Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Angew Chem Int Ed Engl ; 63(14): e202316496, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38348945

ABSTRACT

Brachyury is an oncogenic transcription factor whose overexpression drives chordoma growth. The downmodulation of brachyury in chordoma cells has demonstrated therapeutic potential, however, as a transcription factor it is classically deemed "undruggable". Given that direct pharmacological intervention against brachyury has proven difficult, attempts at intervention have instead targeted upstream kinases. Recently, afatinib, an FDA-approved kinase inhibitor, has been shown to modulate brachyury levels in multiple chordoma cell lines. Herein, we use afatinib as a lead to undertake a structure-based drug design approach, aided by mass-spectrometry and X-ray crystallography, to develop DHC-156, a small molecule that more selectively binds brachyury and downmodulates it as potently as afatinib. We eliminated kinase-inhibition from this novel scaffold while demonstrating that DHC-156 induces the post-translational downmodulation of brachyury that results in an irreversible impairment of chordoma tumor cell growth. In doing so, we demonstrate the feasibility of direct brachyury modulation, which may further be developed into more potent tool compounds and therapies.


Subject(s)
Chordoma , Fetal Proteins , Transcription Factors , Humans , Transcription Factors/metabolism , Chordoma/drug therapy , Chordoma/metabolism , Chordoma/pathology , Afatinib , T-Box Domain Proteins/metabolism
2.
Nat Commun ; 13(1): 6989, 2022 11 16.
Article in English | MEDLINE | ID: mdl-36384931

ABSTRACT

Efficient determination of protein ligandability, or the propensity to bind small-molecules, would greatly facilitate drug development for novel targets. Ligandability is currently assessed using computational methods that typically consider the static structural properties of putative binding sites or by experimental fragment screening. Here, we evaluate ligandability of conserved BTB domains from the cancer-relevant proteins LRF, KAISO, and MIZ1. Using fragment screening, we discover that MIZ1 binds multiple ligands. However, no ligands are uncovered for the structurally related KAISO or LRF. To understand the principles governing ligand-binding by BTB domains, we perform comprehensive NMR-based dynamics studies and find that only the MIZ1 BTB domain exhibits backbone µs-ms time scale motions. Interestingly, residues with elevated dynamics correspond to the binding site of fragment hits and recently defined HUWE1 interaction site. Our data argue that examining protein dynamics using NMR can contribute to identification of cryptic binding sites, and may support prediction of the ligandability of novel challenging targets.


Subject(s)
BTB-POZ Domain , Binding Sites , Proteins/metabolism , Ligands , Protein Binding
3.
Cell Chem Biol ; 28(12): 1716-1727.e6, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34289376

ABSTRACT

GAS41 is an emerging oncogene overexpressed and implicated in multiple cancers, including non-small cell lung cancer (NSCLC). GAS41 is a dimeric protein that contains the YEATS domain, which is involved in the recognition of lysine-acylated histones. Here, we report the development of GAS41 YEATS inhibitors by employing a fragment-based screening approach. These inhibitors bind to GAS41 YEATS domain in a channel constituting a recognition site for acylated lysine on histone proteins. To enhance inhibitory activity, we developed a dimeric analog with nanomolar activity that blocks interactions of GAS41 with acetylated histone H3. Our lead compound engages GAS41 in cells, blocks proliferation of NSCLC cells, and modulates expression of GAS41-dependent genes, validating on-target mechanism of action. This study demonstrates that disruption of GAS41 protein-protein interactions may represent an attractive approach to target lung cancer cells. This work exemplifies the use of bivalent inhibitors as a general strategy to block challenging protein-protein interactions.


Subject(s)
Amides/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Thiophenes/pharmacology , Transcription Factors/antagonists & inhibitors , Amides/chemistry , Antineoplastic Agents/chemistry , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation/drug effects , Cells, Cultured , Drug Screening Assays, Antitumor , Female , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Molecular Structure , Protein Interaction Domains and Motifs/drug effects , Thiophenes/chemistry , Transcription Factors/metabolism
4.
Proc Natl Acad Sci U S A ; 117(44): 27346-27353, 2020 11 03.
Article in English | MEDLINE | ID: mdl-33077600

ABSTRACT

A key functional event in eukaryotic gene activation is the formation of dynamic protein-protein interaction networks between transcriptional activators and transcriptional coactivators. Seemingly incongruent with the tight regulation of transcription, many biochemical and biophysical studies suggest that activators use nonspecific hydrophobic and/or electrostatic interactions to bind to coactivators, with few if any specific contacts. Here a mechanistic dissection of a set of representative dynamic activator•coactivator complexes, comprised of the ETV/PEA3 family of activators and the coactivator Med25, reveals a different molecular recognition model. The data demonstrate that small sequence variations within an activator family significantly redistribute the conformational ensemble of the complex while not affecting overall affinity, and distal residues within the activator-not often considered as contributing to binding-play a key role in mediating conformational redistribution. The ETV/PEA3•Med25 ensembles are directed by specific contacts between the disordered activator and the Med25 interface, which is facilitated by structural shifts of the coactivator binding surface. Taken together, these data highlight the critical role coactivator plasticity plays in recognition of disordered activators and indicate that molecular recognition models of disordered proteins must consider the ability of the binding partners to mediate specificity.


Subject(s)
Transcription Factors/metabolism , Transcriptional Activation/genetics , Amino Acid Sequence/genetics , Humans , Mediator Complex/genetics , Mediator Complex/metabolism , Models, Molecular , Protein Binding/genetics , Protein Interaction Domains and Motifs/genetics , Transcriptional Activation/physiology
5.
Eur J Med Chem ; 207: 112748, 2020 Dec 01.
Article in English | MEDLINE | ID: mdl-32882610

ABSTRACT

The interaction between menin and mixed lineage leukemia (MLL) was identified as an interesting target for treating some cancers including acute leukemia. On the basis of the known crystal structure of the MBM1-menin complex (MBM - menin binding motif), several cyclic peptides were designed. Elaboration of the effective cyclization strategy using a metathesis reaction allowed for a successfully large number of derivatives to be obtained. Subsequent optimization of the loop size, as well as N-terminal, central and C-terminal parts of the studied peptides resulted in structures exhibiting low nanomolar activities. A crystal structure of an inhibitor-menin complex revealed a compact conformation of the ligand molecule, which is stabilized not only by the introduction of a covalent linker but also three intramolecular hydrogen bonds. The inhibitor adopts a figure eight-like conformation, which perfectly fits the cleft of menin. We demonstrated that the development of compact, miniprotein-like structures is a highly effective approach for inhibition of protein-protein interactions.


Subject(s)
Myeloid-Lymphoid Leukemia Protein/metabolism , Peptides/chemistry , Peptides/pharmacology , Proto-Oncogene Proteins/metabolism , Amino Acid Motifs , Humans , Ligands , Models, Molecular , Myeloid-Lymphoid Leukemia Protein/chemistry , Protein Binding/drug effects , Proto-Oncogene Proteins/chemistry
6.
Future Med Chem ; 12(14): 1305-1326, 2020 07.
Article in English | MEDLINE | ID: mdl-32551894

ABSTRACT

Epigenetic protein-protein interactions (PPIs) play essential roles in regulating gene expression, and their dysregulations have been implicated in many diseases. These PPIs are comprised of reader domains recognizing post-translational modifications on histone proteins, and of scaffolding proteins that maintain integrities of epigenetic complexes. Targeting PPIs have become focuses for development of small-molecule inhibitors and anticancer therapeutics. Here we summarize efforts to develop small-molecule inhibitors targeting common epigenetic PPI domains. Potent small molecules have been reported for many domains, yet small domains that recognize methylated lysine side chains on histones are challenging in inhibitor development. We posit that the development of potent inhibitors for difficult-to-prosecute epigenetic PPIs may be achieved by interdisciplinary approaches and extensive explorations of chemical space.


Subject(s)
Epigenesis, Genetic/drug effects , Proteins/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Humans , Protein Binding/drug effects , Protein Processing, Post-Translational/drug effects , Proteins/genetics , Small Molecule Libraries/chemistry
7.
J Clin Invest ; 130(2): 981-997, 2020 02 03.
Article in English | MEDLINE | ID: mdl-31855575

ABSTRACT

The protein-protein interaction between menin and mixed lineage leukemia 1 (MLL1) plays a critical role in acute leukemias with translocations of the MLL1 gene or with mutations in the nucleophosmin 1 (NPM1) gene. As a step toward clinical translation of menin-MLL1 inhibitors, we report development of MI-3454, a highly potent and orally bioavailable inhibitor of the menin-MLL1 interaction. MI-3454 profoundly inhibited proliferation and induced differentiation in acute leukemia cells and primary patient samples with MLL1 translocations or NPM1 mutations. When applied as a single agent, MI-3454 induced complete remission or regression of leukemia in mouse models of MLL1-rearranged or NPM1-mutated leukemia, including patient-derived xenograft models, through downregulation of key genes involved in leukemogenesis. We also identified MEIS1 as a potential pharmacodynamic biomarker of treatment response with MI-3454 in leukemia, and demonstrated that this compound is well tolerated and did not impair normal hematopoiesis in mice. Overall, this study demonstrates, for the first time to our knowledge, profound activity of the menin-MLL1 inhibitor as a single agent in clinically relevant PDX models of leukemia. These data provide a strong rationale for clinical translation of MI-3454 or its analogs for leukemia patients with MLL1 rearrangements or NPM1 mutations.


Subject(s)
Antineoplastic Agents/pharmacology , Histone-Lysine N-Methyltransferase , Leukemia , Mutation , Myeloid-Lymphoid Leukemia Protein , Neoplasms, Experimental , Nuclear Proteins , Proto-Oncogene Proteins , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , K562 Cells , Leukemia/drug therapy , Leukemia/genetics , Leukemia/metabolism , Leukemia/pathology , Myeloid Ecotropic Viral Integration Site 1 Protein/genetics , Myeloid Ecotropic Viral Integration Site 1 Protein/metabolism , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleophosmin , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Remission Induction , U937 Cells
8.
Proc Natl Acad Sci U S A ; 115(36): 8960-8965, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30127017

ABSTRACT

Transcriptional coactivators are a molecular recognition marvel because a single domain within these proteins, the activator binding domain or ABD, interacts with multiple compositionally diverse transcriptional activators. Also remarkable is the structural diversity among ABDs, which range from conformationally dynamic helical motifs to those with a stable core such as a ß-barrel. A significant objective is to define conserved properties of ABDs that allow them to interact with disparate activator sequences. The ABD of the coactivator Med25 (activator interaction domain or AcID) is unique in that it contains secondary structural elements that are on both ends of the spectrum: helices and loops that display significant conformational mobility and a seven-stranded ß-barrel core that is structurally rigid. Using biophysical approaches, we build a mechanistic model of how AcID forms binary and ternary complexes with three distinct activators; despite its static core, Med25 forms short-lived, conformationally mobile, and structurally distinct complexes with each of the cognate partners. Further, ternary complex formation is facilitated by allosteric communication between binding surfaces on opposing faces of the ß-barrel. The model emerging suggests that the conformational shifts and cooperative binding is mediated by a flexible substructure comprised of two dynamic helices and flanking loops, indicating a conserved mechanistic model of activator engagement across ABDs. Targeting a region of this substructure with a small-molecule covalent cochaperone modulates ternary complex formation. Our data support a general strategy for the identification of allosteric small-molecule modulators of ABDs, which are key targets for mechanistic studies as well as therapeutic applications.


Subject(s)
Mediator Complex/antagonists & inhibitors , Mediator Complex/chemistry , Peptides/chemistry , Allosteric Regulation/physiology , Humans , Mediator Complex/metabolism , Protein Domains , Protein Structure, Quaternary , Protein Structure, Secondary
9.
ACS Chem Biol ; 13(9): 2739-2746, 2018 09 21.
Article in English | MEDLINE | ID: mdl-30071723

ABSTRACT

GAS41 is a chromatin-associated protein that belongs to the YEATS family and is involved in the recognition of acetyl-lysine in histone proteins. A unique feature of GAS41 is the presence of a C-terminal coiled-coil domain, which is responsible for protein dimerization. Here, we characterized the specificity of the GAS41 YEATS domain and found that it preferentially binds to acetylated H3K18 and H3K27 peptides. Interestingly, we found that full-length, dimeric GAS41 binds to diacetylated H3 peptides with an enhanced affinity when compared to those for monoacetylated peptides, through a bivalent binding mode. We determined the crystal structure of the GAS41 YEATS domain with H3K23acK27ac to visualize the molecular basis of diacetylated histone binding. Our results suggest a unique binding mode in which full-length GAS41 is a reader of diacetylated histones.


Subject(s)
Histones/metabolism , Transcription Factors/metabolism , Acetylation , Binding Sites , HEK293 Cells , Histones/chemistry , Humans , Models, Molecular , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , Transcription Factors/chemistry
10.
J Med Chem ; 61(17): 7573-7588, 2018 09 13.
Article in English | MEDLINE | ID: mdl-29969259

ABSTRACT

Protein-protein interactions (PPI) between the transcriptional repressor B-cell lymphoma 6 (BCL6) BTB domain (BCL6BTB) and its corepressors have emerged as a promising target for anticancer therapeutics. However, identification of potent, drug-like inhibitors of BCL6BTB has remained challenging. Using NMR-based screening of a library of fragment-like small molecules, we have identified a thiourea compound (7CC5) that binds to BCL6BTB. From this hit, the application of computer-aided drug design (CADD), medicinal chemistry, NMR spectroscopy, and X-ray crystallography has yielded an inhibitor, 15f, that demonstrated over 100-fold improved potency for BCL6BTB. This gain in potency was achieved by a unique binding mode that mimics the binding mode of the corepressor SMRT in the aromatic and the HDCH sites. The structure-activity relationship based on these new inhibitors will have a significant impact on the rational design of novel BCL6 inhibitors, facilitating the identification of therapeutics for the treatment of BCL6-dependent tumors.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Proto-Oncogene Proteins c-bcl-6/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-6/chemistry , Thiourea/chemistry , BTB-POZ Domain , Cell Line, Tumor , Computer-Aided Design , Crystallography, X-Ray , Drug Design , Humans , Hydrogen Bonding , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/pathology , Magnetic Resonance Spectroscopy , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Structure-Activity Relationship
11.
J Med Chem ; 61(11): 4832-4850, 2018 06 14.
Article in English | MEDLINE | ID: mdl-29738674

ABSTRACT

The protein-protein interaction between menin and mixed-lineage leukemia 1 (MLL1) plays an important role in development of acute leukemia with translocations of the MLL1 gene and in solid tumors. Here, we report the development of a new generation of menin-MLL1 inhibitors identified by structure-based optimization of the thienopyrimidine class of compounds. This work resulted in compound 28 (MI-1481), which showed very potent inhibition of the menin-MLL1 interaction (IC50 = 3.6 nM), representing the most potent reversible menin-MLL1 inhibitor reported to date. The crystal structure of the menin-28 complex revealed a hydrogen bond with Glu366 and hydrophobic interactions, which contributed to strong inhibitory activity of 28. Compound 28 also demonstrates pronounced activity in MLL leukemia cells and in vivo in MLL leukemia models. Thus, 28 is a valuable menin-MLL1 inhibitor that can be used for potential therapeutic applications and in further studies regarding the role of menin in cancer.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Proto-Oncogene Proteins/metabolism , Pyrimidines/pharmacology , Animals , Cell Line, Tumor , Drug Design , Female , Humans , Mice , Mice, Inbred C57BL , Models, Molecular , Protein Binding , Protein Conformation , Pyrimidines/chemistry , Pyrimidines/pharmacokinetics
12.
Proc Natl Acad Sci U S A ; 113(31): 8681-6, 2016 08 02.
Article in English | MEDLINE | ID: mdl-27432965

ABSTRACT

The assembly of individual protein subunits into large-scale symmetrical structures is widespread in nature and confers new biological properties. Engineered protein assemblies have potential applications in nanotechnology and medicine; however, a major challenge in engineering assemblies de novo has been to design interactions between the protein subunits so that they specifically assemble into the desired structure. Here we demonstrate a simple, generalizable approach to assemble proteins into cage-like structures that uses short de novo designed coiled-coil domains to mediate assembly. We assembled eight copies of a C3-symmetric trimeric esterase into a well-defined octahedral protein cage by appending a C4-symmetric coiled-coil domain to the protein through a short, flexible linker sequence, with the approximate length of the linker sequence determined by computational modeling. The structure of the cage was verified using a combination of analytical ultracentrifugation, native electrospray mass spectrometry, and negative stain and cryoelectron microscopy. For the protein cage to assemble correctly, it was necessary to optimize the length of the linker sequence. This observation suggests that flexibility between the two protein domains is important to allow the protein subunits sufficient freedom to assemble into the geometry specified by the combination of C4 and C3 symmetry elements. Because this approach is inherently modular and places minimal requirements on the structural features of the protein building blocks, it could be extended to assemble a wide variety of proteins into structures with different symmetries.


Subject(s)
Protein Folding , Protein Multimerization , Protein Structure, Secondary , Proteins/chemistry , Amino Acid Sequence , Cryoelectron Microscopy , Mass Spectrometry/methods , Microscopy, Electron, Transmission , Models, Molecular , Octamer Transcription Factor-2/chemistry , Octamer Transcription Factor-2/ultrastructure , Octamer Transcription Factor-3/chemistry , Octamer Transcription Factor-3/ultrastructure , Proteins/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...