Antitubercular Agents/pharmacology , Caco-2 Cells , Humans , Male , Mice , Microbial Sensitivity Tests , Mycobacterium tuberculosis/pathogenicity , Peptides, Cyclic/pharmacology
12.
Chem Biol ; 21(4): 509-518, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24684906

ABSTRACT

Languishing antibiotic discovery and flourishing antibiotic resistance have prompted the development of alternative untapped sources for antibiotic discovery, including previously uncultured bacteria. Here, we screen extracts from uncultured species against Mycobacterium tuberculosis and identify lassomycin, an antibiotic that exhibits potent bactericidal activity against both growing and dormant mycobacteria, including drug-resistant forms of M. tuberculosis, but little activity against other bacteria or mammalian cells. Lassomycin is a highly basic, ribosomally encoded cyclic peptide with an unusual structural fold that only partially resembles that of other lasso peptides. We show that lassomycin binds to a highly acidic region of the ClpC1 ATPase complex and markedly stimulates its ATPase activity without stimulating ClpP1P2-catalyzed protein breakdown, which is essential for viability of mycobacteria. This mechanism, uncoupling ATPase from proteolytic activity, accounts for the bactericidal activity of lassomycin.


Subject(s)
ATP-Dependent Proteases/antagonists & inhibitors , Anti-Bacterial Agents/pharmacology , Mycobacterium tuberculosis/drug effects , Peptides, Cyclic/pharmacology , Protease Inhibitors/pharmacology , ATP-Dependent Proteases/metabolism , Amino Acid Sequence , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/isolation & purification , Dose-Response Relationship, Drug , Models, Molecular , Molecular Sequence Data , Mycobacterium tuberculosis/enzymology , Peptides, Cyclic/chemistry , Peptides, Cyclic/isolation & purification , Protease Inhibitors/chemistry , Protease Inhibitors/isolation & purification , Structure-Activity Relationship
13.
PLoS Pathog ; 8(2): e1002511, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22359499

ABSTRACT

In most bacteria, Clp protease is a conserved, non-essential serine protease that regulates the response to various stresses. Mycobacteria, including Mycobacterium tuberculosis (Mtb) and Mycobacterium smegmatis, unlike most well studied prokaryotes, encode two ClpP homologs, ClpP1 and ClpP2, in a single operon. Here we demonstrate that the two proteins form a mixed complex (ClpP1P2) in mycobacteria. Using two different approaches, promoter replacement, and a novel system of inducible protein degradation, leading to inducible expression of clpP1 and clpP2, we demonstrate that both genes are essential for growth and that a marked depletion of either one results in rapid bacterial death. ClpP1P2 protease appears important in degrading missense and prematurely terminated peptides, as partial depletion of ClpP2 reduced growth specifically in the presence of antibiotics that increase errors in translation. We further show that the ClpP1P2 protease is required for the degradation of proteins tagged with the SsrA motif, a tag co-translationally added to incomplete protein products. Using active site mutants of ClpP1 and ClpP2, we show that the activity of each subunit is required for proteolysis, for normal growth of Mtb in vitro and during infection of mice. These observations suggest that the Clp protease plays an unusual and essential role in Mtb and may serve as an ideal target for antimycobacterial therapy.


Subject(s)
Bacterial Proteins/metabolism , Microbial Viability , Mycobacterium tuberculosis/physiology , Serine Endopeptidases/metabolism , Tuberculosis/metabolism , Animals , Mice , Mice, Inbred C57BL , Proteolysis , Tuberculosis/genetics
14.
EMBO J ; 31(6): 1529-41, 2012 Mar 21.
Article in English | MEDLINE | ID: mdl-22286948

ABSTRACT

Mycobacterium tuberculosis (Mtb) contains two clpP genes, both of which are essential for viability. We expressed and purified Mtb ClpP1 and ClpP2 separately. Although each formed a tetradecameric structure and was processed, they lacked proteolytic activity. We could, however, reconstitute an active, mixed ClpP1P2 complex after identifying N-blocked dipeptides that stimulate dramatically (>1000-fold) ClpP1P2 activity against certain peptides and proteins. These activators function cooperatively to induce the dissociation of ClpP1 and ClpP2 tetradecamers into heptameric rings, which then re-associate to form the active ClpP1P2 2-ring mixed complex. No analogous small molecule-induced enzyme activation mechanism involving dissociation and re-association of multimeric rings has been described. ClpP1P2 possesses chymotrypsin and caspase-like activities, and ClpP1 and ClpP2 differ in cleavage preferences. The regulatory ATPase ClpC1 was purified and shown to increase hydrolysis of proteins by ClpP1P2, but not peptides. ClpC1 did not activate ClpP1 or ClpP2 homotetradecamers and stimulated ClpP1P2 only when both ATP and a dipeptide activator were present. ClpP1P2 activity, its unusual activation mechanism and ClpC1 ATPase represent attractive drug targets to combat tuberculosis.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Mycobacterium tuberculosis/enzymology , Serine Endopeptidases/chemistry , Serine Endopeptidases/metabolism , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Bacterial Proteins/genetics , Binding Sites , Caspases/metabolism , Chymotrypsin/metabolism , Hydrolysis , Mycobacterium tuberculosis/genetics , Peptide Hydrolases/chemistry , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , Peptides/metabolism , Protein Conformation , Serine Endopeptidases/genetics
15.
Peptides ; 27(7): 1877-85, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16574278

ABSTRACT

Beta-secretase 1 (BACE1) is an aspartic protease believed to play a critical role in Alzheimer's disease. Inhibitors of this enzyme have been designed by incorporating the non-cleavable hydroxyethylene and statine isosteres into peptides corresponding to BACE1 substrate sequences. We sought to develop new methods to quickly characterize and optimize inhibitors based on the statine core. Minimal sequence requirements for binding were first established using both crystallography and peptide spot synthesis. These shortened peptide inhibitors were then optimized by using spot synthesis to perform iterative cycles of substitution and deletion. The present study resulted in the identification of novel "bis-statine" inhibitors shown by crystallography to have a unique binding mode. Our results demonstrate the application of peptide spot synthesis as an effective method for enhancing peptidomimetic drug discovery.


Subject(s)
Amino Acids/chemistry , Biochemistry/methods , Endopeptidases/chemistry , Peptides/chemistry , Protease Inhibitors/pharmacology , Amino Acid Sequence , Amyloid Precursor Protein Secretases , Animals , Biotinylation , CHO Cells , Cricetinae , Crystallization , Crystallography , Models, Molecular , Molecular Sequence Data , Protein Conformation
16.
J Am Chem Soc ; 126(46): 15106-19, 2004 Nov 24.
Article in English | MEDLINE | ID: mdl-15548008

ABSTRACT

We present the structure-based optimization of a series of estrogen receptor-beta (ERbeta) selective ligands. X-ray cocrystal structures of these ligands complexed to both ERalpha and ERbeta are described. We also discuss how molecular modeling was used to take advantage of subtle differences between the two binding cavities in order to optimize selectivity for ERbeta over ERalpha. Quantum chemical calculations are utilized to gain insight into the mechanism of selectivity enhancement. Despite only two relatively conservative residue substitutions in the ligand binding pocket, the most selective compounds have greater than 100-fold selectivity for ERbeta relative to ERalpha when measured using a competitive radioligand binding assay.


Subject(s)
Estrogen Receptor beta/chemistry , Estrogen Receptor beta/metabolism , Amino Acid Sequence , Benzofurans/chemistry , Benzofurans/metabolism , Benzoxazoles/chemistry , Benzoxazoles/metabolism , Binding Sites , Binding, Competitive , Crystallography, X-Ray , Estrogen Receptor alpha/chemistry , Estrogen Receptor alpha/metabolism , Humans , Ligands , Male , Models, Molecular , Molecular Sequence Data , Protein Conformation , Quantum Theory , Radioligand Assay , Structure-Activity Relationship , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...