Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Med Chem ; 64(19): 14498-14512, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34570508

ABSTRACT

Poly-ADP-ribose-polymerase (PARP) inhibitors have achieved regulatory approval in oncology for homologous recombination repair deficient tumors including BRCA mutation. However, some have failed in combination with first-line chemotherapies, usually due to overlapping hematological toxicities. Currently approved PARP inhibitors lack selectivity for PARP1 over PARP2 and some other 16 PARP family members, and we hypothesized that this could contribute to toxicity. Recent literature has demonstrated that PARP1 inhibition and PARP1-DNA trapping are key for driving efficacy in a BRCA mutant background. Herein, we describe the structure- and property-based design of 25 (AZD5305), a potent and selective PARP1 inhibitor and PARP1-DNA trapper with excellent in vivo efficacy in a BRCA mutant HBCx-17 PDX model. Compound 25 is highly selective for PARP1 over other PARP family members, with good secondary pharmacology and physicochemical properties and excellent pharmacokinetics in preclinical species, with reduced effects on human bone marrow progenitor cells in vitro.


Subject(s)
DNA , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases , Humans , Crystallography, X-Ray , DNA/chemistry , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly(ADP-ribose) Polymerase Inhibitors/chemistry , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Substrate Specificity
2.
J Med Chem ; 64(18): 13704-13718, 2021 09 23.
Article in English | MEDLINE | ID: mdl-34491761

ABSTRACT

The epidermal growth factor receptor (EGFR) harboring activating mutations is a clinically validated target in non-small-cell lung cancer, and a number of inhibitors of the EGFR tyrosine kinase domain, including osimertinib, have been approved for clinical use. Resistance to these therapies has emerged due to a variety of molecular events including the C797S mutation which renders third-generation C797-targeting covalent EGFR inhibitors considerably less potent against the target due to the loss of the key covalent-bond-forming residue. We describe the medicinal chemistry optimization of a biochemically potent but modestly cell-active, reversible EGFR inhibitor starting point with sub-optimal physicochemical properties. These studies culminated in the identification of compound 12 that showed improved cell potency, oral exposure, and in vivo activity in clinically relevant EGFR-mutant-driven disease models, including an Exon19 deletion/T790M/C797S triple-mutant mouse xenograft model.


Subject(s)
Antineoplastic Agents/therapeutic use , ErbB Receptors/antagonists & inhibitors , Neoplasms/drug therapy , Organophosphorus Compounds/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Humans , Mice, Nude , Mice, SCID , Mutation , Organophosphorus Compounds/chemical synthesis , Organophosphorus Compounds/metabolism , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/metabolism , Pyrimidines/chemical synthesis , Pyrimidines/metabolism , Rats , Xenograft Model Antitumor Assays
3.
Nat Commun ; 11(1): 4132, 2020 08 17.
Article in English | MEDLINE | ID: mdl-32807781

ABSTRACT

Precise genome editing using CRISPR-Cas9 is a promising therapeutic avenue for genetic diseases, although off-target editing remains a significant safety concern. Guide RNAs shorter than 16 nucleotides in length effectively recruit Cas9 to complementary sites in the genome but do not permit Cas9 nuclease activity. Here we describe CRISPR Guide RNA Assisted Reduction of Damage (CRISPR GUARD) as a method for protecting off-targets sites by co-delivery of short guide RNAs directed against off-target loci by competition with the on-target guide RNA. CRISPR GUARD reduces off-target mutagenesis while retaining on-target editing efficiencies with Cas9 and base editor. However, we discover that short guide RNAs can also support base editing if they contain cytosines within the deaminase activity window. We explore design rules and the universality of this method through in vitro studies and high-throughput screening, revealing CRISPR GUARD as a rapidly implementable strategy to improve the specificity of genome editing for most genomic loci. Finally, we create an online tool for CRISPR GUARD design.


Subject(s)
Gene Editing/methods , RNA, Guide, Kinetoplastida/metabolism , CRISPR-Cas Systems/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/physiology , Humans , Mutagenesis/genetics , Mutagenesis/physiology , RNA, Guide, Kinetoplastida/genetics
4.
Acta Crystallogr D Struct Biol ; 75(Pt 11): 1003-1014, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-31692474

ABSTRACT

Apoptosis is a crucial process by which multicellular organisms control tissue growth, removal and inflammation. Disruption of the normal apoptotic function is often observed in cancer, where cell death is avoided by the overexpression of anti-apoptotic proteins of the Bcl-2 (B-cell lymphoma 2) family, including Mcl-1 (myeloid cell leukaemia 1). This makes Mcl-1 a potential target for drug therapy, through which normal apoptosis may be restored by inhibiting the protective function of Mcl-1. Here, the discovery and biophysical properties of an anti-Mcl-1 antibody fragment are described and the utility of both the scFv and Fab are demonstrated in generating an Mcl-1 crystal system amenable to iterative structure-guided drug design.


Subject(s)
Drug Discovery , Immunoglobulin Fab Fragments/chemistry , Myeloid Cell Leukemia Sequence 1 Protein/chemistry , Single-Chain Antibodies/chemistry , Animals , Apoptosis , CHO Cells , Cloning, Molecular , Cricetulus , Escherichia coli/genetics , Humans
5.
SLAS Discov ; 24(2): 121-132, 2019 02.
Article in English | MEDLINE | ID: mdl-30543471

ABSTRACT

Methods to measure cellular target engagement are increasingly being used in early drug discovery. The Cellular Thermal Shift Assay (CETSA) is one such method. CETSA can investigate target engagement by measuring changes in protein thermal stability upon compound binding within the intracellular environment. It can be performed in high-throughput, microplate-based formats to enable broader application to early drug discovery campaigns, though high-throughput forms of CETSA have only been reported for a limited number of targets. CETSA offers the advantage of investigating the target of interest in its physiological environment and native state, but it is not clear yet how well this technology correlates to more established and conventional cellular and biochemical approaches widely used in drug discovery. We report two novel high-throughput CETSA (CETSA HT) assays for B-Raf and PARP1, demonstrating the application of this technology to additional targets. By performing comparative analyses with other assays, we show that CETSA HT correlates well with other screening technologies and can be applied throughout various stages of hit identification and lead optimization. Our results support the use of CETSA HT as a broadly applicable and valuable methodology to help drive drug discovery campaigns to molecules that engage the intended target in cells.


Subject(s)
Drug Discovery , High-Throughput Screening Assays/methods , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Temperature , Cell Line, Tumor , Humans , Poly (ADP-Ribose) Polymerase-1/metabolism , Proto-Oncogene Proteins B-raf/metabolism
6.
Br J Pharmacol ; 175(4): 618-630, 2018 02.
Article in English | MEDLINE | ID: mdl-29161763

ABSTRACT

BACKGROUND AND PURPOSE: Several anti-angiogenic cancer drugs that inhibit VEGF receptor (VEGFR) signalling for efficacy are associated with a 15-60% incidence of hypertension. Tyrosine kinase inhibitors (TKIs) that have off-target activity at VEGFR-2 may also cause blood pressure elevation as an undesirable side effect. Therefore, the ability to translate VEGFR-2 off-target potency into blood pressure elevation would be useful in development of novel TKIs. Here, we have sought to quantify the relationship between VEGFR-2 inhibition and blood pressure elevation for a range of kinase inhibitors. EXPERIMENTAL APPROACH: Porcine aortic endothelial cells overexpressing VEGFR-2 (PAE) were used to determine IC50 for VEGFR-2 phosphorylation. These IC50 values were compared with published reports of exposure attained during clinical use and the corresponding incidence of all-grade hypertension. Unbound average plasma concentration (Cav,u ) was selected to be the most appropriate pharmacokinetic parameter. The pharmacokinetic-pharmacodynamic (PKPD) relationship for blood pressure elevation was investigated for selected kinase inhibitors, using data derived either from clinical papers or from rat telemetry experiments. KEY RESULTS: All-grade hypertension was predominantly observed when the Cav,u was >0.1-fold of the VEGFR-2 (PAE) IC50 . Furthermore, based on the PKPD analysis, an exposure-dependent blood pressure elevation >1 mmHg was observed only when the Cav,u was >0.1-fold of the VEGFR-2 (PAE) IC50 . CONCLUSIONS AND IMPLICATIONS: Taken together, these data show that the risk of blood pressure elevation is proportional to the amount of VEGFR-2 inhibition, and a margin of >10-fold between VEGFR-2 IC50 and Cav,u appears to confer a minimal risk of hypertension.


Subject(s)
Angiogenesis Inhibitors/toxicity , Blood Pressure/physiology , Hypertension/chemically induced , Protein Kinase Inhibitors/toxicity , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Animals , Axitinib , Blood Pressure/drug effects , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hypertension/metabolism , Imidazoles/toxicity , Indazoles/toxicity , Rats , Swine , Vascular Endothelial Growth Factor Receptor-2/metabolism
7.
Nat Commun ; 8(1): 1610, 2017 11 20.
Article in English | MEDLINE | ID: mdl-29151576

ABSTRACT

The CRISPR-Cas9 RNA-guided endonuclease system allows precise and efficient modification of complex genomes and is continuously developed to enhance specificity, alter targeting and add new functional moieties. However, one area yet to be explored is the base chemistry of the associated RNA molecules. Here we show the design and optimisation of hybrid DNA-RNA CRISPR and tracr molecules based on structure-guided approaches. Through careful mapping of the ribose requirements of Cas9, we develop hybrid versions possessing minimal RNA residues, which are sufficient to direct specific nuclease activity in vitro and in vivo with reduced off-target activity. We identify critical regions within these molecules that require ribose nucleotides and show a direct correlation between binding affinity/stability and cellular activity. This is the first demonstration of a non-RNA-guided Cas9 endonuclease and first step towards eliminating the ribose dependency of Cas9 to develop a XNA-programmable endonuclease.


Subject(s)
Bacterial Proteins/chemistry , DNA/chemistry , Endonucleases/chemistry , RNA, Guide, Kinetoplastida/chemistry , RNA/chemistry , Bacterial Proteins/metabolism , Biocatalysis , CRISPR-Associated Protein 9 , Clustered Regularly Interspaced Short Palindromic Repeats , DNA/genetics , Endonucleases/metabolism , Nucleic Acid Conformation , RNA/genetics , RNA, Guide, Kinetoplastida/genetics
8.
PLoS One ; 11(3): e0151154, 2016.
Article in English | MEDLINE | ID: mdl-26999531

ABSTRACT

MTH1 (NUDT1) is an oncologic target involved in the prevention of DNA damage. We investigate the way MTH1 recognises its substrates and present substrate-bound structures of MTH1 for 8-oxo-dGTP and 8-oxo-rATP as examples of novel strong and weak binding substrate motifs. Investigation of a small set of purine-like fragments using 2D NMR resulted in identification of a fragment with weak potency. The protein-ligand X-Ray structure of this fragment provides insight into the role of water molecules in substrate selectivity. Wider fragment screening by NMR resulted in three new protein structures exhibiting alternative binding configurations to the key Asp-Asp recognition element of the protein. These inhibitor binding modes demonstrate that MTH1 employs an intricate yet promiscuous mechanism of substrate anchoring through its Asp-Asp pharmacophore. The structures suggest that water-mediated interactions convey selectivity towards oxidized substrates over their non-oxidised counterparts, in particular by stabilization of a water molecule in a hydrophobic environment through hydrogen bonding. These findings may be useful in the design of inhibitors of MTH1.


