Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Genes Dev ; 38(1-2): 70-94, 2024 02 13.
Article in English | MEDLINE | ID: mdl-38316520

ABSTRACT

Since genome instability can drive cancer initiation and progression, cells have evolved highly effective and ubiquitous DNA damage response (DDR) programs. However, some cells (for example, in skin) are normally exposed to high levels of DNA-damaging agents. Whether such high-risk cells possess lineage-specific mechanisms that tailor DNA repair to the tissue remains largely unknown. Using melanoma as a model, we show here that the microphthalmia-associated transcription factor MITF, a lineage addition oncogene that coordinates many aspects of melanocyte and melanoma biology, plays a nontranscriptional role in shaping the DDR. On exposure to DNA-damaging agents, MITF is phosphorylated at S325, and its interactome is dramatically remodeled; most transcription cofactors dissociate, and instead MITF interacts with the MRE11-RAD50-NBS1 (MRN) complex. Consequently, cells with high MITF levels accumulate stalled replication forks and display defects in homologous recombination-mediated repair associated with impaired MRN recruitment to DNA damage. In agreement with this, high MITF levels are associated with increased single-nucleotide and copy number variant burdens in melanoma. Significantly, the SUMOylation-defective MITF-E318K melanoma predisposition mutation recapitulates the effects of DNA-PKcs-phosphorylated MITF. Our data suggest that a nontranscriptional function of a lineage-restricted transcription factor contributes to a tissue-specialized modulation of the DDR that can impact cancer initiation.


Subject(s)
Melanoma , Humans , Melanoma/genetics , Microphthalmia-Associated Transcription Factor/genetics , DNA Damage , Genomic Instability/genetics , DNA
2.
Cell ; 150(4): 673-84, 2012 Aug 17.
Article in English | MEDLINE | ID: mdl-22901802

ABSTRACT

A pharmacologic approach to male contraception remains a longstanding challenge in medicine. Toward this objective, we explored the spermatogenic effects of a selective small-molecule inhibitor (JQ1) of the bromodomain and extraterminal (BET) subfamily of epigenetic reader proteins. Here, we report potent inhibition of the testis-specific member BRDT, which is essential for chromatin remodeling during spermatogenesis. Biochemical and crystallographic studies confirm that occupancy of the BRDT acetyl-lysine binding pocket by JQ1 prevents recognition of acetylated histone H4. Treatment of mice with JQ1 reduced seminiferous tubule area, testis size, and spermatozoa number and motility without affecting hormone levels. Although JQ1-treated males mate normally, inhibitory effects of JQ1 evident at the spermatocyte and round spermatid stages cause a complete and reversible contraceptive effect. These data establish a new contraceptive that can cross the blood:testis boundary and inhibit bromodomain activity during spermatogenesis, providing a lead compound targeting the male germ cell for contraception.


Subject(s)
Azepines/pharmacology , Contraceptive Agents, Male/pharmacology , Nuclear Proteins/antagonists & inhibitors , Triazoles/pharmacology , Animals , Azepines/chemistry , Blood-Testis Barrier , Contraceptive Agents, Male/chemistry , Female , Humans , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Models, Molecular , Nuclear Proteins/chemistry , Protein Structure, Tertiary , Sperm Count , Sperm Motility/drug effects , Spermatozoa/drug effects , Testis/cytology , Testis/drug effects , Triazoles/chemistry
3.
Cell ; 149(1): 214-31, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22464331

ABSTRACT

Bromodomains (BRDs) are protein interaction modules that specifically recognize ε-N-lysine acetylation motifs, a key event in the reading process of epigenetic marks. The 61 BRDs in the human genome cluster into eight families based on structure/sequence similarity. Here, we present 29 high-resolution crystal structures, covering all BRD families. Comprehensive crossfamily structural analysis identifies conserved and family-specific structural features that are necessary for specific acetylation-dependent substrate recognition. Screening of more than 30 representative BRDs against systematic histone-peptide arrays identifies new BRD substrates and reveals a strong influence of flanking posttranslational modifications, such as acetylation and phosphorylation, suggesting that BRDs recognize combinations of marks rather than singly acetylated sequences. We further uncovered a structural mechanism for the simultaneous binding and recognition of diverse diacetyl-containing peptides by BRD4. These data provide a foundation for structure-based drug design of specific inhibitors for this emerging target family.


Subject(s)
Histones/chemistry , Protein Processing, Post-Translational , Protein Structure, Tertiary , Acetylation , Amino Acid Sequence , Animals , Crystallography, X-Ray , Genome, Human , Histones/metabolism , Humans , Lysine/metabolism , Models, Molecular , Molecular Sequence Data , Phylogeny , Protein Interaction Domains and Motifs , Proteome/analysis
4.
Mol Cell ; 73(3): 621-638.e17, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30554943

ABSTRACT

Targeting bromodomains (BRDs) of the bromo-and-extra-terminal (BET) family offers opportunities for therapeutic intervention in cancer and other diseases. Here, we profile the interactomes of BRD2, BRD3, BRD4, and BRDT following treatment with the pan-BET BRD inhibitor JQ1, revealing broad rewiring of the interaction landscape, with three distinct classes of behavior for the 603 unique interactors identified. A group of proteins associate in a JQ1-sensitive manner with BET BRDs through canonical and new binding modes, while two classes of extra-terminal (ET)-domain binding motifs mediate acetylation-independent interactions. Last, we identify an unexpected increase in several interactions following JQ1 treatment that define negative functions for BRD3 in the regulation of rRNA synthesis and potentially RNAPII-dependent gene expression that result in decreased cell proliferation. Together, our data highlight the contributions of BET protein modules to their interactomes allowing for a better understanding of pharmacological rewiring in response to JQ1.


Subject(s)
Antineoplastic Agents/pharmacology , Azepines/pharmacology , Molecular Targeted Therapy/methods , Neoplasms/drug therapy , Nuclear Proteins/antagonists & inhibitors , Protein Interaction Maps/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , RNA-Binding Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Triazoles/pharmacology , Antineoplastic Agents/chemistry , Azepines/chemistry , Cell Cycle Proteins , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic , HEK293 Cells , HeLa Cells , Humans , K562 Cells , Models, Molecular , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proteomics/methods , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship , Transcription Factors/genetics , Transcription Factors/metabolism , Triazoles/chemistry
5.
EMBO J ; 37(17)2018 09 03.
Article in English | MEDLINE | ID: mdl-30026309

ABSTRACT

RIPK2 mediates inflammatory signaling by the bacteria-sensing receptors NOD1 and NOD2. Kinase inhibitors targeting RIPK2 are a proposed strategy to ameliorate NOD-mediated pathologies. Here, we reveal that RIPK2 kinase activity is dispensable for NOD2 inflammatory signaling and show that RIPK2 inhibitors function instead by antagonizing XIAP-binding and XIAP-mediated ubiquitination of RIPK2. We map the XIAP binding site on RIPK2 to the loop between ß2 and ß3 of the N-lobe of the kinase, which is in close proximity to the ATP-binding pocket. Through characterization of a new series of ATP pocket-binding RIPK2 inhibitors, we identify the molecular features that determine their inhibition of both the RIPK2-XIAP interaction, and of cellular and in vivoNOD2 signaling. Our study exemplifies how targeting of the ATP-binding pocket in RIPK2 can be exploited to interfere with the RIPK2-XIAP interaction for modulation of NOD signaling.


Subject(s)
Nod2 Signaling Adaptor Protein/metabolism , Protein Kinase Inhibitors/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinase 2/antagonists & inhibitors , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Signal Transduction/drug effects , Animals , Cell Line, Tumor , Female , Humans , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Mice , Nod2 Signaling Adaptor Protein/genetics , Receptor-Interacting Protein Serine-Threonine Kinase 2/genetics , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction/genetics , X-Linked Inhibitor of Apoptosis Protein/genetics , X-Linked Inhibitor of Apoptosis Protein/metabolism
6.
Proc Natl Acad Sci U S A ; 115(20): 5083-5088, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29712860

ABSTRACT

HLA-I molecules play a central role in antigen presentation. They typically bind 9- to 12-mer peptides, and their canonical binding mode involves anchor residues at the second and last positions of their ligands. To investigate potential noncanonical binding modes, we collected in-depth and accurate HLA peptidomics datasets covering 54 HLA-I alleles and developed algorithms to analyze these data. Our results reveal frequent (442 unique peptides) and statistically significant C-terminal extensions for at least eight alleles, including the common HLA-A03:01, HLA-A31:01, and HLA-A68:01. High resolution crystal structure of HLA-A68:01 with such a ligand uncovers structural changes taking place to accommodate C-terminal extensions and helps unraveling sequence and structural properties predictive of the presence of these extensions. Scanning viral proteomes with the C-terminal extension motifs identifies many putative epitopes and we demonstrate direct recognition by human CD8+ T cells of a 10-mer epitope from cytomegalovirus predicted to follow the C-terminal extension binding mode.


Subject(s)
Antigen Presentation/immunology , Epitopes, T-Lymphocyte/immunology , HLA Antigens/immunology , Peptide Fragments/immunology , T-Lymphocytes/immunology , Algorithms , Amino Acid Sequence , Crystallography, X-Ray , Humans , Ligands , Protein Binding
7.
Proc Natl Acad Sci U S A ; 115(37): E8668-E8677, 2018 09 11.
Article in English | MEDLINE | ID: mdl-30150413

ABSTRACT

The close integration of the MAPK, PI3K, and WNT signaling pathways underpins much of development and is deregulated in cancer. In principle, combinatorial posttranslational modification of key lineage-specific transcription factors would be an effective means to integrate critical signaling events. Understanding how this might be achieved is central to deciphering the impact of microenvironmental cues in development and disease. The microphthalmia-associated transcription factor MITF plays a crucial role in the development of melanocytes, the retinal pigment epithelium, osteoclasts, and mast cells and acts as a lineage survival oncogene in melanoma. MITF coordinates survival, differentiation, cell-cycle progression, cell migration, metabolism, and lysosome biogenesis. However, how the activity of this key transcription factor is controlled remains poorly understood. Here, we show that GSK3, downstream from both the PI3K and Wnt pathways, and BRAF/MAPK signaling converges to control MITF nuclear export. Phosphorylation of the melanocyte MITF-M isoform in response to BRAF/MAPK signaling primes for phosphorylation by GSK3, a kinase inhibited by both PI3K and Wnt signaling. Dual phosphorylation, but not monophosphorylation, then promotes MITF nuclear export by activating a previously unrecognized hydrophobic export signal. Nonmelanocyte MITF isoforms exhibit poor regulation by MAPK signaling, but instead their export is controlled by mTOR. We uncover here an unanticipated mode of MITF regulation that integrates the output of key developmental and cancer-associated signaling pathways to gate MITF flux through the import-export cycle. The results have significant implications for our understanding of melanoma progression and stem cell renewal.


Subject(s)
Cell Nucleus/metabolism , Glycogen Synthase Kinase 3/metabolism , MAP Kinase Signaling System , Microphthalmia-Associated Transcription Factor/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Active Transport, Cell Nucleus , Animals , Cell Line, Tumor , Cells, Cultured , HeLa Cells , Humans , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Microphthalmia-Associated Transcription Factor/genetics , Mutation , Phosphorylation , Protein Binding
8.
Mol Cell Proteomics ; 16(6): 1098-1110, 2017 06.
Article in English | MEDLINE | ID: mdl-28373298

ABSTRACT

The Hippo tumor suppressor pathway regulates organ size and tissue homoeostasis in response to diverse signaling inputs. The core of the pathway consists of a short kinase cascade: MST1 and MST2 phosphorylate and activate LATS1 and LATS2, which in turn phosphorylate and inactivate key transcriptional coactivators, YAP1 and TAZ (gene WWTR1). The MOB1 adapter protein regulates both phosphorylation reactions firstly by concurrently binding to the upstream MST and downstream LATS kinases to enable the trans phosphorylation reaction, and secondly by allosterically activating the catalytic function of LATS1 and LATS2 to directly stimulate phosphorylation of YAP and TAZ. Studies of yeast Mob1 and human MOB1 revealed that the ability to recognize phosphopeptide sequences in their interactors, Nud1 and MST2 respectively, was critical to their roles in regulating the Mitotic Exit Network in yeast and the Hippo pathway in metazoans. However, the underlying rules of phosphopeptide recognition by human MOB1, the implications of binding specificity for Hippo pathway signaling, and the generality of phosphopeptide binding function to other human MOB family members remained elusive.Employing proteomics, peptide arrays and biochemical analyses, we systematically examine the phosphopeptide binding specificity of MOB1 and find it to be highly complementary to the substrate phosphorylation specificity of MST1 and MST2. We demonstrate that autophosphorylation of MST1 and MST2 on several threonine residues provides multiple MOB1 binding sites with varying binding affinities which in turn contribute to a redundancy of MST1-MOB1 protein interactions in cells. The crystal structures of MOB1A in complex with two favored phosphopeptide sites in MST1 allow for a full description of the MOB1A phosphopeptide-binding consensus. Lastly, we show that the phosphopeptide binding properties of MOB1A are conserved in all but one of the seven MOB family members in humans, thus providing a starting point for uncovering their elusive cellular functions.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Protein Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/chemistry , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Phosphopeptides/metabolism , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Recombinant Proteins/metabolism , Serine-Threonine Kinase 3 , Signal Transduction
9.
Bioorg Med Chem ; 26(11): 2937-2957, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29776834

ABSTRACT

Ligands for the bromodomain and extra-terminal domain (BET) family of bromodomains have shown promise as useful therapeutic agents for treating a range of cancers and inflammation. Here we report that our previously developed 3,5-dimethylisoxazole-based BET bromodomain ligand (OXFBD02) inhibits interactions of BRD4(1) with the RelA subunit of NF-κB, in addition to histone H4. This ligand shows a promising profile in a screen of the NCI-60 panel but was rapidly metabolised (t½â€¯= 39.8 min). Structure-guided optimisation of compound properties led to the development of the 3-pyridyl-derived OXFBD04. Molecular dynamics simulations assisted our understanding of the role played by an internal hydrogen bond in altering the affinity of this series of molecules for BRD4(1). OXFBD04 shows improved BRD4(1) affinity (IC50 = 166 nM), optimised physicochemical properties (LE = 0.43; LLE = 5.74; SFI = 5.96), and greater metabolic stability (t½â€¯= 388 min).


Subject(s)
Nuclear Proteins/chemistry , Transcription Factors/chemistry , Biological Assay , Blotting, Western , Cell Cycle Proteins , Crystallography, X-Ray , Drug Stability , Heterocyclic Compounds, 4 or More Rings/chemistry , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Inhibitory Concentration 50 , Ligands , Luciferases/chemistry , MCF-7 Cells , Molecular Dynamics Simulation , Molecular Structure , Structure-Activity Relationship
10.
Nat Chem Biol ; 10(4): 305-12, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24584101

ABSTRACT

Concomitant inhibition of multiple cancer-driving kinases is an established strategy to improve the durability of clinical responses to targeted therapies. The difficulty of discovering kinase inhibitors with an appropriate multitarget profile has, however, necessitated the application of combination therapies, which can pose major clinical development challenges. Epigenetic reader domains of the bromodomain family have recently emerged as new targets for cancer therapy. Here we report that several clinical kinase inhibitors also inhibit bromodomains with therapeutically relevant potencies and are best classified as dual kinase-bromodomain inhibitors. Nanomolar activity on BRD4 by BI-2536 and TG-101348, which are clinical PLK1 and JAK2-FLT3 kinase inhibitors, respectively, is particularly noteworthy as these combinations of activities on independent oncogenic pathways exemplify a new strategy for rational single-agent polypharmacological targeting. Furthermore, structure-activity relationships and co-crystal structures identify design features that enable a general platform for the rational design of dual kinase-bromodomain inhibitors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/chemical synthesis , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Design , Polypharmacology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Blotting, Western , Calorimetry , Cell Line, Tumor , Crystallization , Drug Interactions , Drug Screening Assays, Antitumor , Epigenesis, Genetic , High-Throughput Screening Assays , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Proto-Oncogene Proteins c-myc/metabolism , Pteridines/pharmacology , Pyrrolidines/pharmacology , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Structure-Activity Relationship , Sulfonamides/pharmacology
11.
Nature ; 468(7327): 1067-73, 2010 Dec 23.
Article in English | MEDLINE | ID: mdl-20871596

ABSTRACT

Epigenetic proteins are intently pursued targets in ligand discovery. So far, successful efforts have been limited to chromatin modifying enzymes, or so-called epigenetic 'writers' and 'erasers'. Potent inhibitors of histone binding modules have not yet been described. Here we report a cell-permeable small molecule (JQ1) that binds competitively to acetyl-lysine recognition motifs, or bromodomains. High potency and specificity towards a subset of human bromodomains is explained by co-crystal structures with bromodomain and extra-terminal (BET) family member BRD4, revealing excellent shape complementarity with the acetyl-lysine binding cavity. Recurrent translocation of BRD4 is observed in a genetically-defined, incurable subtype of human squamous carcinoma. Competitive binding by JQ1 displaces the BRD4 fusion oncoprotein from chromatin, prompting squamous differentiation and specific antiproliferative effects in BRD4-dependent cell lines and patient-derived xenograft models. These data establish proof-of-concept for targeting protein-protein interactions of epigenetic 'readers', and provide a versatile chemical scaffold for the development of chemical probes more broadly throughout the bromodomain family.


Subject(s)
Azirines/pharmacology , Dihydropyridines/pharmacology , Models, Molecular , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Azirines/chemical synthesis , Azirines/chemistry , Binding Sites , Carcinoma, Squamous Cell/physiopathology , Cell Cycle Proteins , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chromatin/metabolism , Dihydropyridines/chemical synthesis , Dihydropyridines/chemistry , Female , Humans , Mice , Mice, Nude , Molecular Sequence Data , Protein Binding/drug effects , Protein Structure, Tertiary , Recombinant Proteins/metabolism , Sequence Alignment , Skin Neoplasms/physiopathology , Stereoisomerism
12.
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
13.
J Am Chem Soc ; 136(26): 9308-19, 2014 Jul 02.
Article in English | MEDLINE | ID: mdl-24946055

ABSTRACT

Small-molecule inhibitors that target bromodomains outside of the bromodomain and extra-terminal (BET) sub-family are lacking. Here, we describe highly potent and selective ligands for the bromodomain module of the human lysine acetyl transferase CBP/p300, developed from a series of 5-isoxazolyl-benzimidazoles. Our starting point was a fragment hit, which was optimized into a more potent and selective lead using parallel synthesis employing Suzuki couplings, benzimidazole-forming reactions, and reductive aminations. The selectivity of the lead compound against other bromodomain family members was investigated using a thermal stability assay, which revealed some inhibition of the structurally related BET family members. To address the BET selectivity issue, X-ray crystal structures of the lead compound bound to the CREB binding protein (CBP) and the first bromodomain of BRD4 (BRD4(1)) were used to guide the design of more selective compounds. The crystal structures obtained revealed two distinct binding modes. By varying the aryl substitution pattern and developing conformationally constrained analogues, selectivity for CBP over BRD4(1) was increased. The optimized compound is highly potent (Kd = 21 nM) and selective, displaying 40-fold selectivity over BRD4(1). Cellular activity was demonstrated using fluorescence recovery after photo-bleaching (FRAP) and a p53 reporter assay. The optimized compounds are cell-active and have nanomolar affinity for CBP/p300; therefore, they should be useful in studies investigating the biological roles of CBP and p300 and to validate the CBP and p300 bromodomains as therapeutic targets.


Subject(s)
CREB-Binding Protein/chemistry , E1A-Associated p300 Protein/chemistry , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Binding Sites , CREB-Binding Protein/genetics , CREB-Binding Protein/metabolism , Chemistry Techniques, Synthetic , Crystallography, X-Ray , Drug Discovery , Drug Evaluation, Preclinical/methods , E1A-Associated p300 Protein/metabolism , Fluorescence Recovery After Photobleaching , Genes, p53 , HeLa Cells/drug effects , Humans , Indoles/chemistry , Isoxazoles/chemistry , Ligands , Microsomes, Liver/drug effects , Models, Molecular , Molecular Structure , Protein Structure, Tertiary , Small Molecule Libraries/metabolism , Structure-Activity Relationship
14.
Angew Chem Int Ed Engl ; 53(24): 6126-30, 2014 Jun 10.
Article in English | MEDLINE | ID: mdl-24821300

ABSTRACT

The benzoxazinone and dihydroquinoxalinone fragments were employed as novel acetyl lysine mimics in the development of CREBBP bromodomain ligands. While the benzoxazinone series showed low affinity for the CREBBP bromodomain, expansion of the dihydroquinoxalinone series resulted in the first potent inhibitors of a bromodomain outside the BET family. Structural and computational studies reveal that an internal hydrogen bond stabilizes the protein-bound conformation of the dihydroquinoxalinone series. The side chain of this series binds in an induced-fit pocket forming a cation-π interaction with R1173 of CREBBP. The most potent compound inhibits binding of CREBBP to chromatin in U2OS cells.


Subject(s)
CREB-Binding Protein/genetics , Cations/chemistry , Epigenomics/methods , Ligands , Models, Molecular , Protein Binding
15.
J Mol Biol ; 436(7): 168212, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-37481158

ABSTRACT

The human methyltransferase MLL4 plays a critical role in embryogenesis and development, and aberrant activity of MLL4 is linked to neurodegenerative and developmental disorders and cancer. MLL4 contains the catalytic SET domain that catalyzes mono methylation of lysine 4 of histone H3 (H3K4me1) and seven plant homeodomain (PHD) fingers, six of which have not been structurally and functionally characterized. Here, we demonstrate that the triple PHD finger cassette of MLL4, harboring its fourth, fifth and sixth PHD fingers (MLL4PHD456) forms an integrated module, maintains the binding selectivity of the PHD6 finger toward acetylated lysine 16 of histone H4 (H4K16ac), and is capable of binding to DNA. Our findings highlight functional correlation between H4K16ac and H3K4me1, two major histone modifications that are recognized and written, respectively, by MLL4.


Subject(s)
Histone-Lysine N-Methyltransferase , Histones , PHD Zinc Fingers , Humans , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Lysine/metabolism , Protein Binding
16.
bioRxiv ; 2023 Apr 21.
Article in English | MEDLINE | ID: mdl-37131595

ABSTRACT

Since genome instability can drive cancer initiation and progression, cells have evolved highly effective and ubiquitous DNA Damage Response (DDR) programs. However, some cells, in skin for example, are normally exposed to high levels of DNA damaging agents. Whether such high-risk cells possess lineage-specific mechanisms that tailor DNA repair to the tissue remains largely unknown. Here we show, using melanoma as a model, that the microphthalmia-associated transcription factor MITF, a lineage addition oncogene that coordinates many aspects of melanocyte and melanoma biology, plays a non-transcriptional role in shaping the DDR. On exposure to DNA damaging agents, MITF is phosphorylated by ATM/DNA-PKcs, and unexpectedly its interactome is dramatically remodelled; most transcription (co)factors dissociate, and instead MITF interacts with the MRE11-RAD50-NBS1 (MRN) complex. Consequently, cells with high MITF levels accumulate stalled replication forks, and display defects in homologous recombination-mediated repair associated with impaired MRN recruitment to DNA damage. In agreement, high MITF levels are associated with increased SNV burden in melanoma. Significantly, the SUMOylation-defective MITF-E318K melanoma predisposition mutation recapitulates the effects of ATM/DNA-PKcs-phosphorylated MITF. Our data suggest that a non-transcriptional function of a lineage-restricted transcription factor contributes to a tissue-specialised modulation of the DDR that can impact cancer initiation.

17.
Nat Commun ; 14(1): 6051, 2023 09 28.
Article in English | MEDLINE | ID: mdl-37770430

ABSTRACT

The ability of transcription factors to discriminate between different classes of binding sites associated with specific biological functions underpins effective gene regulation in development and homeostasis. How this is achieved is poorly understood. The microphthalmia-associated transcription factor MITF is a lineage-survival oncogene that plays a crucial role in melanocyte development and melanoma. MITF suppresses invasion, reprograms metabolism and promotes both proliferation and differentiation. How MITF distinguishes between differentiation and proliferation-associated targets is unknown. Here we show that compared to many transcription factors MITF exhibits a very long residence time which is reduced by p300/CBP-mediated MITF acetylation at K206. While K206 acetylation also decreases genome-wide MITF DNA-binding affinity, it preferentially directs DNA binding away from differentiation-associated CATGTG motifs toward CACGTG elements. The results reveal an acetylation-mediated switch that suppresses differentiation and provides a mechanistic explanation of why a human K206Q MITF mutation is associated with Waardenburg syndrome.


