Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 98
Filter
1.
Nature ; 611(7937): 721-726, 2022 11.
Article in English | MEDLINE | ID: mdl-36108675

ABSTRACT

Small-ring cage hydrocarbons are popular bioisosteres (molecular replacements) for commonly found para-substituted benzene rings in drug design1. The utility of these cage structures derives from their superior pharmacokinetic properties compared with their parent aromatics, including improved solubility and reduced susceptibility to metabolism2,3. A prime example is the bicyclo[1.1.1]pentane motif, which is mainly synthesized by ring-opening of the interbridgehead bond of the strained hydrocarbon [1.1.1]propellane with radicals or anions4. By contrast, scaffolds mimicking meta-substituted arenes are lacking because of the challenge of synthesizing saturated isosteres that accurately reproduce substituent vectors5. Here we show that bicyclo[3.1.1]heptanes (BCHeps), which are hydrocarbons for which the bridgehead substituents map precisely onto the geometry of meta-substituted benzenes, can be conveniently accessed from [3.1.1]propellane. We found that [3.1.1]propellane can be synthesized on a multigram scale, and readily undergoes a range of radical-based transformations to generate medicinally relevant carbon- and heteroatom-substituted BCHeps, including pharmaceutical analogues. Comparison of the absorption, distribution, metabolism and excretion (ADME) properties of these analogues reveals enhanced metabolic stability relative to their parent arene-containing drugs, validating the potential of this meta-arene analogue as an sp3-rich motif in drug design. Collectively, our results show that BCHeps can be prepared on useful scales using a variety of methods, offering a new surrogate for meta-substituted benzene rings for implementation in drug discovery programmes.


Subject(s)
Bridged Bicyclo Compounds , Drug Design , Heptanes , Anions/chemistry , Benzene/chemistry , Bridged Bicyclo Compounds/chemical synthesis , Bridged Bicyclo Compounds/chemistry , Drug Discovery , Heptanes/chemical synthesis , Heptanes/chemistry , Pentanes/chemical synthesis , Pentanes/chemistry , Solubility
2.
Mol Cell Proteomics ; 22(8): 100609, 2023 08.
Article in English | MEDLINE | ID: mdl-37385347

ABSTRACT

Dampening functional levels of the mitochondrial deubiquitylating enzyme Ubiquitin-specific protease 30 (USP30) has been suggested as an effective therapeutic strategy against neurodegenerative disorders such as Parkinson's Disease. USP30 inhibition may counteract the deleterious effects of impaired turnover of damaged mitochondria, which is inherent to both familial and sporadic forms of the disease. Small-molecule inhibitors targeting USP30 are currently in development, but little is known about their precise nature of binding to the protein. We have integrated biochemical and structural approaches to gain novel mechanistic insights into USP30 inhibition by a small-molecule benzosulfonamide-containing compound, USP30inh. Activity-based protein profiling mass spectrometry confirmed target engagement, high selectivity, and potency of USP30inh for USP30 against 49 other deubiquitylating enzymes in a neuroblastoma cell line. In vitro characterization of USP30inh enzyme kinetics inferred slow and tight binding behavior, which is comparable with features of covalent modification of USP30. Finally, we blended hydrogen-deuterium exchange mass spectrometry and computational docking to elucidate the molecular architecture and geometry of USP30 complex formation with USP30inh, identifying structural rearrangements at the cleft of the USP30 thumb and palm subdomains. These studies suggest that USP30inh binds to this thumb-palm cleft, which guides the ubiquitin C terminus into the active site, thereby preventing ubiquitin binding and isopeptide bond cleavage, and confirming its importance in the inhibitory process. Our data will pave the way for the design and development of next-generation inhibitors targeting USP30 and associated deubiquitinylases.