Subject(s)
Phosphoric Monoester Hydrolases/metabolism , Amino Acid Motifs , Aspartic Acid/metabolism , Binding Sites , Crystallography, X-Ray , Deoxyguanine Nucleotides/chemistry , Deoxyguanine Nucleotides/metabolism , Hydrogen Bonding , Kinetics , Magnetic Resonance Spectroscopy , Phosphoric Monoester Hydrolases/chemistry , Protein Structure, Secondary , Substrate Specificity , Water
9.
J Med Chem ; 59(6): 2346-61, 2016 Mar 24.
Article in English | MEDLINE | ID: mdl-26878898

ABSTRACT

Recent literature has claimed that inhibition of the enzyme MTH1 can eradicate cancer. MTH1 is one of the "housekeeping" enzymes that are responsible for hydrolyzing damaged nucleotides in cells and thus prevent them from being incorporated into DNA. We have developed orthogonal and chemically distinct tool compounds to those published in the literature to allow us to test the hypothesis that inhibition of MTH1 has wide applicability in the treatment of cancer. Here we present the work that led to the discovery of three structurally different series of MTH1 inhibitors with excellent potency, selectivity, and proven target engagement in cells. None of these compounds elicited the reported cellular phenotype, and additional siRNA and CRISPR experiments further support these observations. Critically, the difference between the responses of our highly selective inhibitors and published tool compounds suggests that the effect reported for the latter may be due to off-target cytotoxic effects. As a result, we conclude that the role of MTH1 in carcinogenesis and utility of its inhibition is yet to be established.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , DNA Repair Enzymes/antagonists & inhibitors , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Cell Survival/drug effects , Crystallography, X-Ray , Drug Discovery , Humans , Models, Molecular , RNA, Small Interfering/pharmacology , Rats , Structure-Activity Relationship
10.
Structure ; 21(12): 2143-51, 2013 Dec 03.
Article in English | MEDLINE | ID: mdl-24207125

ABSTRACT

Reactivation of p53 by release of the functional protein from its inhibition by MDM2 provides an efficient, nongenotoxic approach to a wide variety of cancers. We present the cocrystal structures of two complexes of MDM2 with inhibitors based on 6-chloroindole scaffolds. Both molecules bound to a distinct conformational state of MDM2 with nM-µM affinities. In contrast to other structurally characterized antagonists, which mimic three amino acids of p53 (Phe19, Trp23, and Leu26), the compounds induced an additional hydrophobic pocket on the MDM2 surface and unveiled a four-point binding mode. The enlarged interaction interface of the inhibitors resulted in extension of small molecules binding toward the "lid" segment of MDM2 (residues 19-23)--a nascent element that interferes with p53 binding. As supported by protein engineering and molecular dynamics studies, employing these unstable elements of MDM2 provides an efficient and yet unexplored alternative in development of MDM2-p53 association inhibitors.


Subject(s)
Dipeptides/chemistry , Hydroxamic Acids/chemistry , Proto-Oncogene Proteins c-mdm2/chemistry , Tryptophan/analogs & derivatives , Tumor Suppressor Protein p53/chemistry , Crystallography, X-Ray , Humans , Molecular Dynamics Simulation , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Protein Interaction Domains and Motifs , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Tryptophan/chemistry , Tumor Suppressor Protein p53/antagonists & inhibitors
11.
Proc Natl Acad Sci U S A ; 110(44): 17814-9, 2013 Oct 29.
Article in English | MEDLINE | ID: mdl-24127580

ABSTRACT

MDM2 and MDMX are homologous proteins that bind to p53 and regulate its activity. Both contain three folded domains and ~70% intrinsically disordered regions. Previous detailed structural and biophysical studies have concentrated on the isolated folded domains. The N-terminal domains of both exhibit high affinity for the disordered N-terminal of p53 (p53TAD) and inhibit its transactivation function. Here, we have studied full-length MDMX and found a ~100-fold weaker affinity for p53TAD than does its isolated N-terminal domain. We found from NMR spectroscopy and binding studies that MDMX (but not MDM2) contains a conserved, disordered self-inhibitory element that competes intramolecularly for binding with p53TAD. This motif, which we call the WWW element, is centered around residues Trp200 and Trp201. Deletion or mutation of the element increased binding affinity of MDMX to that of the isolated N-terminal domain level. The self-inhibition of MDMX implies a regulatory, allosteric mechanism of its activity. MDMX rests in a latent state in which its binding activity with p53TAD is masked by autoinhibition. Activation of MDMX would require binding to a regulatory protein. The inhibitory function of the WWW element may explain the oncogenic effects of an alternative splicing variant of MDMX that does not contain the WWW element and is found in some aggressive cancers.


Subject(s)
Nuclear Proteins/genetics , Oncogenes/genetics , Proto-Oncogene Proteins/genetics , Regulatory Elements, Transcriptional/genetics , Tumor Suppressor Protein p53/antagonists & inhibitors , Calorimetry , Cell Cycle Proteins , Chromatography, Gel , Humans , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Tumor Suppressor Protein p53/genetics , Ubiquitination , Ultracentrifugation
12.
Proc Natl Acad Sci U S A ; 109(39): 15752-6, 2012 Sep 25.
Article in English | MEDLINE | ID: mdl-22972749

ABSTRACT

The tumor suppressor p53 is a homotetramer of 4 × 393 residues. Its core domain and tetramerization domain are linked and flanked by intrinsically disordered sequences, which hinder its full structural characterization. There is an outstanding problem of the state of the tetramerization domain. Structural studies on the isolated tetramerization domain show it is in a folded tetrameric conformation, but there are conflicting models from electron microscopy of the full-length protein, one of which proposes that the domain is not tetramerically folded and the tetrameric protein is stabilized by interactions between the N and C termini. Here, we present methyl-transverse relaxation optimized NMR spectroscopy (methyl-TROSY) investigations on the full-length and separate domains of the protein with its methionine residues enriched with (13)C to probe its quaternary structure. We obtained high-quality spectra of both the full-length tetrameric p53 and its DNA complex, observing the environment at 11 specific methyl sites. The tetramerization domain was as tetramerically folded in the full-length constructs as in the isolated domain. The N and C termini were intrinsically disordered in both the full-length protein and its complex with a 20-residue specific DNA sequence. Additionally, we detected in the interface of the core (DNA-binding) and N-terminal parts of the protein a slow conformational exchange process that was modulated by specific recognition of DNA, indicating allosteric processes.


Subject(s)
DNA/chemistry , Protein Folding , Tumor Suppressor Protein p53/chemistry , Allosteric Regulation , DNA/metabolism , Humans , Nuclear Magnetic Resonance, Biomolecular , Protein Structure, Quaternary , Protein Structure, Tertiary , Structure-Activity Relationship , Tumor Suppressor Protein p53/metabolism
13.
PLoS One ; 7(6): e37518, 2012.
Article in English | MEDLINE | ID: mdl-22675482

ABSTRACT

SJ-172550 (1) was previously discovered in a biochemical high throughput screen for inhibitors of the interaction of MDMX and p53 and characterized as a reversible inhibitor (J. Biol. Chem. 2010; 285:10786). Further study of the biochemical mode of action of 1 has shown that it acts through a complicated mechanism in which the compound forms a covalent but reversible complex with MDMX and locks MDMX into a conformation that is unable to bind p53. The relative stability of this complex is influenced by many factors including the reducing potential of the media, the presence of aggregates, and other factors that influence the conformational stability of the protein. This complex mechanism of action hinders the further development of compound 1 as a selective MDMX inhibitor.


Subject(s)
Acetates/pharmacology , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Pyrazoles/pharmacology , Tumor Suppressor Protein p53/metabolism , Acetates/chemistry , Amino Acid Sequence , Buffers , Cell Cycle Proteins , Humans , Inhibitory Concentration 50 , Models, Biological , Molecular Sequence Data , Nuclear Proteins/chemistry , Peptides/metabolism , Pliability/drug effects , Protein Binding/drug effects , Protein Conformation , Protein Stability/drug effects , Proto-Oncogene Proteins/chemistry , Pyrazoles/chemistry , Temperature , Tumor Suppressor Protein p53/chemistry
14.
PLoS One ; 7(3): e32839, 2012.
Article in English | MEDLINE | ID: mdl-22427896

ABSTRACT

Although there is no shortage of potential drug targets, there are only a handful known low-molecular-weight inhibitors of protein-protein interactions (PPIs). One problem is that current efforts are dominated by low-yield high-throughput screening, whose rigid framework is not suitable for the diverse chemotypes present in PPIs. Here, we developed a novel pharmacophore-based interactive screening technology that builds on the role anchor residues, or deeply buried hot spots, have in PPIs, and redesigns these entry points with anchor-biased virtual multicomponent reactions, delivering tens of millions of readily synthesizable novel compounds. Application of this approach to the MDM2/p53 cancer target led to high hit rates, resulting in a large and diverse set of confirmed inhibitors, and co-crystal structures validate the designed compounds. Our unique open-access technology promises to expand chemical space and the exploration of the human interactome by leveraging in-house small-scale assays and user-friendly chemistry to rationally design ligands for PPIs with known structure.


Subject(s)
Chemistry, Pharmaceutical/methods , Drug Discovery/methods , Protein Interaction Mapping/methods , Small Molecule Libraries , Software , Systems Biology/methods , Crystallography , Internet , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/chemistry , Systems Biology/trends , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/chemistry
16.
Chem Biol Drug Des ; 76(2): 116-29, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20492448

ABSTRACT

1,4-Thienodiazepine-2,5-diones have been synthesized via the Ugi-Deprotection-Cyclization (UDC) approach starting from Gewald 2-aminothiophenes in a convergent and versatile manner. The resulting scaffold is unprecedented, cyclic, and peptidomimetic with four points of diversity introduced from readily available starting materials. In addition to eighteen synthesized and characterized compounds, a virtual compound library was generated and evaluated for chemical space distribution and drug-like properties. A small focused compound library of 1,4-thienodiazepine-2,5-diones has been screened for the activity against p53-Mdm2 interaction. Biological evaluations demonstrated that some compounds exhibited promising antagonistic activity.


Subject(s)
Azepines/chemistry , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Azepines/chemical synthesis , Azepines/pharmacology , Binding Sites , Computer Simulation , Crystallography, X-Ray , Humans , Magnetic Resonance Spectroscopy , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Thiophenes/chemistry , Tumor Suppressor Protein p53/antagonists & inhibitors
17.
J Am Chem Soc ; 131(22): 7500-1, 2009 Jun 10.
Article in English | MEDLINE | ID: mdl-19422216

ABSTRACT

NMR-based drug screening methods provide the most reliable characterization of binding propensities of ligands to their target proteins. Unique to NMR is its capability to detect weak microM-mM bindings. NMR assays are, however, one of the least effective methods in terms of the amount of protein required and the time needed for acquiring NMR experiments. We have recently described a time efficient 1D proton NMR assay for studying the effect of antagonists on protein-protein interactions. The method, named AIDA-NMR (for Antagonist Induced Dissociation Assay-NMR), can provide information on whether an antagonist of a protein-protein interaction is strong enough to dissociate the complex and, in addition, whether its interaction is through denaturation, precipitation, or release of a protein in its functional folded state. AIDA requires a large protein fragment (larger than ca. 30 kDa) to bind to a small reporter protein (smaller than ca.12 kDa). Here, we present an extension of this method, named SEI AIDA (SEI, for Selective Excitation-Inversion). The SEI AIDA uses tryptophan-bearing proteins, and by selectively exciting only the proton NMR signals of the (N)H(epsilon) indole side chains of tryptophans, the acquisition time of the AIDA experiment can be reduced by an order of magnitude relative to the corresponding 1D AIDA that uses hard pulses. Thus, at 600 MHz, the (N)H(epsilon) signal of a 35 microM protein complex can be acquired in only 2.5 min, making the SEI AIDA suitable for high-throughput screening pipelines in drug discovery.


Subject(s)
Nuclear Magnetic Resonance, Biomolecular/methods , Proteins/chemistry , Tryptophan/chemistry , Proteins/analysis
18.
Biochem J ; 419(3): 555-64, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19175361

ABSTRACT

Staphylococcus aureus is a dangerous human pathogen whose antibiotic resistance is steadily increasing and no efficient vaccine is as yet available. This serious threat drives extensive studies on staphylococcal physiology and pathogenicity pathways, especially virulence factors. Spl (serine protease-like) proteins encoded by an operon containing up to six genes are a good example of poorly characterized secreted proteins probably involved in virulence. In the present study, we describe an efficient heterologous expression system for SplA and detailed biochemical and structural characterization of the recombinant SplA protease. The enzyme shares a significant sequence homology to V8 protease and epidermolytic toxins which are well documented staphylococcal virulence factors. SplA has a very narrow substrate specificity apparently imposed by the precise recognition of three amino acid residues positioned N-terminal to the hydrolysed peptide bond. To explain determinants of this extended specificity we resolve the crystal structure of SplA and define the consensus model of substrate binding. Furthermore we demonstrate that artificial N-terminal elongation of mature SplA mimicking a naturally present signal peptide abolishes enzymatic activity. The probable physiological role of the process is discussed. Of interest, even though precise N-terminal trimming is a common regulatory mechanism among S1 family enzymes, the crystal structure of SplA reveals novel significantly different mechanistic details.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Serine Endopeptidases/chemistry , Serine Endopeptidases/metabolism , Staphylococcus aureus/enzymology , Amino Acid Sequence , Animals , Anions , Biocatalysis , Chymotrypsin/chemistry , Crystallography, X-Ray , Histidine , Hydrogen Bonding , Models, Molecular , Molecular Sequence Data , Protein Structure, Secondary , Recombinant Proteins/biosynthesis , Substrate Specificity
19.
Acta Biochim Pol ; 55(3): 525-36, 2008.
Article in English | MEDLINE | ID: mdl-18769737

ABSTRACT

Thermolysins constitute a family of secreted bacterial metalloproteases expressed, among others, by several pathogens. Strains of Staphylococcus pseudintermedius isolated from diseased dogs and judged as protease-positive, by skim milk agar plate culture, were investigated for protease content. No proteolytic activity was detected when the bacteria were grown in regular liquid media. Unexpectedly, supplementation of the medium with calcium ions resulted in expression of a metalloprotease and profound changes in the profile of extracellular proteins. On the basis of homology to other staphylococcal metalloproteases, the nucleotide sequence of the gene encoding this protease (Pst) and its flanking regions was determined. The full-length pst codes for a protein with an open reading frame of 505 amino acids. The internal region contains the HEXXH catalytic domain that is conserved in members of the thermolysin family. Regardless of the presence of calcium in the medium, the expression of the protease gene was of the same intensity. This suggests that regulation of the metalloprotease production by calcium ions is at a post-transcriptional level. Isolates of S. pseudintermedius exhibit a proteolytic phenotype due to the metalloprotease expression, however only in presence of calcium ions, which most probably stabilize the structure of the protease.


Subject(s)
Metalloendopeptidases/chemistry , Metalloendopeptidases/genetics , Staphylococcus/enzymology , Staphylococcus/genetics , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Calcium/metabolism , Cloning, Molecular , DNA Primers/genetics , DNA, Bacterial/genetics , Dogs , Evolution, Molecular , Gene Expression , Genes, Bacterial , Metalloendopeptidases/metabolism , Models, Molecular , Molecular Sequence Data , Polymorphism, Genetic , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Staphylococcus/isolation & purification , Staphylococcus/pathogenicity , Substrate Specificity
20.
Nat Methods ; 5(7): 605-7, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18536722

ABSTRACT

Live imaging of the actin cytoskeleton is crucial for the study of many fundamental biological processes, but current approaches to visualize actin have several limitations. Here we describe Lifeact, a 17-amino-acid peptide, which stained filamentous actin (F-actin) structures in eukaryotic cells and tissues. Lifeact did not interfere with actin dynamics in vitro and in vivo and in its chemically modified peptide form allowed visualization of actin dynamics in nontransfectable cells.


Subject(s)
Actins/metabolism , Coloring Agents/chemistry , Peptides/chemistry , Staining and Labeling/methods , Animals , Cells, Cultured , Cytoskeleton/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Mice , Microscopy, Fluorescence , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...