Subject(s)
Melanoma , Microphthalmia-Associated Transcription Factor , Humans , Cell Line, Tumor , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Acetylation , Melanoma/genetics , Melanoma/metabolism , Melanocytes/metabolism
18.
Bioorg Med Chem ; 20(6): 1878-86, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22137933

ABSTRACT

Benzodiazepines are psychoactive drugs with anxiolytic, sedative, skeletal muscle relaxant and amnestic properties. Recently triazolo-benzodiazepines have been also described as potent and highly selective protein interaction inhibitors of bromodomain and extra-terminal (BET) proteins, a family of transcriptional co-regulators that play a key role in cancer cell survival and proliferation, but the requirements for high affinity interaction of this compound class with bromodomains has not been described. Here we provide insight into the structure-activity relationship (SAR) and selectivity of this versatile scaffold. In addition, using high resolution crystal structures we compared the binding mode of a series of benzodiazepine (BzD) and related triazolo-benzotriazepines (BzT) derivatives including clinically approved drugs such as alprazolam and midazolam. Our analysis revealed the importance of the 1-methyl triazolo ring system for BET binding and suggests modifications for the development of further high affinity bromodomain inhibitors.


Subject(s)
Benzazepines/chemistry , Benzazepines/pharmacology , Nuclear Proteins/metabolism , Protein Interaction Maps/drug effects , Transcription Factors/metabolism , Alprazolam/chemistry , Alprazolam/pharmacology , Benzodiazepines/chemistry , Benzodiazepines/pharmacology , Cell Cycle Proteins , GABA Modulators/chemistry , GABA Modulators/pharmacology , Humans , Midazolam/chemistry , Midazolam/pharmacology , Models, Molecular , Nuclear Proteins/chemistry , Protein Structure, Tertiary/drug effects , Transcription Factors/chemistry
19.
ChemMedChem ; 17(20): e202200343, 2022 10 19.
Article in English | MEDLINE | ID: mdl-36040095

ABSTRACT

The bromodomain and extra-terminal (BET) family of proteins includes BRD2, BRD3, BRD4, and the testis-specific protein, BRDT, each containing two N-terminal tandem bromodomain (BRD) modules. Potent and selective inhibitors targeting the two bromodomains are required to elucidate their biological role(s), with potential clinical applications. In this study, we designed and synthesized a series of benzimidazole-6-sulfonamides starting from the azobenzene compounds MS436 (7 a) and MS611 (7 b) that exhibited preference for the first (BD1) over the second (BD2) BRD of BET family members. The most-promising compound (9 a) showed good binding potency and improved metabolic stability and selectivity towards BD1 with respect to the parent compounds.


Subject(s)
Nuclear Proteins , Sulfonamides , Male , Humans , Sulfonamides/pharmacology , Benzo(a)pyrene , Transcription Factors/metabolism , Imidazoles/pharmacology , Benzimidazoles/pharmacology , Cell Cycle Proteins/metabolism
20.
J Inherit Metab Dis ; 34(3): 671-6, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21445611

ABSTRACT

Fumarate hydratase catalyzes the stereospecific hydration across the olefinic double bond in fumarate leading to L-malate. The enzyme is expressed in mitochondrial and cytosolic compartments, and participates in the Krebs cycle in mitochondria, as well as in regulation of cytosolic fumarate levels. Fumarate hydratase deficiency is an autosomal recessive trait presenting as metabolic disorder with severe encephalopathy, seizures and poor neurological outcome. Heterozygous mutations are associated with a predisposition to cutaneous and uterine leiomyomas and to renal cancer. The crystal structure of human fumarate hydratase shows that mutations can be grouped into two distinct classes either affecting structural integrity of the core enzyme architecture, or are localized around the enzyme active site. An interactive version of this manuscript (which may contain additional mutations appended after acceptance of this manuscript) may be found on the SSIEM website at: http://www.ssiem.org/resources/structures/FH .


Subject(s)
Fumarate Hydratase/chemistry , Fumarate Hydratase/genetics , Metabolism, Inborn Errors/etiology , Catalytic Domain/genetics , Crystallography, X-Ray , Fumarate Hydratase/deficiency , Humans , Metabolism, Inborn Errors/genetics , Mitochondrial Diseases/etiology , Mitochondrial Diseases/genetics , Models, Molecular , Mutant Proteins/chemistry , Mutation/physiology , Protein Conformation , Protein Folding , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL