Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Cell Chem Biol ; 31(7): 1247-1263.e16, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-38537632

ABSTRACT

This study describes the identification and target deconvolution of small molecule inhibitors of oncogenic Yes-associated protein (YAP1)/TAZ activity with potent anti-tumor activity in vivo. A high-throughput screen (HTS) of 3.8 million compounds was conducted using a cellular YAP1/TAZ reporter assay. Target deconvolution studies identified the geranylgeranyltransferase-I (GGTase-I) complex as the direct target of YAP1/TAZ pathway inhibitors. The small molecule inhibitors block the activation of Rho-GTPases, leading to subsequent inactivation of YAP1/TAZ and inhibition of cancer cell proliferation in vitro. Multi-parameter optimization resulted in BAY-593, an in vivo probe with favorable PK properties, which demonstrated anti-tumor activity and blockade of YAP1/TAZ signaling in vivo.


Subject(s)
Adaptor Proteins, Signal Transducing , Antineoplastic Agents , Cell Proliferation , High-Throughput Screening Assays , Signal Transduction , Transcription Factors , YAP-Signaling Proteins , Humans , Transcription Factors/metabolism , Transcription Factors/antagonists & inhibitors , YAP-Signaling Proteins/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Signal Transduction/drug effects , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Animals , Cell Proliferation/drug effects , Mice , rho GTP-Binding Proteins/metabolism , rho GTP-Binding Proteins/antagonists & inhibitors , Cell Line, Tumor , Phosphoproteins/metabolism , Phosphoproteins/antagonists & inhibitors , Drug Screening Assays, Antitumor , Alkyl and Aryl Transferases/antagonists & inhibitors , Alkyl and Aryl Transferases/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Drug Discovery , Mice, Nude , Acyltransferases/antagonists & inhibitors , Acyltransferases/metabolism , Phenotype , Structure-Activity Relationship , Transcriptional Coactivator with PDZ-Binding Motif Proteins
2.
J Med Chem ; 67(2): 1225-1242, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38228402

ABSTRACT

Interleukin-1 receptor-associated kinase 4 (IRAK4) plays a critical role in innate inflammatory processes. Here, we describe the discovery of two clinical candidate IRAK4 inhibitors, BAY1834845 (zabedosertib) and BAY1830839, starting from a high-throughput screening hit derived from Bayer's compound library. By exploiting binding site features distinct to IRAK4 using an in-house docking model, liabilities of the original hit could surprisingly be overcome to confer both candidates with a unique combination of good potency and selectivity. Favorable DMPK profiles and activity in animal inflammation models led to the selection of these two compounds for clinical development in patients.


Subject(s)
High-Throughput Screening Assays , Indazoles , Interleukin-1 Receptor-Associated Kinases , Pyridines , Animals , Humans , Binding Sites , Inflammation
3.
J Med Chem ; 65(24): 16420-16431, 2022 12 22.
Article in English | MEDLINE | ID: mdl-36475653

ABSTRACT

Despite advances in the treatment of heart failure in recent years, options for patients are still limited and the disease is associated with considerable morbidity and mortality. Modulating cyclic guanosine monophosphate levels within the natriuretic peptide signaling pathway by inhibiting PDE9A has been associated with beneficial effects in preclinical heart failure models. We herein report the identification of BAY-7081, a potent, selective, and orally bioavailable PDE9A inhibitor with very good aqueous solubility starting from a high-throughput screening hit. Key aspect of the optimization was a switch in metabolism of our lead structures from glucuronidation to oxidation. The switch proved being essential for the identification of compounds with improved pharmacokinetic profiles. By studying a tool compound in a transverse aortic constriction mouse model, we were able to substantiate the relevance of PDE9A inhibition in heart diseases.


Subject(s)
Cyclic GMP , Heart Failure , Mice , Animals , Cyclic GMP/metabolism , High-Throughput Screening Assays , 3',5'-Cyclic-AMP Phosphodiesterases
4.
SLAS Discov ; 26(8): 947-960, 2021 09.
Article in English | MEDLINE | ID: mdl-34154424

ABSTRACT

SMYD3 (SET and MYND domain-containing protein 3) is a protein lysine methyltransferase that was initially described as an H3K4 methyltransferase involved in transcriptional regulation. SMYD3 has been reported to methylate and regulate several nonhistone proteins relevant to cancer, including mitogen-activated protein kinase kinase kinase 2 (MAP3K2), vascular endothelial growth factor receptor 1 (VEGFR1), and the human epidermal growth factor receptor 2 (HER2). In addition, overexpression of SMYD3 has been linked to poor prognosis in certain cancers, suggesting SMYD3 as a potential oncogene and attractive cancer drug target. Here we report the discovery of a novel SMYD3 inhibitor. We performed a thermal shift assay (TSA)-based high-throughput screening (HTS) with 410,000 compounds and identified a novel benzodiazepine-based SMYD3 inhibitor series. Crystal structures revealed that this series binds to the substrate binding site and occupies the hydrophobic lysine binding pocket via an unprecedented hydrogen bonding pattern. Biochemical assays showed substrate competitive behavior. Following optimization and extensive biophysical validation with surface plasmon resonance (SPR) analysis and isothermal titration calorimetry (ITC), we identified BAY-6035, which shows nanomolar potency and selectivity against kinases and other PKMTs. Furthermore, BAY-6035 specifically inhibits methylation of MAP3K2 by SMYD3 in a cellular mechanistic assay with an IC50 <100 nM. Moreover, we describe a congeneric negative control to BAY-6035. In summary, BAY-6035 is a novel selective and potent SMYD3 inhibitor probe that will foster the exploration of the biological role of SMYD3 in diseased and nondiseased tissues.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Discovery/methods , High-Throughput Screening Assays/methods , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/chemistry , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Protein Binding , Small Molecule Libraries , Structure-Activity Relationship
5.
ChemMedChem ; 16(7): 1116-1125, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33513288

ABSTRACT

Protein arginine N-methyl transferase 4 (PRMT4) asymmetrically dimethylates the arginine residues of histone H3 and nonhistone proteins. The overexpression of PRMT4 in several cancers has stimulated interest in the discovery of inhibitors as biological tools and, potentially, therapeutics. Although several PRMT4 inhibitors have been reported, most display poor selectivity against other members of the PRMT family of methyl transferases. Herein, we report the structure-based design of a new class of alanine-containing 3-arylindoles as potent and selective PRMT4 inhibitors, and describe key structure-activity relationships for this class of compounds.


Subject(s)
Alanine/pharmacology , Antineoplastic Agents/pharmacology , Drug Design , Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Neoplasms/drug therapy , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Alanine/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , HEK293 Cells , Humans , Indoles/chemical synthesis , Indoles/chemistry , Molecular Structure , Neoplasms/metabolism , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Structure-Activity Relationship
6.
iScience ; 23(9): 101517, 2020 Sep 25.
Article in English | MEDLINE | ID: mdl-32927263

ABSTRACT

Structural mutants of p53 induce global p53 protein destabilization and misfolding, followed by p53 protein aggregation. First evidence indicates that p53 can be part of protein condensates and that p53 aggregation potentially transitions through a condensate-like state. We show condensate-like states of fluorescently labeled structural mutant p53 in the nucleus of living cancer cells. We furthermore identified small molecule compounds that interact with the p53 protein and lead to dissolution of p53 structural mutant condensates. The same compounds lead to condensation of a fluorescently tagged p53 DNA-binding mutant, indicating that the identified compounds differentially alter p53 condensation behavior depending on the type of p53 mutation. In contrast to p53 aggregation inhibitors, these compounds are active on p53 condensates and do not lead to mutant p53 reactivation. Taken together our study provides evidence for structural mutant p53 condensation in living cells and tools to modulate this process.