Subject(s)
Deubiquitinating Enzymes , Mitophagy , Deubiquitinating Enzymes/antagonists & inhibitors , Deubiquitinating Enzymes/metabolism , Mitochondrial Proteins/metabolism , Mitophagy/physiology , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Sulfonamides/pharmacology
3.
J Biol Chem ; 299(12): 105382, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37866628

ABSTRACT

Proteomic studies have identified moesin (MSN), a protein containing a four-point-one, ezrin, radixin, moesin (FERM) domain, and the receptor CD44 as hub proteins found within a coexpression module strongly linked to Alzheimer's disease (AD) traits and microglia. These proteins are more abundant in Alzheimer's patient brains, and their levels are positively correlated with cognitive decline, amyloid plaque deposition, and neurofibrillary tangle burden. The MSN FERM domain interacts with the phospholipid phosphatidylinositol 4,5-bisphosphate (PIP2) and the cytoplasmic tail of CD44. Inhibiting the MSN-CD44 interaction may help limit AD-associated neuronal damage. Here, we investigated the feasibility of developing inhibitors that target this protein-protein interaction. We have employed structural, mutational, and phage-display studies to examine how CD44 binds to the FERM domain of MSN. Interestingly, we have identified an allosteric site located close to the PIP2 binding pocket that influences CD44 binding. These findings suggest a mechanism in which PIP2 binding to the FERM domain stimulates CD44 binding through an allosteric effect, leading to the formation of a neighboring pocket capable of accommodating a receptor tail. Furthermore, high-throughput screening of a chemical library identified two compounds that disrupt the MSN-CD44 interaction. One compound series was further optimized for biochemical activity, specificity, and solubility. Our results suggest that the FERM domain holds potential as a drug development target. Small molecule preliminary leads generated from this study could serve as a foundation for additional medicinal chemistry efforts with the goal of controlling microglial activity in AD by modifying the MSN-CD44 interaction.


Subject(s)
Alzheimer Disease , Protein Binding , Humans , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , FERM Domains , Hyaluronan Receptors/metabolism , Protein Binding/drug effects , Proteomics
4.
J Biol Chem ; 296: 100521, 2021.
Article in English | MEDLINE | ID: mdl-33684443

ABSTRACT

The human dedicator of cytokinesis (DOCK) family consists of 11 structurally conserved proteins that serve as atypical RHO guanine nucleotide exchange factors (RHO GEFs). These regulatory proteins act as mediators in numerous cellular cascades that promote cytoskeletal remodeling, playing roles in various crucial processes such as differentiation, migration, polarization, and axon growth in neurons. At the molecular level, DOCK DHR2 domains facilitate nucleotide dissociation from small GTPases, a process that is otherwise too slow for rapid spatiotemporal control of cellular signaling. Here, we provide an overview of the biological and structural characteristics for the various DOCK proteins and describe how they differ from other RHO GEFs and between DOCK subfamilies. The expression of the family varies depending on cell or tissue type, and they are consequently implicated in a broad range of disease phenotypes, particularly in the brain. A growing body of available structural information reveals the mechanism by which the catalytic DHR2 domain elicits nucleotide dissociation and also indicates strategies for the discovery and design of high-affinity small-molecule inhibitors. Such compounds could serve as chemical probes to interrogate the cellular function and provide starting points for drug discovery of this important class of enzymes.


Subject(s)
Rho Guanine Nucleotide Exchange Factors/metabolism , Catalytic Domain , GTP Phosphohydrolases/metabolism , Protein Conformation , Rho Guanine Nucleotide Exchange Factors/chemistry
5.
Biol Chem ; 403(4): 391-402, 2022 03 28.
Article in English | MEDLINE | ID: mdl-35191283

ABSTRACT

Ubiquitination is a key regulatory mechanism vital for maintenance of cellular homeostasis. Protein degradation is induced by E3 ligases via attachment of ubiquitin chains to substrates. Pharmacological exploitation of this phenomenon via targeted protein degradation (TPD) can be achieved with molecular glues or bifunctional molecules facilitating the formation of ternary complexes between an E3 ligase and a given protein of interest (POI), resulting in ubiquitination of the substrate and subsequent proteolysis by the proteasome. Recently, the development of novel covalent fragment screening approaches has enabled the identification of first-in-class ligands for E3 ligases and deubiquitinases revealing so far unexplored binding sites which highlights the potential of these methods to uncover and expand druggable space for new target classes.


Subject(s)
Proteasome Endopeptidase Complex , Ubiquitin , Ligands , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
6.
J Biol Chem ; 295(52): 17973-17985, 2020 12 25.
Article in English | MEDLINE | ID: mdl-33028632

ABSTRACT

The molecular mechanisms of reduced frataxin (FXN) expression in Friedreich's ataxia (FRDA) are linked to epigenetic modification of the FXN locus caused by the disease-associated GAA expansion. Here, we identify that SUV4-20 histone methyltransferases, specifically SUV4-20 H1, play an important role in the regulation of FXN expression and represent a novel therapeutic target. Using a human FXN-GAA-Luciferase repeat expansion genomic DNA reporter model of FRDA, we screened the Structural Genomics Consortium epigenetic probe collection. We found that pharmacological inhibition of the SUV4-20 methyltransferases by the tool compound A-196 increased the expression of FXN by ∼1.5-fold in the reporter cell line. In several FRDA cell lines and patient-derived primary peripheral blood mononuclear cells, A-196 increased FXN expression by up to 2-fold, an effect not seen in WT cells. SUV4-20 inhibition was accompanied by a reduction in H4K20me2 and H4K20me3 and an increase in H4K20me1, but only modest (1.4-7.8%) perturbation in genome-wide expression was observed. Finally, based on the structural activity relationship and crystal structure of A-196, novel small molecule A-196 analogs were synthesized and shown to give a 20-fold increase in potency for increasing FXN expression. Overall, our results suggest that histone methylation is important in the regulation of FXN expression and highlight SUV4-20 H1 as a potential novel therapeutic target for FRDA.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Fibroblasts/pathology , Friedreich Ataxia/pathology , Gene Silencing , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Iron-Binding Proteins/metabolism , Case-Control Studies , Fibroblasts/metabolism , Friedreich Ataxia/genetics , Friedreich Ataxia/metabolism , Heterochromatin , Humans , Iron-Binding Proteins/antagonists & inhibitors , Iron-Binding Proteins/genetics , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Frataxin
7.
Angew Chem Int Ed Engl ; 59(16): 6342-6366, 2020 04 16.
Article in English | MEDLINE | ID: mdl-30869179

ABSTRACT

The Ras superfamily of small GTPases are guanine-nucleotide-dependent switches essential for numerous cellular processes. Mutations or dysregulation of these proteins are associated with many diseases, but unsuccessful attempts to target the small GTPases directly have resulted in them being classed as "undruggable". The GTP-dependent signaling of these proteins is controlled by their regulators; guanine nucleotide exchange factors (GEFs), GTPase activating proteins (GAPs), and in the Rho and Rab subfamilies, guanine nucleotide dissociation inhibitors (GDIs). This review covers the recent small molecule and biologics strategies to target the small GTPases through their regulators. It seeks to critically re-evaluate recent chemical biology practice, such as the presence of PAINs motifs and the cell-based readout using compounds that are weakly potent or of unknown specificity. It highlights the vast scope of potential approaches for targeting the small GTPases in the future through their regulatory proteins.


Subject(s)
Monomeric GTP-Binding Proteins/metabolism , Small Molecule Libraries/chemistry , Binding Sites , Drug Evaluation, Preclinical , Humans , Molecular Dynamics Simulation , Monomeric GTP-Binding Proteins/antagonists & inhibitors , Monomeric GTP-Binding Proteins/classification , Peptides/chemistry , Peptides/metabolism , Phylogeny , Protein Binding , Small Molecule Libraries/metabolism , Structure-Activity Relationship
8.
J Am Chem Soc ; 141(22): 8951-8968, 2019 06 05.
Article in English | MEDLINE | ID: mdl-31060360

ABSTRACT

Covalent probes can display unmatched potency, selectivity, and duration of action; however, their discovery is challenging. In principle, fragments that can irreversibly bind their target can overcome the low affinity that limits reversible fragment screening, but such electrophilic fragments were considered nonselective and were rarely screened. We hypothesized that mild electrophiles might overcome the selectivity challenge and constructed a library of 993 mildly electrophilic fragments. We characterized this library by a new high-throughput thiol-reactivity assay and screened them against 10 cysteine-containing proteins. Highly reactive and promiscuous fragments were rare and could be easily eliminated. In contrast, we found hits for most targets. Combining our approach with high-throughput crystallography allowed rapid progression to potent and selective probes for two enzymes, the deubiquitinase OTUB2 and the pyrophosphatase NUDT7. No inhibitors were previously known for either. This study highlights the potential of electrophile-fragment screening as a practical and efficient tool for covalent-ligand discovery.


Subject(s)
Drug Evaluation, Preclinical/methods , Electrons , HEK293 Cells , Humans , Ligands , Models, Molecular , Molecular Weight , Protein Conformation , Time Factors
9.
Angew Chem Int Ed Engl ; 58(4): 1007-1012, 2019 01 21.
Article in English | MEDLINE | ID: mdl-30589164

ABSTRACT

Bromodomain-containing proteins are epigenetic modulators involved in a wide range of cellular processes, from recruitment of transcription factors to pathological disruption of gene regulation and cancer development. Since the druggability of these acetyl-lysine reader domains was established, efforts were made to develop potent and selective inhibitors across the entire family. Here we report the development of a small molecule-based approach to covalently modify recombinant and endogenous bromodomain-containing proteins by targeting a conserved lysine and a tyrosine residue in the variable ZA or BC loops. Moreover, the addition of a reporter tag allowed in-gel visualization and pull-down of the desired bromodomains.


Subject(s)
Carbamates/chemistry , Histones/chemistry , Lysine/chemistry , Protein Domains , Pyridazines/chemistry , Triazoles/chemistry , Acetylation , Amino Acid Sequence , Binding Sites , Conserved Sequence , Molecular Docking Simulation , Protein Binding
10.
Angew Chem Int Ed Engl ; 58(2): 515-519, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30431220

ABSTRACT

Histone lysine demethylases (KDMs) are involved in the dynamic regulation of gene expression and they play a critical role in several biological processes. Achieving selectivity over the different KDMs has been a major challenge for KDM inhibitor development. Here we report potent and selective KDM5 covalent inhibitors designed to target cysteine residues only present in the KDM5 sub-family. The covalent binding to the targeted proteins was confirmed by MS and time-dependent inhibition. Additional competition assays show that compounds were non 2-OG competitive. Target engagement and ChIP-seq analysis showed that the compounds inhibited the KDM5 members in cells at nano- to micromolar levels and induce a global increase of the H3K4me3 mark at transcriptional start sites.

11.
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
12.
J Org Chem ; 83(16): 9510-9516, 2018 08 17.
Article in English | MEDLINE | ID: mdl-29932332

ABSTRACT

Sulfonimidamides are an emerging bioisosteric replacement in medicinal chemistry projects, and therefore new chemistries are necessary to access this functionality. The general synthesis of CF3-sulfonimidamides from an activated bench-stable transfer reagent is described. A diverse reaction scope is demonstrated, with a wide range of nucleophilic amines being tolerated in this transformation. The CF3-sulfonimidamides obtained contain an additional diversity point, in the form a protected imine, that could be unmasked to allow late stage modifications.

13.
Bioorg Med Chem ; 26(11): 2965-2972, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29567296

ABSTRACT

The polyadenosine-diphosphate-ribose polymerase 14 (PARP14) has been implicated in DNA damage response pathways for homologous recombination. PARP14 contains three (ADP ribose binding) macrodomains (MD) whose exact contribution to overall PARP14 function in pathology remains unclear. A medium throughput screen led to the identification of N-(2(-9H-carbazol-1-yl)phenyl)acetamide (GeA-69, 1) as a novel allosteric PARP14 MD2 (second MD of PARP14) inhibitor. We herein report medicinal chemistry around this novel chemotype to afford a sub-micromolar PARP14 MD2 inhibitor. This chemical series provides a novel starting point for further development of PARP14 chemical probes.


Subject(s)
Cysteine Endopeptidases/chemistry , Drug Discovery , Poly(ADP-ribose) Polymerase Inhibitors/chemical synthesis , Poly(ADP-ribose) Polymerases/chemistry , Allosteric Regulation , Carbazoles/chemistry , Humans , Inhibitory Concentration 50 , Models, Biological , Molecular Docking Simulation , Molecular Structure , Poly(ADP-ribose) Polymerase Inhibitors/chemistry , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/drug effects , Structure-Activity Relationship
14.
Proc Natl Acad Sci U S A ; 112(34): 10768-73, 2015 Aug 25.
Article in English | MEDLINE | ID: mdl-26261308

ABSTRACT

Th17 responses are critical to a variety of human autoimmune diseases, and therapeutic targeting with monoclonal antibodies against IL-17 and IL-23 has shown considerable promise. Here, we report data to support selective bromodomain blockade of the transcriptional coactivators CBP (CREB binding protein) and p300 as an alternative approach to inhibit human Th17 responses. We show that CBP30 has marked molecular specificity for the bromodomains of CBP and p300, compared with 43 other bromodomains. In unbiased cellular testing on a diverse panel of cultured primary human cells, CBP30 reduced immune cell production of IL-17A and other proinflammatory cytokines. CBP30 also inhibited IL-17A secretion by Th17 cells from healthy donors and patients with ankylosing spondylitis and psoriatic arthritis. Transcriptional profiling of human T cells after CBP30 treatment showed a much more restricted effect on gene expression than that observed with the pan-BET (bromo and extraterminal domain protein family) bromodomain inhibitor JQ1. This selective targeting of the CBP/p300 bromodomain by CBP30 will potentially lead to fewer side effects than with the broadly acting epigenetic inhibitors currently in clinical trials.


Subject(s)
Benzimidazoles/pharmacology , Immunosuppressive Agents/pharmacology , Interleukin-17/metabolism , Isoxazoles/pharmacology , Th17 Cells/drug effects , p300-CBP Transcription Factors/antagonists & inhibitors , Adult , Aged , Arthritis, Psoriatic/metabolism , Arthritis, Psoriatic/pathology , Azepines/pharmacology , Benzimidazoles/chemistry , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Calorimetry , Cells, Cultured , Crystallography, X-Ray , Drug Evaluation, Preclinical , Female , Gene Expression Regulation/drug effects , Humans , Immunosuppressive Agents/chemistry , Interleukin-17/biosynthesis , Interleukin-17/genetics , Isoxazoles/chemistry , Kinetics , Male , Middle Aged , Models, Molecular , Molecular Structure , Protein Conformation , Protein Structure, Tertiary/drug effects , Recombinant Proteins/metabolism , Spondylitis, Ankylosing/metabolism , Spondylitis, Ankylosing/pathology , Structure-Activity Relationship , Th17 Cells/immunology , Triazoles/pharmacology
15.
Angew Chem Int Ed Engl ; 57(50): 16302-16307, 2018 12 10.
Article in English | MEDLINE | ID: mdl-30288907

ABSTRACT

YEATS domain (YD) containing proteins are an emerging class of epigenetic targets in drug discovery. Dysregulation of these modified lysine-binding proteins has been linked to the onset and progression of cancers. We herein report the discovery and characterisation of the first small-molecule chemical probe, SGC-iMLLT, for the YD of MLLT1 (ENL/YEATS1) and MLLT3 (AF9/YEATS3). SGC-iMLLT is a potent and selective inhibitor of MLLT1/3-histone interactions. Excellent selectivity over other human YD proteins (YEATS2/4) and bromodomains was observed. Furthermore, our probe displays cellular target engagement of MLLT1 and MLLT3. The first small-molecule X-ray co-crystal structures with the MLLT1 YD are also reported. This first-in-class probe molecule can be used to understand MLLT1/3-associated biology and the therapeutic potential of small-molecule YD inhibitors.


Subject(s)
Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/chemistry , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/chemistry , Small Molecule Libraries/chemistry , Transcription Factors/antagonists & inhibitors , Transcription Factors/chemistry , Crystallography, X-Ray , Histones/metabolism , Humans , Molecular Docking Simulation , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Protein Domains , Protein Interaction Maps/drug effects , Small Molecule Libraries/pharmacology , Transcription Factors/metabolism
16.
Angew Chem Int Ed Engl ; 56(49): 15555-15559, 2017 12 04.
Article in English | MEDLINE | ID: mdl-28976073

ABSTRACT

Histone lysine demethylases (KDMs) are of critical importance in the epigenetic regulation of gene expression, yet there are few selective, cell-permeable inhibitors or suitable tool compounds for these enzymes. We describe the discovery of a new class of inhibitor that is highly potent towards the histone lysine demethylases KDM2A/7A. A modular synthetic approach was used to explore the chemical space and accelerate the investigation of key structure-activity relationships, leading to the development of a small molecule with around 75-fold selectivity towards KDM2A/7A versus other KDMs, as well as cellular activity at low micromolar concentrations.


Subject(s)
Drug Discovery , Enzyme Inhibitors/pharmacology , F-Box Proteins/antagonists & inhibitors , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , F-Box Proteins/metabolism , HeLa Cells , Humans , Jumonji Domain-Containing Histone Demethylases/metabolism , Molecular Structure , Structure-Activity Relationship
17.
Angew Chem Int Ed Engl ; 56(3): 827-831, 2017 01 16.
Article in English | MEDLINE | ID: mdl-27966810

ABSTRACT

The p300/CBP-associated factor (PCAF) and related GCN5 bromodomain-containing lysine acetyl transferases are members of subfamily I of the bromodomain phylogenetic tree. Iterative cycles of rational inhibitor design and biophysical characterization led to the discovery of the triazolopthalazine-based L-45 (dubbed L-Moses) as the first potent, selective, and cell-active PCAF bromodomain (Brd) inhibitor. Synthesis from readily available (1R,2S)-(-)-norephedrine furnished L-45 in enantiopure form. L-45 was shown to disrupt PCAF-Brd histone H3.3 interaction in cells using a nanoBRET assay, and a co-crystal structure of L-45 with the homologous Brd PfGCN5 from Plasmodium falciparum rationalizes the high selectivity for PCAF and GCN5 bromodomains. Compound L-45 shows no observable cytotoxicity in peripheral blood mononuclear cells (PBMC), good cell-permeability, and metabolic stability in human and mouse liver microsomes, supporting its potential for in vivo use.


Subject(s)
Azo Compounds/pharmacology , Drug Discovery , Hydralazine/pharmacology , Molecular Probes/pharmacology , p300-CBP Transcription Factors/antagonists & inhibitors , Azo Compounds/chemical synthesis , Azo Compounds/chemistry , Dose-Response Relationship, Drug , Hydralazine/chemical synthesis , Hydralazine/chemistry , Molecular Probes/chemical synthesis , Molecular Probes/chemistry , Molecular Structure , Structure-Activity Relationship
18.
Drug Discov Today Technol ; 19: 73-80, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27769361

ABSTRACT

The bromodomain family of proteins are 'readers' of acetylated lysines of histones, a key mark in the epigenetic code of gene regulation. Without high quality chemical probes with which to study these proteins, their biological function, and potential use in therapeutics, remains unknown. Recently, a number of chemical ligands were reported for the previously unprobed bromodomain proteins BRD7 and BRD9. Herein the development and characterisation of probes against these proteins is detailed, including the preliminary biological activity of BRD7 and BRD9 assessed using these probes. Future studies utilising these chemically-diverse compounds in parallel will allow for a confident assessment of the role of BRD7/9, and give multiple entry points into any subsequent pharmaceutical programs.


Subject(s)
Chromosomal Proteins, Non-Histone/chemistry , Protein Domains , Transcription Factors/chemistry , Animals , Chromosomal Proteins, Non-Histone/metabolism , Drug Design , Humans , Ligands , Transcription Factors/metabolism
19.
Proc Natl Acad Sci U S A ; 110(49): 19754-9, 2013 Dec 03.
Article in English | MEDLINE | ID: mdl-24248379

ABSTRACT

Bromodomains have emerged as attractive candidates for the development of inhibitors targeting gene transcription. Inhibitors of the bromo and extraterminal (BET) family recently showed promising activity in diverse disease models. However, the pleiotropic nature of BET proteins regulating tissue-specific transcription has raised safety concerns and suggested that attempts should be made for domain-specific targeting. Here, we report that RVX-208, a compound currently in phase II clinical trials, is a BET bromodomain inhibitor specific for second bromodomains (BD2s). Cocrystal structures revealed binding modes of RVX-208 and its synthetic precursor, and fluorescent recovery after photobleaching demonstrated that RVX-208 displaces BET proteins from chromatin. However, gene-expression data showed that BD2 inhibition only modestly affects BET-dependent gene transcription. Our data demonstrate the feasibility of specific targeting within the BET family resulting in different transcriptional outcomes and highlight the importance of BD1 in transcriptional regulation.


Subject(s)
Models, Molecular , Quinazolines/chemistry , Quinazolines/pharmacology , Transcription Factors/antagonists & inhibitors , Transcription Factors/chemistry , Crystallization , Fluorescence Recovery After Photobleaching , Hep G2 Cells , Humans , Molecular Structure , Oligonucleotide Array Sequence Analysis , Protein Binding , Protein Structure, Tertiary/physiology , Quinazolinones
20.
Angew Chem Int Ed Engl ; 54(21): 6217-21, 2015 May 18.
Article in English | MEDLINE | ID: mdl-25864491

ABSTRACT

The bromodomain-containing proteins BRD9 and BRD7 are part of the human SWI/SNF chromatin-remodeling complexes BAF and PBAF. To date, no selective inhibitor for BRD7/9 has been reported despite its potential value as a biological tool or as a lead for future therapeutics. The quinolone-fused lactam LP99 is now reported as the first potent and selective inhibitor of the BRD7 and BRD9 bromodomains. Development of LP99 from a fragment hit was expedited through balancing structure-based inhibitor design and biophysical characterization against tractable chemical synthesis: Complexity-building nitro-Mannich/lactamization cascade processes allowed for early structure-activity relationship studies whereas an enantioselective organocatalytic nitro-Mannich reaction enabled the synthesis of the lead scaffold in enantioenriched form and on scale. This epigenetic probe was shown to inhibit the association of BRD7 and BRD9 to acetylated histones in vitro and in cells. Moreover, LP99 was used to demonstrate that BRD7/9 plays a role in regulating pro-inflammatory cytokine secretion.


Subject(s)
Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Drug Discovery , Lactams/chemistry , Lactams/pharmacology , Transcription Factors/antagonists & inhibitors , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/metabolism , Humans , Models, Molecular , Transcription Factors/chemistry , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL