Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Acta Crystallogr F Struct Biol Commun ; 76(Pt 1): 40-46, 2020 Jan 01.
Article in English | MEDLINE | ID: mdl-31929185

ABSTRACT

Direct soaking of protein crystals with small-molecule fragments grouped into complementary clusters is a useful technique when assessing the potential of a new crystal system to support structure-guided drug discovery. It provides a robustness check prior to any extensive crystal screening, a double check for assay binding cutoffs and structural data for binding pockets that may or may not be picked out in assay measurements. The structural output from this technique for three novel fragment molecules identified to bind to the antibacterial target Acinetobacter baumannii undecaprenyl pyrophosphate synthase are reported, and the different physicochemical requirements of a successful antibiotic are compared with traditional medicines.


Subject(s)
Acinetobacter baumannii/enzymology , Alkyl and Aryl Transferases/chemistry , Bacterial Proteins/chemistry , Crystallography, X-Ray/methods , Drug Discovery , Alkyl and Aryl Transferases/isolation & purification , Anti-Bacterial Agents/chemistry , Bacterial Proteins/isolation & purification , Catalytic Domain , Crystallization , Escherichia coli , Gene Expression/genetics , Models, Molecular , Protein Conformation , X-Ray Diffraction
2.
J Med Chem ; 59(15): 7299-304, 2016 Aug 11.
Article in English | MEDLINE | ID: mdl-27379833

ABSTRACT

Undecaprenyl pyrophosphate synthase (UppS) is an essential enzyme in bacterial cell wall synthesis. Here we report the discovery of Staphylococcus aureus UppS inhibitors from an Encoded Library Technology screen and demonstrate binding to the hydrophobic substrate site through cocrystallography studies. The use of bacterial strains with regulated uppS expression and inhibitor resistant mutant studies confirmed that the whole cell activity was the result of UppS inhibition, validating UppS as a druggable antibacterial target.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Anti-Bacterial Agents/pharmacology , Drug Discovery , Enzyme Inhibitors/pharmacology , Pyrazoles/pharmacology , Staphylococcus aureus/drug effects , Alkyl and Aryl Transferases/metabolism , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Staphylococcus aureus/enzymology , Structure-Activity Relationship
3.
Nat Chem Biol ; 10(3): 181-7, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24390428

ABSTRACT

Although therapeutic interventions of signal-transduction cascades with targeted kinase inhibitors are a well-established strategy, drug-discovery efforts to identify targeted phosphatase inhibitors have proven challenging. Herein we report a series of allosteric, small-molecule inhibitors of wild-type p53-induced phosphatase (Wip1), an oncogenic phosphatase common to multiple cancers. Compound binding to Wip1 is dependent on a 'flap' subdomain located near the Wip1 catalytic site that renders Wip1 structurally divergent from other members of the protein phosphatase 2C (PP2C) family and that thereby confers selectivity for Wip1 over other phosphatases. Treatment of tumor cells with the inhibitor GSK2830371 increases phosphorylation of Wip1 substrates and causes growth inhibition in both hematopoietic tumor cell lines and Wip1-amplified breast tumor cells harboring wild-type TP53. Oral administration of Wip1 inhibitors in mice results in expected pharmacodynamic effects and causes inhibition of lymphoma xenograft growth. To our knowledge, GSK2830371 is the first orally active, allosteric inhibitor of Wip1 phosphatase.


Subject(s)
Aminopyridines/chemistry , Dipeptides/chemistry , Enzyme Inhibitors/pharmacology , Phosphoprotein Phosphatases/antagonists & inhibitors , Administration, Oral , Allosteric Regulation , Amino Acid Motifs , Aminopyridines/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Catalytic Domain , Cell Line, Tumor , Dipeptides/pharmacology , Disease Models, Animal , Drug Screening Assays, Antitumor , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , Female , Heterografts , Humans , Mice , Mice, SCID , Models, Biological , Neoplasms , Protein Phosphatase 2C
4.
PLoS One ; 8(7): e67583, 2013.
Article in English | MEDLINE | ID: mdl-23844038

ABSTRACT

Mitogen-Activated Protein Kinase (MAPK) pathway activation has been implicated in many types of human cancer. BRAF mutations that constitutively activate MAPK signalling and bypass the need for upstream stimuli occur with high prevalence in melanoma, colorectal carcinoma, ovarian cancer, papillary thyroid carcinoma, and cholangiocarcinoma. In this report we characterize the novel, potent, and selective BRAF inhibitor, dabrafenib (GSK2118436). Cellular inhibition of BRAF(V600E) kinase activity by dabrafenib resulted in decreased MEK and ERK phosphorylation and inhibition of cell proliferation through an initial G1 cell cycle arrest, followed by cell death. In a BRAF(V600E)-containing xenograft model of human melanoma, orally administered dabrafenib inhibited ERK activation, downregulated Ki67, and upregulated p27, leading to tumor growth inhibition. However, as reported for other BRAF inhibitors, dabrafenib also induced MAPK pathway activation in wild-type BRAF cells through CRAF (RAF1) signalling, potentially explaining the squamous cell carcinomas and keratoacanthomas arising in patients treated with BRAF inhibitors. In addressing this issue, we showed that concomitant administration of BRAF and MEK inhibitors abrogated paradoxical BRAF inhibitor-induced MAPK signalling in cells, reduced the occurrence of skin lesions in rats, and enhanced the inhibition of human tumor xenograft growth in mouse models. Taken together, our findings offer preclinical proof of concept for dabrafenib as a specific and highly efficacious BRAF inhibitor and provide evidence for its potential clinical benefits when used in combination with a MEK inhibitor.


Subject(s)
Melanoma/drug therapy , Melanoma/metabolism , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Drug Evaluation, Preclinical , Enzyme Activation/drug effects , Female , Humans , Imidazoles/administration & dosage , Melanoma/pathology , Mice , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mutation , Oximes/administration & dosage , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Pyridones/administration & dosage , Pyrimidinones/administration & dosage , Signal Transduction/drug effects , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
5.
Arch Biochem Biophys ; 503(2): 207-12, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20816748

ABSTRACT

USP7, also known as the hepes simplex virus associated ubiquitin-specific protease (HAUSP), deubiquitinates both mdm2 and p53, and plays an important role in regulating the level and activity of p53. Here, we report that deletion of the TRAF-like domain at the N-terminus of USP7, previously reported to contain the mdm2/p53 binding site, has no effect on USP7 mediated deubiquitination of Ub(n)-mdm2 and Ub(n)-p53. Amino acids 208-1102 were identified to be the minimal length of USP7 that retains proteolytic activity, similar to full length enzyme, towards not only a truncated model substrate Ub-AFC, but also Ub(n)-mdm2, Ub(n)-p53. In contrast, the catalytic domain of USP7 (amino acids 208-560) has 50-700 fold less proteolytic activity towards different substrates. Moreover, inhibition of the catalytic domain of USP7 by Ubal is also different from the full length or TRAF-like domain deleted proteins. Using glutathione pull-down methods, we demonstrate that the C-terminal domain of USP7 contains additional binding sites, a.a. 801-1050 and a.a. 880-1050 for mdm2 and p53, respectively. The additional USP7 binding site on mdm2 is mapped to be the C-terminal RING finger domain (a.a. 425-491). We propose that the C-terminal domain of USP7 is responsible for maintaining the active conformation for catalysis and inhibitor binding, and contains the prime side of the proteolytic active site.


Subject(s)
Proto-Oncogene Proteins c-mdm2/chemistry , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/metabolism , Ubiquitin Thiolesterase/chemistry , Amino Acid Motifs/genetics , Binding Sites/genetics , Catalytic Domain/genetics , Genes, p53 , Humans , Protein Binding/genetics , Protein Structure, Tertiary/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Tumor Suppressor Protein p53/genetics , Ubiquitin-Specific Peptidase 7 , Ubiquitination
6.
Protein Expr Purif ; 73(2): 167-76, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20457255

ABSTRACT

Phosphoinositide 3-kinases have been targeted for therapeutic research because they are key components of a cell signaling cascade controlling proliferation, growth, and survival. Direct activation of the PI3Kalpha pathway contributes to the development and progression of solid tumors in breast, endometrial, colon, ovarian, and gastric cancers. In the context of a drug discovery effort, the availability of a robust crystallographic system is a means to understand the subtle differences between ATP competitive inhibitor interactions with the active site and their selectivity against other PI3Kinase enzymes. To generate a suitable recombinant design for this purpose, a p85alpha-p110alpha fusion system was developed which enabled the expression and purification of a stoichiometrically homogeneous, constitutively active enzyme for structure determination with potent ATP competitive inhibitors (Raha et al., in preparation) [56]. This approach has yielded preparations with activity and inhibition characteristics comparable to those of the full-length PI3Kalpha from which X-ray diffracting crystals were grown with inhibitors bound in the active site.


Subject(s)
Class II Phosphatidylinositol 3-Kinases/metabolism , Crystallography, X-Ray , Enzyme Inhibitors/pharmacology , Adenosine Triphosphate/metabolism , Animals , Artificial Gene Fusion , Baculoviridae/metabolism , Binding Sites , Cells, Cultured , Class II Phosphatidylinositol 3-Kinases/chemistry , Class II Phosphatidylinositol 3-Kinases/genetics , Class Ia Phosphatidylinositol 3-Kinase/genetics , Drug Design , Inhibitory Concentration 50 , Models, Molecular , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Spodoptera/cytology , Spodoptera/metabolism , X-Ray Diffraction
7.
Anal Biochem ; 383(2): 311-5, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18814837

ABSTRACT

Differential activation of the phosphoinositide 3-kinase (PI3K)/AKT pathway has been linked to cancer. Activation occurs through gene amplification and activating mutations. High-frequency mutations in the gene encoding the p110alpha catalytic subunit of PI3K (PIK3CA) have been observed in a variety of tumors including colon, brain, breast, ovarian, and gastric. Inhibition of PI3K kinase activity may provide a specific way to treat multiple types of human cancer. A scintillation proximity assay (SPA) was developed to detect phosphatidylinositol 3-kinase catalytic activity. Using this assay format, steady-state kinetic parameters were compared for the PI3K class IA enzymes p110alpha, p110beta, and p110delta, each coexpressed with the regulatory subunit p85alpha or splice variant p55alpha. Inhibition by the natural product wortmannin and LY294002 was detected with potencies consistent with alternate assay formats. Other biochemical assay formats have been described for phosphoinositide 3-kinases but each has its unique limitations. The simple, inexpensive, sensitive high-throughput nature of the SPA format has advanced our knowledge of isoform-specific enzymology and will facilitate the discovery of novel PI3K inhibitors.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , Protein Subunits/metabolism , Scintillation Counting/methods , Biocatalysis/drug effects , Biological Products/pharmacology , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacology , Humans , Hydrogen-Ion Concentration , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Microspheres , Phosphoinositide-3 Kinase Inhibitors , Protein Subunits/antagonists & inhibitors , Titrimetry
8.
Biochem J ; 409(2): 519-24, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-17877460

ABSTRACT

The PIK3CA gene, encoding the p110alpha catalytic subunit of Class IA PI3Ks (phosphoinositide 3-kinases), is frequently mutated in many human tumours. The three most common tumour-derived alleles of p110alpha, H1047R, E542K and E545K, were shown to potently activate PI3K signalling in human epithelial cells. In the present study, we examine the biochemical activity of the recombinantly purified PI3K oncogenic mutants. The kinetic characterizations of the wt (wild-type) and the three 'hot spot' PI3K mutants show that the mutants all have approx. 2-fold increase in lipid kinase activities. Interestingly, the phosphorylated IRS-1 (insulin receptor substrate-1) protein shows activation of the lipid kinase activity for the wt and H1047R but not E542K and E545K PI3Kalpha, suggesting that these mutations represent different mechanisms of lipid kinase activation and hence transforming activity in cancer cells.


Subject(s)
Oncogenes , Phosphatidylinositol 3-Kinases/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adenosine Triphosphate/metabolism , Alleles , Catalytic Domain , Class I Phosphatidylinositol 3-Kinases , Enzyme Activation , Humans , Insulin Receptor Substrate Proteins , Kinetics , Mutation , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Tumor Cells, Cultured
9.
Proc Natl Acad Sci U S A ; 103(20): 7625-30, 2006 May 16.
Article in English | MEDLINE | ID: mdl-16684877

ABSTRACT

Heat shock protein (Hsp)90 is emerging as an important therapeutic target for the treatment of cancer. Two analogues of the Hsp90 inhibitor geldanamycin are currently in clinical trials. Geldanamycin (GA) and its analogues have been reported to bind purified Hsp90 with low micromolar potency, in stark contrast to their low nanomolar antiproliferative activity in cell culture and their potent antitumor activity in animal models. Several models have been proposed to account for the approximately 100-fold-greater potency in cell culture, including that GA analogues bind with greater affinity to a five-protein Hsp90 complex than to Hsp90 alone. We have determined that GA and the fluorescent analogue BODIPY-GA (BDGA) both demonstrate slow, tight binding to purified Hsp90. BDGA, used to characterize the kinetics of ligand-Hsp90 interactions, was found to bind Hsp90alpha with k(off) = 2.5 x 10(-3) min(-1), t(1/2) = 4.6 h, and Ki* = 10 nM. It was found that BDGA binds to a functional multiprotein Hsp90 complex with kinetics and affinity identical to that of Hsp90 alone. Also, BDGA binds to Hsp90 from multiple cell lysates in a time-dependent manner with similar kinetics. Therefore, our results indicate that the high potency of GA in cell culture and in vivo can be accounted for by its time-dependent, tight binding to Hsp90 alone. In the broader context, these studies highlight the essentiality of detailed biochemical characterization of drug-target interactions for the effective translation of in vitro pharmacology to cellular and in vivo efficacy.


Subject(s)
Antibiotics, Antineoplastic , HSP90 Heat-Shock Proteins , Quinones , Animals , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/metabolism , Benzoquinones , Boron Compounds/chemistry , Boron Compounds/metabolism , Cells, Cultured , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Humans , Lactams, Macrocyclic , Protein Binding , Quinones/chemistry , Quinones/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...