7.
Int J Cancer ; 145(5): 1382-1394, 2019 09 01.
Article in English | MEDLINE | ID: mdl-30828788

ABSTRACT

Darolutamide is a novel androgen receptor (AR) antagonist with a distinct chemical structure compared to other AR antagonists and currently in clinical Phase 3 trials for prostate cancer. Using cell-based transactivation assays, we demonstrate that darolutamide, its diastereomers and its main metabolite keto-darolutamide are strong, competitive antagonists for AR wild type, and also for several mutants identified in prostate cancer patients for which other AR antagonists show reduced antagonism or even agonism. Darolutamide, its two diastereomers and main metabolite are also strong antagonists in assays measuring AR N/C interaction and homodimerization. Molecular modeling suggests that the flexibility of darolutamide allows accommodation in the W742C/L mutated AR ligand-binding pocket while for enzalutamide the loss of the important hydrophobic interaction with W742 leads to reduced AR interaction. This correlates with an antagonistic pattern profile of coregulator recruitment for darolutamide. In vitro efficacy studies performed with androgen-dependent prostate cancer cell lines show that darolutamide strongly reduces cell viability and potently inhibits spheroid formation. Also, a marked down-regulation of androgen target genes paralleled by decreased AR binding to gene regulatory regions is seen. In vivo studies reveal that oral dosing of darolutamide markedly reduces growth of the LAPC-4 cell line-derived xenograft and of the KuCaP-1 patient-derived xenograft. Altogether, these results substantiate a unique antagonistic profile of darolutamide and support further development as a prostate cancer drug.


Subject(s)
Androgen Receptor Antagonists/pharmacology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/metabolism , Pyrazoles/pharmacology , Receptors, Androgen/metabolism , Androgen Receptor Antagonists/chemistry , Animals , Cell Line, Tumor , Gene Expression/drug effects , Humans , Male , Mice , Mice, SCID , Models, Molecular , Prostatic Neoplasms, Castration-Resistant/genetics , Protein Domains , Pyrazoles/chemistry , Receptors, Androgen/chemistry , Receptors, Androgen/genetics , Xenograft Model Antitumor Assays
8.
Nat Chem Biol ; 12(7): 539-45, 2016 07.
Article in English | MEDLINE | ID: mdl-27214403

ABSTRACT

Members of the KDM5 (also known as JARID1) family are 2-oxoglutarate- and Fe(2+)-dependent oxygenases that act as histone H3K4 demethylases, thereby regulating cell proliferation and stem cell self-renewal and differentiation. Here we report crystal structures of the catalytic core of the human KDM5B enzyme in complex with three inhibitor chemotypes. These scaffolds exploit several aspects of the KDM5 active site, and their selectivity profiles reflect their hybrid features with respect to the KDM4 and KDM6 families. Whereas GSK-J1, a previously identified KDM6 inhibitor, showed about sevenfold less inhibitory activity toward KDM5B than toward KDM6 proteins, KDM5-C49 displayed 25-100-fold selectivity between KDM5B and KDM6B. The cell-permeable derivative KDM5-C70 had an antiproliferative effect in myeloma cells, leading to genome-wide elevation of H3K4me3 levels. The selective inhibitor GSK467 exploited unique binding modes, but it lacked cellular potency in the myeloma system. Taken together, these structural leads deliver multiple starting points for further rational and selective inhibitor design.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Histone Demethylases/antagonists & inhibitors , Jumonji Domain-Containing Histone Demethylases/chemistry , Jumonji Domain-Containing Histone Demethylases/metabolism , Multiple Myeloma/drug therapy , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemistry , Histone Demethylases/metabolism , Humans , Models, Molecular , Multiple Myeloma/pathology , Protein Conformation , Structure-Activity Relationship
9.
J Med Chem ; 59(3): 1176-83, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26824386

ABSTRACT

Protein methyltransferases (PMTs) are a promising target class in oncology and other disease areas. They are composed of SET domain methyltransferases and structurally unrelated Rossman-fold enzymes that include protein arginine methyltransferases (PRMTs). In the absence of a well-defined medicinal chemistry tool-kit focused on PMTs, most current inhibitors were identified by screening large and diverse libraries of leadlike molecules. So far, no successful fragment-based approach was reported against this target class. Here, by deconstructing potent PRMT inhibitors, we find that chemical moieties occupying the substrate arginine-binding site can act as efficient fragment inhibitors. Screening a fragment library against PRMT6 produced numerous hits, including a 300 nM inhibitor (ligand efficiency of 0.56) that decreased global histone 3 arginine 2 methylation in cells, and can serve as a warhead for the development of PRMT chemical probes.


Subject(s)
Drug Discovery , Enzyme Inhibitors/pharmacology , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Repressor Proteins/antagonists & inhibitors , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Models, Molecular , Molecular Structure , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , Structure-Activity Relationship
10.
Bioorg Med Chem Lett ; 26(1): 60-7, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26614408

ABSTRACT

We have identified a class of azabenzimidazoles as potent and selective JAK1 inhibitors. Investigations into the SAR are presented along with the structural features required to achieve selectivity for JAK1 versus other JAK family members. An example from the series demonstrated highly selective inhibition of JAK1 versus JAK2 and JAK3, along with inhibition of pSTAT3 in vivo, enabling it to serve as a JAK1 selective tool compound to further probe the biology of JAK1 selective inhibitors.


Subject(s)
Imidazoles/pharmacology , Janus Kinase 1/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Cell Cycle/drug effects , Cell Death/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Female , Humans , Imidazoles/chemical synthesis , Imidazoles/chemistry , Janus Kinase 1/metabolism , Mice , Mice, Nude , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , STAT3 Transcription Factor/metabolism , Structure-Activity Relationship
11.
J Biol Chem ; 290(32): 19597-613, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-26105055

ABSTRACT

For host colonization, the human fungal pathogen Candida glabrata is known to utilize a large family of highly related surface-exposed cell wall proteins, the lectin-like epithelial adhesins (Epas). To reveal the structure-function relationships within the entire Epa family, we have performed a large scale functional analysis of the adhesion (A) domains of 17 Epa paralogs in combination with three-dimensional structural studies of selected members with cognate ligands. Our study shows that most EpaA domains exert lectin-like functions and together recognize a wide variety of glycans with terminal galactosides for conferring epithelial cell adhesion. We further identify several conserved and variable structural features within the diverse Epa ligand binding pockets, which affect affinity and specificity. These features rationalize why mere phylogenetic relationships within the Epa family are weak indicators for functional classification and explain how Epa-like adhesins have evolved in C. glabrata and related fungal species.


Subject(s)
Candida glabrata/chemistry , Cell Adhesion Molecules/chemistry , Fungal Proteins/chemistry , Lectins/chemistry , Polysaccharides/chemistry , Amino Acid Sequence , Binding Sites , Biological Evolution , Candida glabrata/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Gene Expression , Genetic Variation , Lectins/genetics , Lectins/metabolism , Models, Molecular , Molecular Sequence Data , Phylogeny , Polysaccharides/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment
12.
J Med Chem ; 58(11): 4845-50, 2015 Jun 11.
Article in English | MEDLINE | ID: mdl-26000468

ABSTRACT

3,4-disubstituted pyrrolidines originally designed to inhibit the closely related HIV-1 protease were evaluated as privileged structures against HTLV-1 protease (HTLV-1 PR). The most potent inhibitor of this series exhibits two-digit nanomolar affinity and represents, to the best of our knowledge, the most potent nonpeptidic inhibitor of HTLV-1 PR described so far. The X-ray structures of two representatives bound to HTLV-1 PR were determined, and the structural basis of their affinity is discussed.


Subject(s)
Aspartic Acid Endopeptidases/chemistry , HIV-1/drug effects , Human T-lymphotropic virus 1/drug effects , Protease Inhibitors/pharmacology , Pyrrolidines/pharmacology , Aspartic Acid Endopeptidases/metabolism , HIV-1/enzymology , Human T-lymphotropic virus 1/enzymology , Humans , Models, Molecular , Molecular Structure , Protease Inhibitors/chemistry , Protein Conformation , Pyrrolidines/chemistry , Structure-Activity Relationship
13.
Angew Chem Int Ed Engl ; 54(9): 2849-53, 2015 Feb 23.
Article in English | MEDLINE | ID: mdl-25630461

ABSTRACT

Successful lead optimization in structure-based drug discovery depends on the correct deduction and interpretation of the underlying structure-activity relationships (SAR) to facilitate efficient decision-making on the next candidates to be synthesized. Consequently, the question arises, how frequently a binding mode (re)-validation is required, to ensure not to be misled by invalid assumptions on the binding geometry. We present an example in which minor chemical modifications within one inhibitor series lead to surprisingly different binding modes. X-ray structure determination of eight inhibitors derived from one core scaffold resulted in four different binding modes in the aspartic protease endothiapepsin, a well-established surrogate for e.g. renin and ß-secretase. In addition, we suggest an empirical metrics that might serve as an indicator during lead optimization to qualify compounds as candidates for structural revalidation.


Subject(s)
Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Endopeptidases/pharmacology , Protease Inhibitors/pharmacology , Thiophenes/pharmacology , Aspartic Acid Endopeptidases/chemical synthesis , Aspartic Acid Endopeptidases/chemistry , Aspartic Acid Endopeptidases/metabolism , Binding Sites/drug effects , Dose-Response Relationship, Drug , Models, Molecular , Molecular Structure , Protease Inhibitors/chemical synthesis , Protease Inhibitors/chemistry , Structure-Activity Relationship , Thiophenes/chemical synthesis , Thiophenes/chemistry
14.
Antimicrob Agents Chemother ; 59(2): 1100-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25487800

ABSTRACT

Dengue fever is a severe, widespread, and neglected disease with more than 2 million diagnosed infections per year. The dengue virus NS2B/NS3 protease (PR) represents a prime target for rational drug design. At the moment, there are no clinical PR inhibitors (PIs) available. We have identified diaryl (thio)ethers as candidates for a novel class of PIs. Here, we report the selective and noncompetitive inhibition of the serotype 2 and 3 dengue virus PR in vitro and in cells by benzothiazole derivatives exhibiting 50% inhibitory concentrations (IC50s) in the low-micromolar range. Inhibition of replication of DENV serotypes 1 to 3 was specific, since all substances influenced neither hepatitis C virus (HCV) nor HIV-1 replication. Molecular docking suggests binding at a specific allosteric binding site. In addition to the in vitro assays, a cell-based PR assay was developed to test these substances in a replication-independent way. The new compounds inhibited the DENV PR with IC50s in the low-micromolar or submicromolar range in cells. Furthermore, these novel PIs inhibit viral replication at submicromolar concentrations.


Subject(s)
Dengue Virus/drug effects , Protease Inhibitors/pharmacology , Serine Endopeptidases/metabolism , Viral Nonstructural Proteins/metabolism , Antiviral Agents/pharmacology , Cell Line , Dengue Virus/enzymology , HIV-1/drug effects , Humans , Molecular Docking Simulation , Virus Replication/drug effects
15.
J Med Chem ; 57(14): 6266-72, 2014 Jul 24.
Article in English | MEDLINE | ID: mdl-25006983

ABSTRACT

HTLV-1 protease (HTLV-1 PR) is an aspartic protease which represents a promising drug target for the discovery of novel anti-HTLV-1 drugs. The X-ray structure of HTLV-1 PR in complex with the well-known and approved HIV-1 PR inhibitor Indinavir was determined at 2.40 Å resolution. In this contribution, we describe the first crystal structure in complex with a nonpeptidic inhibitor that accounts for rationalizing the rather moderate affinity of Indinavir against HTLV-1 PR and provides the basis for further structure-guided optimization strategies.


Subject(s)
Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Endopeptidases/chemistry , HIV Protease Inhibitors/chemistry , HIV Protease Inhibitors/pharmacology , Indinavir/chemistry , Indinavir/pharmacology , Aspartic Acid Endopeptidases/metabolism , Crystallography, X-Ray , Dose-Response Relationship, Drug , Models, Molecular , Molecular Structure , Structure-Activity Relationship
16.
Angew Chem Int Ed Engl ; 53(8): 2230-4, 2014 Feb 17.
Article in English | MEDLINE | ID: mdl-24446383

ABSTRACT

Lasso peptides belong to the class of ribosomally synthesized and post-translationally modified peptides. Their common distinguishing feature is an N-terminal macrolactam ring that is threaded by the C-terminal tail. This lasso fold is maintained through steric interactions. The isolation and characterization of xanthomonins I-III, the first lasso peptides featuring macrolactam rings consisting of only seven amino acids, is now presented. The crystal structure of xanthomonin I and the NMR structure of xanthomonin II were also determined. A total of 25 variants of xanthomonin II were generated to probe different aspects of the biosynthesis, stability, and fold maintenance. These mutational studies reveal the limits such a small ring imposes on the threading and show that every plug amino acid larger than serine is able to maintain a heat-stable lasso fold in the xanthomonin II scaffold.


Subject(s)
Lactams/chemistry , Peptides/metabolism , Rotaxanes/metabolism , Amino Acid Sequence , Crystallography, X-Ray , Magnetic Resonance Spectroscopy , Molecular Sequence Data , Multigene Family , Mutagenesis , Peptides/chemistry , Peptides/genetics , Protein Structure, Tertiary , Rotaxanes/chemistry , Xanthomonas/genetics , Xanthomonas/metabolism
17.
Angew Chem Int Ed Engl ; 53(1): 305-9, 2014 Jan 03.
Article in English | MEDLINE | ID: mdl-24258965

ABSTRACT

The probe-based discovery of the first small-molecule inhibitor of the repair enzyme 8-oxo-dGTPase (MTH1) is presented, which is an unconventional cyclometalated ruthenium half-sandwich complex. The organometallic inhibitor with low-nanomolar activity displays astonishing specificity, as verified in tests with an extended panel of protein kinases and other ATP binding proteins. The binding of the organometallic inhibitor to MTH1 is investigated by protein crystallography.


Subject(s)
DNA Repair Enzymes/chemistry , Phosphoric Monoester Hydrolases/chemistry , Ruthenium/chemistry , DNA Repair Enzymes/pharmacology , Humans , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Phosphoric Monoester Hydrolases/pharmacology
18.
PLoS One ; 8(3): e58937, 2013.
Article in English | MEDLINE | ID: mdl-23516580

ABSTRACT

Enterohemorrhagic and enteropathogenic E. coli (EHEC and EPEC) can cause severe and potentially life-threatening infections. Their pathogenicity is mediated by at least 40 effector proteins which they inject into their host cells by a type-III secretion system leading to the subversion of several cellular pathways. However, the molecular function of several effectors remains unknown, even though they contribute to virulence. Here we show that one of them, NleF, binds to caspase-4, -8, and -9 in yeast two-hybrid, LUMIER, and direct interaction assays. NleF inhibits the catalytic activity of the caspases in vitro and in cell lysate and prevents apoptosis in HeLa and Caco-2 cells. We have solved the crystal structure of the caspase-9/NleF complex which shows that NleF uses a novel mode of caspase inhibition, involving the insertion of the carboxy-terminus of NleF into the active site of the protease. In conformance with our structural model, mutagenized NleF with truncated or elongated carboxy-termini revealed a complete loss in caspase binding and apoptosis inhibition. Evasion of apoptosis helps pathogenic E. coli and other pathogens to take over the host cell by counteracting the cell's ability to self-destruct upon infection. Recently, two other effector proteins, namely NleD and NleH, were shown to interfere with apoptosis. Even though NleF is not the only effector protein capable of apoptosis inhibition, direct inhibition of caspases by bacterial effectors has not been reported to date. Also unique so far is its mode of inhibition that resembles the one obtained for synthetic peptide-type inhibitors and as such deviates substantially from previously reported caspase-9 inhibitors such as the BIR3 domain of XIAP.


Subject(s)
Caspase Inhibitors/metabolism , Caspases/metabolism , Escherichia coli O157 , Escherichia coli Proteins/metabolism , Virulence Factors/metabolism , Caspase Inhibitors/chemistry , Caspase Inhibitors/pharmacology , Caspases/chemistry , Cell Line , Escherichia coli O157/physiology , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/pharmacology , Humans , Models, Molecular , Protein Conformation , Reproducibility of Results , Virulence Factors/chemistry , Virulence Factors/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...