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










Publication year range
1.
J Med Chem ; 67(2): 1061-1078, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38198226

ABSTRACT

Hedgehog signaling is involved in embryonic development and cancer growth. Functional activity of secreted Hedgehog signaling proteins is dependent on N-terminal palmitoylation, making the palmitoyl transferase Hedgehog acyltransferase (HHAT), a potential drug target and a series of 4,5,6,7-tetrahydrothieno[3,2-c]pyridines have been identified as HHAT inhibitors. Based on structural data, we designed and synthesized 37 new analogues which we profiled alongside 13 previously reported analogues in enzymatic and cellular assays. Our results show that a central amide linkage, a secondary amine, and (R)-configuration at the 4-position of the core are three key factors for inhibitory potency. Several potent analogues with low- or sub-µM IC50 against purified HHAT also inhibit Sonic Hedgehog (SHH) palmitoylation in cells and suppress the SHH signaling pathway. This work identifies IMP-1575 as the most potent cell-active chemical probe for HHAT function, alongside an inactive control enantiomer, providing tool compounds for validation of HHAT as a target in cellular assays.


Subject(s)
Hedgehog Proteins , Hedgehog Proteins/metabolism , Pyridines/chemistry , Pyridines/pharmacology
2.
Cell Chem Biol ; 30(6): 573-590.e6, 2023 06 15.
Article in English | MEDLINE | ID: mdl-37130519

ABSTRACT

The natural product family of the fusicoccanes (FCs) has been shown to display anti-cancer activity, especially when combined with established therapeutic agents. FCs stabilize 14-3-3 protein-protein interactions (PPIs). Here, we tested combinations of a small library of FCs with interferon α (IFNα) on different cancer cell lines and report a proteomics approach to identify the specific 14-3-3 PPIs that are induced by IFNα and stabilized by FCs in OVCAR-3 cells. Among the identified 14-3-3 target proteins are THEMIS2, receptor interacting protein kinase 2 (RIPK2), EIF2AK2, and several members of the LDB1 complex. Biophysical and structural biology studies confirm these 14-3-3 PPIs as physical targets of FC stabilization, and transcriptome as well as pathway analyses suggest possible explanations for the observed synergistic effect of IFNα/FC treatment on cancer cells. This study elucidates the polypharmacological effects of FCs in cancer cells and identifies potential targets from the vast interactome of 14-3-3s for therapeutic intervention in oncology.


Subject(s)
Interferon-alpha , Ovarian Neoplasms , Humans , Female , Interferon-alpha/pharmacology , Apoptosis , Cell Line, Tumor , Cell Death
4.
J Med Chem ; 65(24): 16818-16828, 2022 12 22.
Article in English | MEDLINE | ID: mdl-36484727

ABSTRACT

The ubiquitously expressed glucocorticoid receptor (GR) is a nuclear receptor that controls a broad range of biological processes and is activated by steroidal glucocorticoids such as hydrocortisone or dexamethasone. Glucocorticoids are used to treat a wide variety of conditions, from inflammation to cancer but suffer from a range of side effects that motivate the search for safer GR modulators. GR is also regulated outside the steroid-binding site through protein-protein interactions (PPIs) with 14-3-3 adapter proteins. Manipulation of these PPIs will provide insights into noncanonical GR signaling as well as a new level of control over GR activity. We report the first molecular glues that selectively stabilize the 14-3-3/GR PPI using the related nuclear receptor estrogen receptor α (ERα) as a selectivity target to drive design. These 14-3-3/GR PPI stabilizers can be used to dissect noncanonical GR signaling and enable the development of novel atypical GR modulators.


Subject(s)
Glucocorticoids , Receptors, Glucocorticoid , Glucocorticoids/metabolism , Receptors, Glucocorticoid/metabolism , 14-3-3 Proteins/metabolism , Gene Expression Regulation , Binding Sites , Steroids , Dexamethasone
5.
Methods Mol Biol ; 2374: 13-26, 2022.
Article in English | MEDLINE | ID: mdl-34562239

ABSTRACT

Palmitoylation of the Hedgehog family of proteins is a critical step in the Hedgehog signaling pathway and is performed by the membrane-bound O-acyltransferase enzyme Hedgehog acyltransferase (HHAT). Measurement of HHAT activity has traditionally relied on radiolabeled fatty acid substrates, which imposes considerable constraints on throughput, cost, and safety, consequently hindering the efficient identification and development of small-molecule HHAT inhibitors. The Acylation-coupled Lipophilic Induction of Polarisation (Acyl-cLIP) assay was recently developed in our lab as a novel platform to evaluate lipidation of peptides in real time and high throughput. In this chapter, we describe the isolation of active HHAT from HEK293a cells and application of the Acyl-cLIP assay to characterize HHAT inhibitors. Our methodology uses standard chemical biology lab equipment and yields high-quality kinetic data from minimal sample volumes. The assay uses standard 384-well plates and is easily adapted to medium- or high-throughput screening formats.


