Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 108
Filter
1.
J Proteome Res ; 22(9): 2959-2972, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37582225

ABSTRACT

Proteins often undergo structural perturbations upon binding to other proteins or ligands or when they are subjected to environmental changes. Hydrogen-deuterium exchange mass spectrometry (HDX-MS) can be used to explore conformational changes in proteins by examining differences in the rate of deuterium incorporation in different contexts. To determine deuterium incorporation rates, HDX-MS measurements are typically made over a time course. Recently introduced methods show that incorporating the temporal dimension into the statistical analysis improves power and interpretation. However, these approaches have technical assumptions that hinder their flexibility. Here, we propose a more flexible methodology by reframing these methods in a Bayesian framework. Our proposed framework has improved algorithmic stability, allows us to perform uncertainty quantification, and can calculate statistical quantities that are inaccessible to other approaches. We demonstrate the general applicability of the method by showing it can perform rigorous model selection on a spike-in HDX-MS experiment, improved interpretation in an epitope mapping experiment, and increased sensitivity in a small molecule case-study. Bayesian analysis of an HDX experiment with an antibody dimer bound to an E3 ubiquitin ligase identifies at least two interaction interfaces where previous methods obtained confounding results due to the complexities of conformational changes on binding. Our findings are consistent with the cocrystal structure of these proteins, demonstrating a bayesian approach can identify important binding epitopes from HDX data. We also generate HDX-MS data of the bromodomain-containing protein BRD4 in complex with GSK1210151A to demonstrate the increased sensitivity of adopting a Bayesian approach.


Subject(s)
Deuterium Exchange Measurement , Hydrogen Deuterium Exchange-Mass Spectrometry , Bayes Theorem , Deuterium/chemistry , Deuterium Exchange Measurement/methods , Nuclear Proteins , Mass Spectrometry/methods , Transcription Factors
2.
J Proteome Res ; 21(4): 849-864, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35258980

ABSTRACT

Proteomics is a data-rich science with complex experimental designs and an intricate measurement process. To obtain insights from the large data sets produced, statistical methods, including machine learning, are routinely applied. For a quantity of interest, many of these approaches only produce a point estimate, such as a mean, leaving little room for more nuanced interpretations. By contrast, Bayesian statistics allows quantification of uncertainty through the use of probability distributions. These probability distributions enable scientists to ask complex questions of their proteomics data. Bayesian statistics also offers a modular framework for data analysis by making dependencies between data and parameters explicit. Hence, specifying complex hierarchies of parameter dependencies is straightforward in the Bayesian framework. This allows us to use a statistical methodology which equals, rather than neglects, the sophistication of experimental design and instrumentation present in proteomics. Here, we review Bayesian methods applied to proteomics, demonstrating their potential power, alongside the challenges posed by adopting this new statistical framework. To illustrate our review, we give a walk-through of the development of a Bayesian model for dynamic organic orthogonal phase-separation (OOPS) data.


Subject(s)
Machine Learning , Proteomics , Bayes Theorem , Probability , Uncertainty
3.
Proc Natl Acad Sci U S A ; 116(19): 9318-9323, 2019 05 07.
Article in English | MEDLINE | ID: mdl-30962368

ABSTRACT

Visceral leishmaniasis (VL), caused by the protozoan parasites Leishmania donovani and Leishmania infantum, is one of the major parasitic diseases worldwide. There is an urgent need for new drugs to treat VL, because current therapies are unfit for purpose in a resource-poor setting. Here, we describe the development of a preclinical drug candidate, GSK3494245/DDD01305143/compound 8, with potential to treat this neglected tropical disease. The compound series was discovered by repurposing hits from a screen against the related parasite Trypanosoma cruzi Subsequent optimization of the chemical series resulted in the development of a potent cidal compound with activity against a range of clinically relevant L. donovani and L. infantum isolates. Compound 8 demonstrates promising pharmacokinetic properties and impressive in vivo efficacy in our mouse model of infection comparable with those of the current oral antileishmanial miltefosine. Detailed mode of action studies confirm that this compound acts principally by inhibition of the chymotrypsin-like activity catalyzed by the ß5 subunit of the L. donovani proteasome. High-resolution cryo-EM structures of apo and compound 8-bound Leishmania tarentolae 20S proteasome reveal a previously undiscovered inhibitor site that lies between the ß4 and ß5 proteasome subunits. This induced pocket exploits ß4 residues that are divergent between humans and kinetoplastid parasites and is consistent with all of our experimental and mutagenesis data. As a result of these comprehensive studies and due to a favorable developability and safety profile, compound 8 is being advanced toward human clinical trials.


Subject(s)
Antiprotozoal Agents/administration & dosage , Leishmania donovani/drug effects , Leishmania infantum/drug effects , Leishmaniasis, Visceral/diagnostic imaging , Proteasome Inhibitors/administration & dosage , Protozoan Proteins/antagonists & inhibitors , Animals , Antiprotozoal Agents/chemistry , Binding Sites , Disease Models, Animal , Drug Evaluation, Preclinical , Humans , Leishmania donovani/chemistry , Leishmania donovani/enzymology , Leishmania infantum/chemistry , Leishmania infantum/enzymology , Leishmaniasis, Visceral/parasitology , Male , Mice , Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/chemistry , Protein Conformation , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism
4.
Bioconjug Chem ; 32(2): 279-289, 2021 02 17.
Article in English | MEDLINE | ID: mdl-33523652

ABSTRACT

Reducing the required frequence of drug dosing can improve the adherence of patients to chronic treatments. Hence, drugs with longer in vivo half-lives are highly desirable. One of the most promising approaches to extend the in vivo half-life of drugs is conjugation to human serum albumin (HSA). In this work, we describe the use of AlbuBinder 1, a small-molecule noncovalent HSA binder, to extend the in vivo half-life and pharmacology of small-molecule BMP1/TLL inhibitors in humanized mice (HSA KI/KI). A series of conjugates of AlbuBinder 1 with BMP1/TLL inhibitors were prepared. In particular, conjugate c showed good solubility and a half-life extension of >20-fold versus the parent molecule in the HSA KI/KI mice, reaching half-lives of >48 h with maintained maximal inhibition of plasma BMP1/TLL. The same conjugate showed a half-life of only 3 h in the wild-type mice, suggesting that the half-life extension was principally due to specific interactions with HSA. It is envisioned that conjugation to AlbuBinder 1 should be applicable to a wide range of small molecule or peptide drugs with short half-lives. In this context, AlbuBinders represent a viable alternative to existing half-life extension technologies.


Subject(s)
Metalloproteases/metabolism , Protease Inhibitors/pharmacology , Serum Albumin, Human/metabolism , Small Molecule Libraries/metabolism , Animals , Bone Morphogenetic Protein 1/metabolism , Half-Life , Humans , Mice , Proof of Concept Study , Protease Inhibitors/pharmacokinetics
5.
Chemistry ; 27(71): 17880-17888, 2021 Dec 20.
Article in English | MEDLINE | ID: mdl-34328642

ABSTRACT

We present a one-step Ugi reaction protocol for the expedient synthesis of photoaffinity probes for live-cell MS-based proteomics. The reaction couples an amine affinity function with commonly used photoreactive groups, and a variety of handle functionalities. Using this technology, a series of pan-BET (BET: bromodomain and extra-terminal domain) selective bromodomain photoaffinity probes were obtained by parallel synthesis. Studies on the effects of photoreactive group, linker length and irradiation wavelength on photocrosslinking efficiency provide valuable insights into photoaffinity probe design. Optimal probes were progressed to MS-based proteomics to capture the BET family of proteins from live cells and reveal their potential on- and off-target profiles.