Subject(s)
Acylation , Acyltransferases/metabolism , Hedgehog Proteins/metabolism , Lipoylation , Surgical Instruments
6.
Mol Cell ; 81(24): 5025-5038.e10, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34890564

ABSTRACT

The Sonic Hedgehog (SHH) morphogen pathway is fundamental for embryonic development and stem cell maintenance and is implicated in various cancers. A key step in signaling is transfer of a palmitate group to the SHH N terminus, catalyzed by the multi-pass transmembrane enzyme Hedgehog acyltransferase (HHAT). We present the high-resolution cryo-EM structure of HHAT bound to substrate analog palmityl-coenzyme A and a SHH-mimetic megabody, revealing a heme group bound to HHAT that is essential for HHAT function. A structure of HHAT bound to potent small-molecule inhibitor IMP-1575 revealed conformational changes in the active site that occlude substrate binding. Our multidisciplinary analysis provides a detailed view of the mechanism by which HHAT adapts the membrane environment to transfer an acyl chain across the endoplasmic reticulum membrane. This structure of a membrane-bound O-acyltransferase (MBOAT) superfamily member provides a blueprint for other protein-substrate MBOATs and a template for future drug discovery.


Subject(s)
Acyltransferases/antagonists & inhibitors , Acyltransferases/metabolism , Enzyme Inhibitors/pharmacology , Hedgehog Proteins/metabolism , Membrane Proteins/metabolism , Acylation , Acyltransferases/genetics , Acyltransferases/ultrastructure , Allosteric Regulation , Animals , COS Cells , Catalytic Domain , Chlorocebus aethiops , Cryoelectron Microscopy , HEK293 Cells , Heme/metabolism , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/ultrastructure , Molecular Dynamics Simulation , Palmitoyl Coenzyme A/metabolism , Protein Conformation , Signal Transduction , Structure-Activity Relationship
7.
ACS Med Chem Lett ; 12(6): 976-982, 2021 Jun 10.
Article in English | MEDLINE | ID: mdl-34136078

ABSTRACT

The systematic discovery of functional fragments binding to the composite interface of protein complexes is a first critical step for the development of orthosteric stabilizers of protein-protein interactions (PPIs). We have previously shown that disulfide trapping successfully yielded covalent stabilizers for the PPI of 14-3-3 with the estrogen receptor ERα. Here we provide an assessment of the composite PPI target pocket and the molecular characteristics of various fragments binding to a specific subpocket. Evaluating structure-activity relationships highlights the basic principles for PPI stabilization by these covalent fragments that engage a relatively large and exposed binding pocket at the protein/peptide interface with a "molecular glue" mode of action.

8.
Angew Chem Int Ed Engl ; 60(24): 13542-13547, 2021 06 07.
Article in English | MEDLINE | ID: mdl-33768725

ABSTRACT

The mammalian membrane-bound O-acyltransferase (MBOAT) superfamily is involved in biological processes including growth, development and appetite sensing. MBOATs are attractive drug targets in cancer and obesity; however, information on the binding site and molecular mechanisms underlying small-molecule inhibition is elusive. This study reports rational development of a photochemical probe to interrogate a novel small-molecule inhibitor binding site in the human MBOAT Hedgehog acyltransferase (HHAT). Structure-activity relationship investigation identified single enantiomer IMP-1575, the most potent HHAT inhibitor reported to-date, and guided design of photocrosslinking probes that maintained HHAT-inhibitory potency. Photocrosslinking and proteomic sequencing of HHAT delivered identification of the first small-molecule binding site in a mammalian MBOAT. Topology and homology data suggested a potential mechanism for HHAT inhibition which was confirmed by kinetic analysis. Our results provide an optimal HHAT tool inhibitor IMP-1575 (Ki =38 nM) and a strategy for mapping small molecule interaction sites in MBOATs.


Subject(s)
Acetyltransferases/antagonists & inhibitors , Affinity Labels/chemistry , Small Molecule Libraries/chemistry , Acetyltransferases/metabolism , Binding Sites , Humans , Kinetics , Light , Palmitoyl Coenzyme A/antagonists & inhibitors , Palmitoyl Coenzyme A/metabolism , Pyridines/chemistry , Pyridines/metabolism , Small Molecule Libraries/metabolism , Structure-Activity Relationship
9.
Angew Chem Weinheim Bergstr Ger ; 133(24): 13654-13659, 2021 Jun 07.
Article in English | MEDLINE | ID: mdl-38504937

ABSTRACT

The mammalian membrane-bound O-acyltransferase (MBOAT) superfamily is involved in biological processes including growth, development and appetite sensing. MBOATs are attractive drug targets in cancer and obesity; however, information on the binding site and molecular mechanisms underlying small-molecule inhibition is elusive. This study reports rational development of a photochemical probe to interrogate a novel small-molecule inhibitor binding site in the human MBOAT Hedgehog acyltransferase (HHAT). Structure-activity relationship investigation identified single enantiomer IMP-1575, the most potent HHAT inhibitor reported to-date, and guided design of photocrosslinking probes that maintained HHAT-inhibitory potency. Photocrosslinking and proteomic sequencing of HHAT delivered identification of the first small-molecule binding site in a mammalian MBOAT. Topology and homology data suggested a potential mechanism for HHAT inhibition which was confirmed by kinetic analysis. Our results provide an optimal HHAT tool inhibitor IMP-1575 (K i=38 nM) and a strategy for mapping small molecule interaction sites in MBOATs.

10.
ACS Med Chem Lett ; 11(5): 1041-1046, 2020 May 14.
Article in English | MEDLINE | ID: mdl-32435423

ABSTRACT

Protein-protein interactions (PPIs) play an important role in numerous biological processes such as cell-cycle regulation and multiple diseases. The family of 14-3-3 proteins is an attractive target as they serve as binding partner to various proteins and are therefore capable of regulating their biological activities. Discovering small-molecule modulators, in particular stabilizers, of such complexes via traditional screening approaches is a challenging task. Herein, we pioneered the first application of dynamic combinatorial chemistry (DCC) to a PPI target, to find modulators of 14-3-3 proteins. Evaluation of the amplified hits from the DCC experiments for their binding affinity via surface plasmon resonance (SPR), revealed that the low-micromolar (K D 15-16 µM) acylhydrazones are 14-3-3/synaptopodin PPI stabilizers. Thus, DCC appears to be ideally suited for the discovery of not only modulators but even the more elusive stabilizers of notoriously challenging PPIs.

11.
Chemistry ; 26(31): 7131-7139, 2020 Jun 02.
Article in English | MEDLINE | ID: mdl-32255539

ABSTRACT

Protein-protein interactions (PPIs) of 14-3-3 proteins are a model system for studying PPI stabilization. The complex natural product Fusicoccin A stabilizes many 14-3-3 PPIs but is not amenable for use in SAR studies, motivating the search for more drug-like chemical matter. However, drug-like 14-3-3 PPI stabilizers enabling such studies have remained elusive. An X-ray crystal structure of a PPI in complex with an extremely low potency stabilizer uncovered an unexpected non-protein interacting, ligand-chelated Mg2+ leading to the discovery of metal-ion-dependent 14-3-3 PPI stabilization potency. This originates from a novel chelation-controlled bioactive conformation stabilization effect. Metal chelation has been associated with pan-assay interference compounds (PAINS) and frequent hitter behavior, but chelation can evidently also lead to true potency gains and find use as a medicinal chemistry strategy to guide compound optimization. To demonstrate this, we exploited the effect to design the first potent, selective, and drug-like 14-3-3 PPI stabilizers.


Subject(s)
14-3-3 Proteins/chemistry , Biological Products/chemistry , Chelating Agents/chemistry , Metals/chemistry , Drug Discovery , Glycosides , Humans , Molecular Conformation , Protein Binding
12.
Cell Chem Biol ; 27(6): 657-667.e6, 2020 06 18.
Article in English | MEDLINE | ID: mdl-32220335

ABSTRACT

Targeting protein-protein interactions (PPIs) is a promising approach in the development of drugs for many indications. 14-3-3 proteins are a family of phosphoprotein-binding molecules with critical functions in dozens of cell signaling networks. 14-3-3s are abundant in the central nervous system, and the small molecule fusicoccin-A (FC-A), a tool compound that can be used to manipulate 14-3-3 PPIs, enhances neurite outgrowth in cultured neurons. New semisynthetic FC-A derivatives with improved binding affinity for 14-3-3 complexes have recently been developed. Here, we use a series of screens that identify these compounds as potent inducers of neurite outgrowth through a polypharmacological mechanism. Using proteomics and X-ray crystallography, we discover that these compounds extensively regulate the 14-3-3 interactome by stabilizing specific PPIs, while disrupting others. These results provide new insights into the development of drugs to target 14-3-3 PPIs, a potential therapeutic strategy for CNS diseases.


Subject(s)
14-3-3 Proteins/antagonists & inhibitors , Glycosides/pharmacology , Neurites/drug effects , Small Molecule Libraries/pharmacology , 14-3-3 Proteins/isolation & purification , 14-3-3 Proteins/metabolism , Animals , Cells, Cultured , Crystallography, X-Ray , Dose-Response Relationship, Drug , Female , Glycosides/chemistry , Male , Models, Molecular , Molecular Conformation , Neurites/metabolism , Neuronal Outgrowth/drug effects , Protein Binding/drug effects , Rats , Rats, Sprague-Dawley , Small Molecule Libraries/chemistry
13.
Chem Commun (Camb) ; 55(98): 14809-14812, 2019 Dec 05.
Article in English | MEDLINE | ID: mdl-31763628

ABSTRACT

Here we describe the synthesis of a series of α,ß-phosphopeptides, based on the phosphoepitope site on YAP1 (yes-associated protein 1), and the biochemical, biophysical and structural characterization of their binding to 14-3-3 proteins. The impact of systematic mono- and di-substitution of α → ß3 amino acid residues around the phosphoserine residue are discussed. Our results confirm the important role played by the +2 proline residue in the thermodynamics and structure of the phosphoepitope/14-3-3 interaction.


Subject(s)
14-3-3 Proteins/metabolism , Phosphopeptides/metabolism , 14-3-3 Proteins/chemistry , Adaptor Proteins, Signal Transducing/chemistry , Amino Acid Sequence , Amino Acids/chemistry , Amino Acids/metabolism , Binding Sites , Crystallography, X-Ray , Humans , Phosphopeptides/chemical synthesis , Phosphopeptides/chemistry , Protein Binding , Protein Structure, Secondary , Thermodynamics , Transcription Factors/chemistry , YAP-Signaling Proteins
14.
Chem Sci ; 10(10): 2869-2874, 2019 Mar 14.
Article in English | MEDLINE | ID: mdl-30996864

ABSTRACT

A cooperativity framework to describe and interpret small-molecule stabilization of protein-protein interactions (PPI) is presented. The stabilization of PPIs is a versatile and emerging therapeutic strategy to target specific combinations of protein partners within the protein interactome. Currently, the potency of PPI stabilizers is typically expressed by their apparent affinity or EC50. Here, we propose that the effect of a PPI stabilizer be best described involving the cooperativity factor, α, between the stabilizer and binding partners in addition to the intrinsic affinity, K D II, of the stabilizer for one of the apo-proteins. By way of illustration, we combine fluorescence polarization measurements with thermodynamic modeling to determine the α and K D II for the PPI stabilization of 14-3-3 and TASK3 by fusicoccin-A (FC-A) and validate our approach by studying other PPI-partners of 14-3-3 proteins. Finally, we characterize a library of different stabilizer compounds, and perform structure-activity relationship studies in which molecular changes could be attributed to either changes in cooperativity or intrinsic affinity. Such insights should aid in the development of more effective protein-protein stabilizer drugs.

15.
Angew Chem Int Ed Engl ; 57(41): 13470-13474, 2018 10 08.
Article in English | MEDLINE | ID: mdl-30025189

ABSTRACT

The natural product family of fusicoccanes are stabilizers of 14-3-3 mediated protein-protein interactions (PPIs), some of which possess antitumor activity. In this study, the first use of molecular dynamics (MD) to rationally design PPI stabilizers with increased potency is presented. Synthesis of a focused library, with subsequent characterization by fluorescence polarization, mutational studies, and X-ray crystallography confirmed the power of the MD-based design approach, revealing the potential for an additional hydrogen bond with the 14-3-3 protein to lead to significantly increased potency. Additionally, these compounds exert their action in a cellular environment with increased potency. The newly found polar interaction could provide an anchoring point for new small-molecule PPI stabilizers. These results facilitate the development of fusicoccanes towards drugs or tool compounds, as well as allowing the study of the fundamental principles behind PPI stabilization.


Subject(s)
14-3-3 Proteins/chemistry , Biological Products/chemistry , Diterpenes/chemistry , Cell Line, Tumor , Crystallography, X-Ray , Drug Design , Fluorescence Polarization , Humans , Hydrogen Bonding , Molecular Dynamics Simulation , Molecular Structure , Protein Binding
16.
ACS Chem Neurosci ; 9(11): 2639-2654, 2018 11 21.
Article in English | MEDLINE | ID: mdl-29722962

ABSTRACT

Current molecular hypotheses have not yet delivered marketable treatments for Alzheimer's disease (AD), arguably due to a lack of understanding of AD biology and an overreliance on conventional drug modalities. Protein-protein interactions (PPIs) are emerging drug targets, which show promise for the treatment of, e.g., cancer, but are still underexploited for treating neurodegenerative diseases. 14-3-3 binding to phosphorylated Tau is a promising PPI drug target based on its reported destabilizing effect on microtubules, leading to enhanced neurofibrillary tangle formation as a potential cause of AD-related neurodegeneration. Inhibition of 14-3-3/Tau may therefore be neuroprotective. Previously, we reported the structure-guided development of modified peptide inhibitors of 14-3-3/Tau. Here, we report further efforts to optimize the binding mode and activity of our modified Tau peptides through a combination of chemical synthesis, biochemical assays, and X-ray crystallography. Most notably, we were able to characterize two different high-affinity binding modes, both of which inhibited 14-3-3-binding to full-length PKA-phosphorylated Tau protein in vitro as measured by NMR spectroscopy. Our findings, besides producing useful tool inhibitor compounds for studying 14-3-3/Tau, have enhanced our understanding of the molecular parameters for inhibiting 14-3-3/Tau, which are important milestones toward the establishment of our 14-3-3 PPI hypothesis.


Subject(s)
14-3-3 Proteins/metabolism , Alzheimer Disease/metabolism , Neurofibrillary Tangles/metabolism , tau Proteins/metabolism , 14-3-3 Proteins/chemistry , Crystallography, X-Ray , Drug Discovery , Humans , Magnetic Resonance Spectroscopy , Microtubules/metabolism , Phosphorylation , Protein Binding , tau Proteins/chemistry
17.
Expert Opin Drug Discov ; 12(9): 925-940, 2017 09.
Article in English | MEDLINE | ID: mdl-28695752

ABSTRACT

INTRODUCTION: PPIs are involved in every disease and specific modulation of these PPIs with small molecules would significantly improve our prospects of developing therapeutic agents. Both industry and academia have engaged in the identification and use of PPI inhibitors. However in comparison, the opposite strategy of employing small-molecule stabilizers of PPIs is underrepresented in drug discovery. Areas covered: PPI stabilization has not been exploited in a systematic manner. Rather, this concept validated by a number of therapeutically used natural products like rapamycin and paclitaxel has been shown retrospectively to be the basis of the activity of synthetic molecules originating from drug discovery projects among them lenalidomide and tafamidis. Here, the authors cover the growing number of synthetic small-molecule PPI stabilizers to advocate for a stronger consideration of this as a drug discovery approach. Expert opinion: Both the natural products and the growing number of synthetic molecules show that PPI stabilization is a viable strategy for drug discovery. There is certainly a significant challenge to adapt compound libraries, screening techniques and downstream methodologies to identify, characterize and optimize PPI stabilizers, but the examples of molecules reviewed here in our opinion justify these efforts.


Subject(s)
Drug Design , Drug Discovery/methods , Proteins/metabolism , Biological Products/pharmacology , Humans , Pharmaceutical Preparations/chemical synthesis , Pharmaceutical Preparations/chemistry , Pharmaceutical Preparations/metabolism , Protein Binding , Protein Stability , Small Molecule Libraries
18.
ACS Chem Neurosci ; 8(9): 2065-2077, 2017 09 20.
Article in English | MEDLINE | ID: mdl-28691794

ABSTRACT

Retinoid X receptors (RXRs) play key roles in many physiological processes in both the periphery and central nervous system. In addition, RXRs form heterodimers with other nuclear receptors to exert their physiological effects. The nuclear receptor related 1 protein (NURR1) is particularly interesting because of its role in promoting differentiation and survival of dopamine neurons. However, only a small number of RXR-heterodimer selective modulators are available, with limited chemical diversity. This work describes the synthesis, biochemical evaluation, and structural elucidation of a novel series of RXR ligands with strongly biased interactions with RXRα-NURR1 heterodimers. Targeted modifications to the small molecule biaryl scaffold caused local RXRα side-chain disturbances and displacement of secondary structural elements upon ligand binding. This resulted in the repositioning of protein helices in the heterodimer interface of RXRα, alterations in homo- versus heterodimer formation, and modulation of activation function 2 (AF2). The data provide a rationale for the design of RXR ligands consisting of a highly conserved hydrophilic region, strongly contributing to the ligand affinity, and a variable hydrophobic region, which efficiently probes the effects of structural changes at the level of the ligand on co-regulator recruitment or the RXRα-NURR1 dimerization interface.


Subject(s)
Nuclear Receptor Subfamily 4, Group A, Member 2/chemistry , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Retinoid X Receptor alpha/chemistry , Retinoid X Receptor alpha/metabolism , Drug Design , Escherichia coli , Esters/chemistry , Ethers/chemistry , Humans , Hydrogen Bonding , Ligands , Models, Molecular , Molecular Structure , Nuclear Receptor Subfamily 4, Group A, Member 2/agonists , Nuclear Receptor Subfamily 4, Group A, Member 2/antagonists & inhibitors , Protein Binding , Protein Multimerization , Retinoid X Receptor alpha/agonists , Retinoid X Receptor alpha/antagonists & inhibitors , Two-Hybrid System Techniques
19.
FEBS Lett ; 591(16): 2449-2457, 2017 08.
Article in English | MEDLINE | ID: mdl-28640363

ABSTRACT

14-3-3 proteins are positive regulators of the tumor suppressor p53, the mutation of which is implicated in many human cancers. Current strategies for targeting of p53 involve restoration of wild-type function or inhibition of the interaction with MDM2, its key negative regulator. Despite the efficacy of these strategies, the alternate approach of stabilizing the interaction of p53 with positive regulators and, thus, enhancing tumor suppressor activity, has not been explored. Here, we report the first example of small-molecule stabilization of the 14-3-3 - p53 protein-protein interaction (PPI) and demonstrate the potential of this approach as a therapeutic modality. We also observed a disconnect between biophysical and crystallographic data in the presence of a stabilizing molecule, which is unusual in 14-3-3 PPIs.


Subject(s)
14-3-3 Proteins/metabolism , Glycosides/pharmacology , Tumor Suppressor Protein p53/metabolism , 14-3-3 Proteins/chemistry , Models, Molecular , Protein Binding/drug effects , Protein Stability , Protein Structure, Secondary , Tumor Suppressor Protein p53/chemistry
20.
Angew Chem Int Ed Engl ; 56(20): 5480-5484, 2017 05 08.
Article in English | MEDLINE | ID: mdl-28407400

ABSTRACT

Spiroketals are structural motifs found in many biologically active natural products, which has stimulated considerable efforts toward their synthesis and interest in their use as drug lead compounds. Despite this, the use of spiroketals, and especially bisbenzanulated spiroketals, in a structure-based drug discovery setting has not been convincingly demonstrated. Herein, we report the rational design of a bisbenzannulated spiroketal that potently binds to the retinoid X receptor (RXR) thereby inducing partial co-activator recruitment. We solved the crystal structure of the spiroketal-hRXRα-TIF2 ternary complex, and identified a canonical allosteric mechanism as a possible explanation for the partial agonist behavior of our spiroketal. Our co-crystal structure, the first of a designed spiroketal-protein complex, suggests that spiroketals can be designed to selectively target other nuclear receptor subtypes.


Subject(s)
Furans/chemistry , Nuclear Receptor Coactivator 2/chemistry , Retinoid X Receptor alpha/chemistry , Spiro Compounds/chemistry , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Structure
SELECTION OF CITATIONS
SEARCH DETAIL
...