Subject(s)
Proteomics
6.
Bioorg Med Chem Lett ; 41: 127973, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33753261

ABSTRACT

α1-antitrypsin deficiency is characterised by the misfolding and intracellular polymerisation of mutant α1-antitrypsin protein within the endoplasmic reticulum (ER) of hepatocytes. Small molecules that bind and stabilise Z α1-antitrypsin were identified via a DNA-encoded library screen. A subsequent structure based optimisation led to a series of highly potent, selective and cellular active α1-antitrypsin correctors.


Subject(s)
Drug Design , Protein Folding , alpha 1-Antitrypsin/metabolism , Crystallization , Drug Development/methods , Drug Evaluation, Preclinical , Endoplasmic Reticulum/metabolism , Gene Library , Hepatocytes/metabolism , Humans , Models, Molecular , Protein Conformation , alpha 1-Antitrypsin/genetics
7.
Angew Chem Int Ed Engl ; 60(43): 23327-23334, 2021 10 18.
Article in English | MEDLINE | ID: mdl-34416073

ABSTRACT

Focal adhesion kinase (FAK) is a key mediator of tumour progression and metastasis. To date, clinical trials of FAK inhibitors have reported disappointing efficacy for oncology indications. We report the design and characterisation of GSK215, a potent, selective, FAK-degrading Proteolysis Targeting Chimera (PROTAC) based on a binder for the VHL E3 ligase and the known FAK inhibitor VS-4718. X-ray crystallography revealed the molecular basis of the highly cooperative FAK-GSK215-VHL ternary complex, and GSK215 showed differentiated in-vitro pharmacology compared to VS-4718. In mice, a single dose of GSK215 induced rapid and prolonged FAK degradation, giving a long-lasting effect on FAK levels (≈96 h) and a marked PK/PD disconnect. This tool PROTAC molecule is expected to be useful for the study of FAK-degradation biology in vivo, and our results indicate that FAK degradation may be a differentiated clinical strategy versus FAK inhibition for the treatment of cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Focal Adhesion Kinase 1/antagonists & inhibitors , Proteolysis/drug effects , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Benzamides/chemistry , Benzamides/pharmacokinetics , Benzamides/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Dipeptides/chemistry , Dipeptides/pharmacokinetics , Dipeptides/pharmacology , Focal Adhesion Kinase 1/metabolism , Humans , Mice , Molecular Structure , Ubiquitin-Protein Ligases/metabolism
8.
Angew Chem Int Ed Engl ; 59(47): 21096-21105, 2020 11 16.
Article in English | MEDLINE | ID: mdl-32745361

ABSTRACT

Advances in genomic analyses enable the identification of new proteins that are associated with disease. To validate these targets, tool molecules are required to demonstrate that a ligand can have a disease-modifying effect. Currently, as tools are reported for only a fraction of the proteome, platforms for ligand discovery are essential to leverage insights from genomic analyses. Fragment screening offers an efficient approach to explore chemical space. Presented here is a fragment-screening platform, termed PhABits (PhotoAffinity Bits), which utilizes a library of photoreactive fragments to covalently capture fragment-protein interactions. Hits can be profiled to determine potency and the site of crosslinking, and subsequently developed as reporters in a competitive displacement assay to identify novel hit matter. The PhABit platform is envisioned to be widely applicable to novel protein targets, identifying starting points in the development of therapeutics.


Subject(s)
Antineoplastic Agents/analysis , Bridged Bicyclo Compounds, Heterocyclic/analysis , Cross-Linking Reagents/chemistry , Photoaffinity Labels/chemistry , Pyrazoles/analysis , Quinoxalines/analysis , Sulfonamides/analysis , Vemurafenib/analysis , Antineoplastic Agents/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Humans , Ligands , Molecular Structure , Proteins/antagonists & inhibitors , Proteins/chemistry , Pyrazoles/pharmacology , Quinoxalines/pharmacology , Sulfonamides/pharmacology , Vemurafenib/pharmacology
9.
J Am Chem Soc ; 141(6): 2703-2712, 2019 02 13.
Article in English | MEDLINE | ID: mdl-30657686

ABSTRACT

Modification of proteins with polyubiquitin chains is a key regulatory mechanism to control cellular behavior and alterations in the ubiquitin system are linked to many diseases. Linear (M1-linked) polyubiquitin chains play pivotal roles in several cellular signaling pathways mediating immune and inflammatory responses and apoptotic cell death. These chains are formed by the linear ubiquitin chain assembly complex (LUBAC), a multiprotein E3 ligase that consists of 3 subunits, HOIP, HOIL-1L, and SHARPIN. Herein, we describe the discovery of inhibitors targeting the active site cysteine of the catalytic subunit HOIP using fragment-based covalent ligand screening. We report the synthesis of a diverse library of electrophilic fragments and demonstrate an integrated use of protein LC-MS, biochemical ubiquitination assays, chemical synthesis, and protein crystallography to enable the first structure-based development of covalent inhibitors for an RBR E3 ligase. Furthermore, using cell-based assays and chemoproteomics, we demonstrate that these compounds effectively penetrate mammalian cells to label and inhibit HOIP and NF-κB activation, making them suitable hits for the development of selective probes to study LUBAC biology. Our results illustrate the power of fragment-based covalent ligand screening to discover lead compounds for challenging targets, which holds promise to be a general approach for the development of cell-permeable inhibitors of thioester-forming E3 ubiquitin ligases.


Subject(s)
Enzyme Inhibitors/pharmacology , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/metabolism , Amino Acid Sequence , Drug Evaluation, Preclinical , Enzyme Inhibitors/metabolism , HEK293 Cells , Humans , Ligands , MCF-7 Cells , Models, Molecular , Protein Structure, Secondary , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology , Ubiquitin-Protein Ligases/chemistry
10.
Biochem Soc Trans ; 47(1): 281-293, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30647139

ABSTRACT

The impact of structural biology on drug discovery is well documented, and the workhorse technique for the past 30 years or so has been X-ray crystallography. With the advent of several technological improvements, including direct electron detectors, automation, better microscope vacuums and lenses, phase plates and improvements in computing power enabled by GPUs, it is now possible to record and analyse images of protein structures containing high-resolution information. This review, from a pharmaceutical perspective, highlights some of the most relevant and interesting protein structures for the pharmaceutical industry and shows examples of how ligand-binding sites, membrane proteins, both big and small, pseudo symmetry and complexes are being addressed by this technique.


Subject(s)
Cryoelectron Microscopy/methods , Drug Discovery , Crystallography, X-Ray
11.
PLoS Biol ; 13(12): e1002316, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26646171

ABSTRACT

Trypanosoma brucei, the causative agent of African sleeping sickness, is transmitted to its mammalian host by the tsetse. In the fly, the parasite's surface is covered with invariant procyclin, while in the mammal it resides extracellularly in its bloodstream form (BF) and is densely covered with highly immunogenic Variant Surface Glycoprotein (VSG). In the BF, the parasite varies this highly immunogenic surface VSG using a repertoire of ~2500 distinct VSG genes. Recent reports in mammalian systems point to a role for histone acetyl-lysine recognizing bromodomain proteins in the maintenance of stem cell fate, leading us to hypothesize that bromodomain proteins may maintain the BF cell fate in trypanosomes. Using small-molecule inhibitors and genetic mutants for individual bromodomain proteins, we performed RNA-seq experiments that revealed changes in the transcriptome similar to those seen in cells differentiating from the BF to the insect stage. This was recapitulated at the protein level by the appearance of insect-stage proteins on the cell surface. Furthermore, bromodomain inhibition disrupts two major BF-specific immune evasion mechanisms that trypanosomes harness to evade mammalian host antibody responses. First, monoallelic expression of the antigenically varied VSG is disrupted. Second, rapid internalization of antibodies bound to VSG on the surface of the trypanosome is blocked. Thus, our studies reveal a role for trypanosome bromodomain proteins in maintaining bloodstream stage identity and immune evasion. Importantly, bromodomain inhibition leads to a decrease in virulence in a mouse model of infection, establishing these proteins as potential therapeutic drug targets for trypanosomiasis. Our 1.25Å resolution crystal structure of a trypanosome bromodomain in complex with I-BET151 reveals a novel binding mode of the inhibitor, which serves as a promising starting point for rational drug design.


Subject(s)
Models, Molecular , Protozoan Proteins/metabolism , Transcription Factors/metabolism , Trypanosoma brucei brucei/physiology , Amino Acid Substitution , Animals , Binding Sites , Cell Line , Gene Knockdown Techniques , Gene Knockout Techniques , Immune Evasion , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Conformation , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Survival Analysis , Transcription Factors/antagonists & inhibitors , Transcription Factors/chemistry , Transcription Factors/genetics , Trypanocidal Agents/pharmacology , Trypanocidal Agents/therapeutic use , Trypanosoma brucei brucei/drug effects , Trypanosoma brucei brucei/immunology , Trypanosoma brucei brucei/pathogenicity , Trypanosomiasis, African/drug therapy , Trypanosomiasis, African/parasitology , Trypanosomiasis, African/physiopathology , Virulence
12.
Nature ; 488(7411): 404-8, 2012 Aug 16.
Article in English | MEDLINE | ID: mdl-22842901

ABSTRACT

The jumonji (JMJ) family of histone demethylases are Fe2+- and α-ketoglutarate-dependent oxygenases that are essential components of regulatory transcriptional chromatin complexes. These enzymes demethylate lysine residues in histones in a methylation-state and sequence-specific context. Considerable effort has been devoted to gaining a mechanistic understanding of the roles of histone lysine demethylases in eukaryotic transcription, genome integrity and epigenetic inheritance, as well as in development, physiology and disease. However, because of the absence of any selective inhibitors, the relevance of the demethylase activity of JMJ enzymes in regulating cellular responses remains poorly understood. Here we present a structure-guided small-molecule and chemoproteomics approach to elucidating the functional role of the H3K27me3-specific demethylase subfamily (KDM6 subfamily members JMJD3 and UTX). The liganded structures of human and mouse JMJD3 provide novel insight into the specificity determinants for cofactor, substrate and inhibitor recognition by the KDM6 subfamily of demethylases. We exploited these structural features to generate the first small-molecule catalytic site inhibitor that is selective for the H3K27me3-specific JMJ subfamily. We demonstrate that this inhibitor binds in a novel manner and reduces lipopolysaccharide-induced proinflammatory cytokine production by human primary macrophages, a process that depends on both JMJD3 and UTX. Our results resolve the ambiguity associated with the catalytic function of H3K27-specific JMJs in regulating disease-relevant inflammatory responses and provide encouragement for designing small-molecule inhibitors to allow selective pharmacological intervention across the JMJ family.


Subject(s)
Enzyme Inhibitors/pharmacology , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Macrophages/drug effects , Macrophages/immunology , Amino Acid Sequence , Animals , Biocatalysis/drug effects , Catalytic Domain , Cells, Cultured , Enzyme Inhibitors/metabolism , Evolution, Molecular , Histones/chemistry , Histones/metabolism , Humans , Inhibitory Concentration 50 , Jumonji Domain-Containing Histone Demethylases/chemistry , Jumonji Domain-Containing Histone Demethylases/classification , Jumonji Domain-Containing Histone Demethylases/metabolism , Lysine/metabolism , Macrophages/enzymology , Macrophages/metabolism , Methylation/drug effects , Mice , Models, Molecular , Substrate Specificity , Tumor Necrosis Factor-alpha/biosynthesis
13.
J Biol Chem ; 291(11): 5500-5511, 2016 Mar 11.
Article in English | MEDLINE | ID: mdl-26728464

ABSTRACT

A potent VEGF inhibitor with novel antibody architecture and antigen binding mode has been developed. The molecule, hereafter referred to as VEGF dual dAb (domain antibody), was evaluated in vitro for binding to VEGF and for potency in VEGF-driven models and compared with other anti-VEGF biologics that have been used in ocular anti-angiogenic therapeutic regimes. VEGF dual dAb is more potent than bevacizumab and ranibizumab for VEGF binding, inhibition of VEGF receptor binding assays (RBAs), and VEGF-driven in vitro models of angiogenesis and displays comparable inhibition to aflibercept (Eylea). VEGF dual dAb is dimeric, and each monomer contains two distinct anti-VEGF domain antibodies attached via linkers to a human IgG1 Fc domain. Mechanistically, the enhanced in vitro potency of VEGF dual dAb, in comparison to other anti-VEGF biologics, can be explained by increased binding stoichiometry. A consistent model of the target engagement has been built based on the x-ray complexes of each of the two isolated domain antibodies with the VEGF antigen.


Subject(s)
Bevacizumab/pharmacology , Ranibizumab/pharmacology , Receptors, Vascular Endothelial Growth Factor/pharmacology , Recombinant Fusion Proteins/pharmacology , Single-Domain Antibodies/pharmacology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Animals , Crystallography, X-Ray , Drug Discovery , HEK293 Cells , Humans , Models, Molecular , Single-Domain Antibodies/chemistry , Swine , Vascular Endothelial Growth Factor A/chemistry
14.
Nat Chem Biol ; 11(3): 189-91, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25622091

ABSTRACT

PAD4 has been strongly implicated in the pathogenesis of autoimmune, cardiovascular and oncological diseases through clinical genetics and gene disruption in mice. New selective PAD4 inhibitors binding a calcium-deficient form of the PAD4 enzyme have validated the critical enzymatic role of human and mouse PAD4 in both histone citrullination and neutrophil extracellular trap formation for, to our knowledge, the first time. The therapeutic potential of PAD4 inhibitors can now be explored.


Subject(s)
Benzimidazoles/pharmacology , Enzyme Inhibitors/pharmacology , Hydrolases/antagonists & inhibitors , Neutrophils/drug effects , Animals , Benzimidazoles/chemical synthesis , Binding, Competitive , Calcium/metabolism , Citrulline/metabolism , Enzyme Inhibitors/chemical synthesis , HEK293 Cells , Histones/metabolism , Humans , In Vitro Techniques , Mice , Models, Molecular , Protein-Arginine Deiminase Type 4 , Protein-Arginine Deiminases , Small Molecule Libraries , Substrate Specificity
15.
Nature ; 478(7370): 529-33, 2011 Oct 02.
Article in English | MEDLINE | ID: mdl-21964340

ABSTRACT

Recurrent chromosomal translocations involving the mixed lineage leukaemia (MLL) gene initiate aggressive forms of leukaemia, which are often refractory to conventional therapies. Many MLL-fusion partners are members of the super elongation complex (SEC), a critical regulator of transcriptional elongation, suggesting that aberrant control of this process has an important role in leukaemia induction. Here we use a global proteomic strategy to demonstrate that MLL fusions, as part of SEC and the polymerase-associated factor complex (PAFc), are associated with the BET family of acetyl-lysine recognizing, chromatin 'adaptor' proteins. These data provided the basis for therapeutic intervention in MLL-fusion leukaemia, via the displacement of the BET family of proteins from chromatin. We show that a novel small molecule inhibitor of the BET family, GSK1210151A (I-BET151), has profound efficacy against human and murine MLL-fusion leukaemic cell lines, through the induction of early cell cycle arrest and apoptosis. I-BET151 treatment in two human leukaemia cell lines with different MLL fusions alters the expression of a common set of genes whose function may account for these phenotypic changes. The mode of action of I-BET151 is, at least in part, due to the inhibition of transcription at key genes (BCL2, C-MYC and CDK6) through the displacement of BRD3/4, PAFc and SEC components from chromatin. In vivo studies indicate that I-BET151 has significant therapeutic value, providing survival benefit in two distinct mouse models of murine MLL-AF9 and human MLL-AF4 leukaemia. Finally, the efficacy of I-BET151 against human leukaemia stem cells is demonstrated, providing further evidence of its potent therapeutic potential. These findings establish the displacement of BET proteins from chromatin as a promising epigenetic therapy for these aggressive leukaemias.


Subject(s)
Chromatin/metabolism , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Oncogene Proteins, Fusion/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Animals , Cell Line, Tumor , Chromatin/genetics , Chromatin Immunoprecipitation , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Protein Binding/drug effects , Proteomics , Transcription, Genetic/drug effects
16.
Angew Chem Int Ed Engl ; 56(41): 12492-12497, 2017 10 02.
Article in English | MEDLINE | ID: mdl-28786545

ABSTRACT

The uridyl peptide antibiotics (UPAs), of which pacidamycin is a member, have a clinically unexploited mode of action and an unusual assembly. Perhaps the most striking feature of these molecules is the biosynthetically unique 3'-deoxyuridine that they share. This moiety is generated by an unusual, small and monomeric dehydratase, Pac13, which catalyses the dehydration of uridine-5'-aldehyde. Here we report the structural characterisation of Pac13 with a series of ligands, and gain insight into the enzyme's mechanism demonstrating that H42 is critical to the enzyme's activity and that the reaction is likely to proceed via an E1cB mechanism. The resemblance of the 3'-deoxy pacidamycin moiety with the synthetic anti-retrovirals, presents a potential opportunity for the utilisation of Pac13 in the biocatalytic generation of antiviral compounds.

17.
Nature ; 468(7327): 1119-23, 2010 Dec 23.
Article in English | MEDLINE | ID: mdl-21068722

ABSTRACT

Interaction of pathogens with cells of the immune system results in activation of inflammatory gene expression. This response, although vital for immune defence, is frequently deleterious to the host due to the exaggerated production of inflammatory proteins. The scope of inflammatory responses reflects the activation state of signalling proteins upstream of inflammatory genes as well as signal-induced assembly of nuclear chromatin complexes that support mRNA expression. Recognition of post-translationally modified histones by nuclear proteins that initiate mRNA transcription and support mRNA elongation is a critical step in the regulation of gene expression. Here we present a novel pharmacological approach that targets inflammatory gene expression by interfering with the recognition of acetylated histones by the bromodomain and extra terminal domain (BET) family of proteins. We describe a synthetic compound (I-BET) that by 'mimicking' acetylated histones disrupts chromatin complexes responsible for the expression of key inflammatory genes in activated macrophages, and confers protection against lipopolysaccharide-induced endotoxic shock and bacteria-induced sepsis. Our findings suggest that synthetic compounds specifically targeting proteins that recognize post-translationally modified histones can serve as a new generation of immunomodulatory drugs.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Gene Expression Regulation/drug effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , Inflammation , Macrophages/drug effects , Acetylation/drug effects , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/therapeutic use , Benzodiazepines , Cells, Cultured , Epigenomics , Genome-Wide Association Study , Heterocyclic Compounds, 4 or More Rings/chemistry , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Inflammation/drug therapy , Inflammation/prevention & control , Kaplan-Meier Estimate , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Models, Molecular , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Salmonella Infections/drug therapy , Salmonella Infections/immunology , Salmonella Infections/physiopathology , Salmonella Infections/prevention & control , Salmonella typhimurium , Sepsis/drug therapy , Sepsis/prevention & control , Shock, Septic/drug therapy , Shock, Septic/prevention & control
18.
Angew Chem Int Ed Engl ; 55(38): 11382-6, 2016 09 12.
Article in English | MEDLINE | ID: mdl-27530368

ABSTRACT

ATAD2 is a cancer-associated protein whose bromodomain has been described as among the least druggable of that target class. Starting from a potent lead, permeability and selectivity were improved through a dual approach: 1) using CF2 as a sulfone bio-isostere to exploit the unique properties of fluorine, and 2) using 1,3-interactions to control the conformation of a piperidine ring. This resulted in the first reported low-nanomolar, selective and cell permeable chemical probe for ATAD2.

SELECTION OF CITATIONS
SEARCH DETAIL