Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
ChemMedChem ; 18(11): e202300051, 2023 06 01.
Article in English | MEDLINE | ID: mdl-36988034

ABSTRACT

The inhibition of the YAP-TEAD protein-protein interaction constitutes a promising therapeutic approach for the treatment of cancers linked to the dysregulation of the Hippo signaling pathway. The identification of a class of small molecules which potently inhibit the YAP-TEAD interaction by binding tightly to the Ω-loop pocket of TEAD has previously been communicated. This report details the further multi-parameter optimization of this class of compounds resulting in advanced analogs combining nanomolar cellular potency with a balanced ADME and off-target profile, and efficacy of these compounds in tumor bearing mice is demonstrated for the first time.


Subject(s)
Neoplasms , Transcription Factors , Animals , Mice , Transcription Factors/metabolism , YAP-Signaling Proteins
2.
ACS Chem Biol ; 18(3): 643-651, 2023 03 17.
Article in English | MEDLINE | ID: mdl-36825662

ABSTRACT

The TEAD transcription factors are the most distal elements of the Hippo pathway, and their transcriptional activity is regulated by several proteins, including YAP. In some cancers, the Hippo pathway is deregulated and inhibitors of the YAP:TEAD interaction are foreseen as new anticancer drugs. The binding of YAP to TEAD is driven by the interaction of an α-helix and an Ω-loop present in its TEAD-binding domain with two distinct pockets at the TEAD surface. Using the mRNA-based display technique to screen a library of in vitro-translated cyclic peptides, we identified a peptide that binds with a nanomolar affinity to TEAD. The X-ray structure of this peptide in complex with TEAD reveals that it interacts with the α-helix pocket. Under our experimental conditions, this peptide can form a ternary complex with TEAD and YAP. Furthermore, combining it with a peptide binding to the Ω-loop pocket gives an additive inhibitory effect on the YAP:TEAD interaction. Overall, our results show that it is possible to identify nanomolar inhibitors of the YAP:TEAD interaction that bind to the α-helix pocket, suggesting that developing such compounds might be a strategy to treat cancers where the Hippo pathway is deregulated.


Subject(s)
Neoplasms , Transcription Factors , Humans , Transcription Factors/metabolism , Protein Conformation, alpha-Helical , TEA Domain Transcription Factors , Peptides/chemistry
3.
Protein Sci ; 32(1): e4545, 2023 01.
Article in English | MEDLINE | ID: mdl-36522189

ABSTRACT

The yes-associated protein (YAP) regulates the transcriptional activity of the TEAD transcription factors that are key in the control of organ morphogenesis. YAP interacts with TEAD via three secondary structure elements: a ß-strand, an α-helix, and an Ω-loop. Earlier results have shown that the ß-strand has only a marginal contribution in the YAP:TEAD interaction, but we show here that it significantly enhances the affinity of YAP for the Drosophila homolog of TEAD, scalloped (Sd). Nuclear magnetic resonance shows that the ß-strand adopts a more rigid conformation once bound to Sd; pre-steady state kinetic measurements show that the YAP:Sd complex is more stable. Although the crystal structures of the YAP:TEAD and YAP:Sd complexes reveal no differences at the binding interface that could explain these results. Molecular Dynamics simulations are in line with our experimental findings regarding ß-strand stability and overall binding affinity of YAP to TEAD and Sd. In particular, RMSF, correlated motion and MMGBSA analyses suggest that ß-sheet fluctuations play a relevant role in YAP53-57 ß-strand dissociation from TEAD4 and contribute to the lower affinity of YAP for TEAD4. Identifying a clear mechanism leading to the difference in YAP's ß-strand stability proved to be challenging, pointing to the potential relevance of multiple modest structural changes or fluctuations for regulation of binding affinity.


Subject(s)
TEA Domain Transcription Factors , Transcription Factors , Transcription Factors/chemistry , DNA-Binding Proteins/chemistry , Protein Conformation, beta-Strand , Gene Expression Regulation , Protein Binding
4.
ChemMedChem ; 17(19): e202200303, 2022 10 06.
Article in English | MEDLINE | ID: mdl-35950546

ABSTRACT

Inhibition of the YAP-TEAD protein-protein interaction is an attractive therapeutic concept under intense investigation with the objective to treat cancers associated with a dysregulation of the Hippo pathway. However, owing to the very extended surface of interaction of the two proteins, the identification of small drug-like molecules able to efficiently prevent YAP from binding to TEAD by direct competition has been elusive so far. We disclose here the discovery of the first class of small molecules potently inhibiting the YAP-TEAD interaction by binding at one of the main interaction sites of YAP at the surface of TEAD. These inhibitors, providing a path forward to pharmacological intervention in the Hippo pathway, evolved from a weakly active virtual screening hit advanced to high potency by structure-based design.


Subject(s)
Neoplasms , Transcription Factors , Adaptor Proteins, Signal Transducing/chemistry , Humans , Transcription Factors/metabolism , YAP-Signaling Proteins
5.
Cell Chem Biol ; 28(9): 1271-1282.e12, 2021 09 16.
Article in English | MEDLINE | ID: mdl-33894161

ABSTRACT

Acute kidney injury (AKI) is a life-threatening disease with no known curative or preventive therapies. Data from multiple animal models and human studies have linked dysregulation of bone morphogenetic protein (BMP) signaling to AKI. Small molecules that potentiate endogenous BMP signaling should have a beneficial effect in AKI. We performed a high-throughput phenotypic screen and identified a series of FK506 analogs that act as potent BMP potentiators by sequestering FKBP12 from BMP type I receptors. We further showed that calcineurin inhibition was not required for this activity. We identified a calcineurin-sparing FK506 analog oxtFK through late-stage functionalization and structure-guided design. OxtFK demonstrated an improved safety profile in vivo relative to FK506. OxtFK stimulated BMP signaling in vitro and in vivo and protected the kidneys in an AKI mouse model, making it a promising candidate for future development as a first-in-class therapeutic for diseases with dysregulated BMP signaling.


Subject(s)
Acute Kidney Injury/drug therapy , Bone Morphogenetic Proteins/metabolism , Tacrolimus/pharmacology , Animals , Cells, Cultured , Disease Models, Animal , High-Throughput Screening Assays , Humans , Male , Mice , Mice, Inbred C57BL , Molecular Structure , Phenotype , Tacrolimus/analogs & derivatives , Tacrolimus/chemistry
6.
Nat Commun ; 12(1): 898, 2021 02 09.
Article in English | MEDLINE | ID: mdl-33563973

ABSTRACT

Radiation sensitivity varies greatly between tissues. The transcription factor p53 mediates the response to radiation; however, the abundance of p53 protein does not correlate well with the extent of radiosensitivity across tissues. Given recent studies showing that the temporal dynamics of p53 influence the fate of cultured cells in response to irradiation, we set out to determine the dynamic behavior of p53 and its impact on radiation sensitivity in vivo. We find that radiosensitive tissues show prolonged p53 signaling after radiation, while more resistant tissues show transient p53 activation. Sustaining p53 using a small molecule (NMI801) that inhibits Mdm2, a negative regulator of p53, reduced viability in cell culture and suppressed tumor growth. Our work proposes a mechanism for the control of radiation sensitivity and suggests tools to alter the dynamics of p53 to enhance tumor clearance. Similar approaches can be used to enhance killing of cancer cells or reduce toxicity in normal tissues following genotoxic therapies.


Subject(s)
Radiation Tolerance , Tumor Suppressor Protein p53/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Humans , Mice , Neoplasms/drug therapy , Neoplasms/radiotherapy , Protein Binding/drug effects , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/metabolism , Radiation Tolerance/drug effects , Tissue Distribution/drug effects , Tumor Burden/drug effects , Tumor Suppressor Protein p53/radiation effects , Xenograft Model Antitumor Assays
7.
RSC Chem Biol ; 2(6): 1661-1668, 2021 Dec 02.
Article in English | MEDLINE | ID: mdl-34977581

ABSTRACT

Mouse double minute 2 homolog (MDM2, Hdm2) is an important negative regulator of the tumor suppressor p53. Using a mRNA based display technique to screen a library of >1012 in vitro-translated cyclic peptides, we have identified a macrocyclic ligand that shows picomolar potency on MDM2. X-Ray crystallography reveals a novel binding mode utilizing a unique pharmacophore to occupy the Phe/Trp/Leu pockets on MDM2. Conjugation of a cyclic cell-penetrating peptide (cCPP) to the initially non cell-permeable ligand enables cellular uptake and a pharmacodynamic response in SJSA-1 cells. The demonstrated enhanced intracellular availability of cyclic peptides that are identified by a display technology exemplifies a process for the application of intracellular tools for drug discovery projects.

8.
J Med Chem ; 62(23): 10816-10832, 2019 12 12.
Article in English | MEDLINE | ID: mdl-31729873

ABSTRACT

Retinoic acid receptor-related orphan receptor gamma-t (RORγt) is considered to be the master transcription factor for the development of Th17 cells that produce proinflammatory cytokines such as IL-17A. Overproportionate Th17 cell abundance is associated with the pathogenesis of many inflammatory conditions including psoriasis. In a high-throughput fluorescence resonance energy transfer (FRET) screen, we identified compound 1 as a hit with promising lipophilic efficiency (LipE). Using structure-based drug design based on a number of X-ray cocrystal structures, we morphed this hit class into potent imidazoles, exemplified by compound 3. To improve the poor absorption, distribution, metabolism, and excretion (ADME) properties of neutral imidazoles, we extended our ligands with carboxylic acid substituents toward a polar, water-rich area of the protein. This highly lipophilicity-efficient modification ultimately led to the discovery of compound 14, a potent and selective inhibitor of RORγt with good ADME properties and excellent in vivo pharmacokinetics. This compound showed good efficacy in an in vivo delayed-type hypersensitivity pharmacology model in rats.


Subject(s)
Hypersensitivity, Delayed/drug therapy , Imidazoles/pharmacology , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Administration, Oral , Animals , Dose-Response Relationship, Drug , Drug Design , Female , Fluorescence Resonance Energy Transfer , Half-Life , Imidazoles/chemistry , Imidazoles/pharmacokinetics , Male , Models, Molecular , Molecular Structure , Rats
9.
Bioorg Med Chem Lett ; 29(16): 2316-2319, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31235263

ABSTRACT

The YAP-TEAD protein-protein interaction is a potential therapeutic target to treat cancers in which the Hippo signaling pathway is deregulated. However, the extremely large surface of interaction between the two proteins presents a formidable challenge for a small molecule interaction disrupter approach. We have accomplished progress towards showing the feasibility of this approach by the identification of a 15-mer peptide able to potently (nanomolar range) disrupt the YAP-TEAD interaction by targeting only one of the two important sites of interaction. This peptide, incorporating non-natural amino acids selected by structure-based design, is derived from the Ω-loop sequence 85-99 of YAP.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Drug Design , Peptides/pharmacology , Small Molecule Libraries/pharmacology , Transcription Factors/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/chemistry , Dose-Response Relationship, Drug , Humans , Molecular Structure , Peptides/chemical synthesis , Peptides/chemistry , Protein Binding/drug effects , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Transcription Factors/chemistry , YAP-Signaling Proteins
10.
ChemMedChem ; 14(14): 1305-1314, 2019 07 17.
Article in English | MEDLINE | ID: mdl-31066983

ABSTRACT

Hdm2 (human MDM2, human double minute 2 homologue) counteracts p53 function by direct binding to p53 and by ubiquitin-dependent p53 protein degradation. Activation of p53 by inhibitors of the p53-Hdm2 interaction is being pursued as a therapeutic strategy in p53 wild-type cancers. In addition, HdmX (human MDMX, human MDM4) was also identified as an important therapeutic target to efficiently reactivate p53, and it is likely that dual inhibition of Hdm2 and HdmX is beneficial. Herein we report four new X-ray structures for Hdm2 and five new X-ray structures for HdmX complexes, involving different classes of synthetic compounds (including the worldwide highest resolutions for Hdm2 and HdmX, at 1.13 and 1.20 Å, respectively). We also reveal the key additive 18-crown-ether, which we discovered to enable HdmX crystallization and show its stabilization of various Lys residues. In addition, we report the previously unpublished details of X-ray structure determinations for eight further Hdm2 complexes, including the clinical trial compounds NVP-CGM097 and NVP-HDM201. An analysis of all compound binding modes reveals new and deepened insight into the possible adaptations and structural states of Hdm2 (e.g., flip of F55, flip of Y67, reorientation of H96) and HdmX (e.g., flip of H55, dimer induction), enabling key binding interactions for different compound classes. To facilitate comparisons, we used the same numbering for Hdm2 (as in Q00987) and HdmX (as in O15151, but minus 1). Taken together, these structural insights should prove useful for the design and optimization of further selective and/or dual Hdm2/HdmX inhibitors.


Subject(s)
Cell Cycle Proteins/metabolism , Heterocyclic Compounds/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins/metabolism , Binding Sites , Cell Cycle Proteins/chemistry , Crystallography, X-Ray , Heterocyclic Compounds/chemistry , Humans , Protein Binding , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins c-mdm2/chemistry
11.
FEBS J ; 286(12): 2381-2398, 2019 06.
Article in English | MEDLINE | ID: mdl-30903741

ABSTRACT

Four TEAD transcription factors (TEAD1-4) mediate the signalling output of the Hippo pathway that controls organ size in humans. TEAD transcriptional activity is regulated via interactions with the YAP, TAZ and VGLL proteins. A mutation in the TEAD1 gene, Tyr421His, has been identified in patients suffering from Sveinsson's chorioretinal atrophy (SCA), an autosomal dominant eye disease. This mutation prevents the YAP/TAZ:TEAD1 interaction. In this study, we measure the affinity of YAP, TAZ and VGLL1 for the four human TEADs and find that they have a similar affinity for all TEADs. We quantitate the effect of the mutation found in SCA patients and show that it destabilizes the YAP/TAZ:TEAD interaction by about 3 kcal·mol-1 . We determine the structure of YAP in complex with this mutant form of TEAD and show that the histidine residue adopts different conformations at the binding interface. The presence of this flexible residue induces the destabilization of several H-bonds and the loss of van der Waals contacts, which explains why the Tyr421HisTEAD1 mutation has such a large destabilizing effect on the formation of the YAP:TEAD complex. DATABASE: The crystallographic data have been deposited at the RSCB Protein Data Bank (PDB, www.pdb.org) with the access codes: 6HIL (wtYAP :Tyr421HisTEAD1 ), 6HIK (wtYAP :Tyr429HisTEAD4 ).


Subject(s)
Cell Cycle Proteins/genetics , Corneal Dystrophies, Hereditary/genetics , DNA-Binding Proteins/chemistry , Nuclear Proteins/chemistry , Protein Interaction Maps/genetics , Retinal Degeneration/genetics , Transcription Factors/chemistry , Transcription Factors/genetics , Acyltransferases , Cell Cycle Proteins/chemistry , Corneal Dystrophies, Hereditary/pathology , Crystallography, X-Ray , DNA-Binding Proteins/genetics , Humans , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/genetics , Mutation/genetics , Nuclear Proteins/genetics , Protein Binding/genetics , Retinal Degeneration/pathology , Signal Transduction/genetics , TEA Domain Transcription Factors
12.
Bioorg Med Chem Lett ; 28(20): 3404-3408, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30217415

ABSTRACT

Small molecule inhibitors of the p53-MDM2 protein complex are under intense investigation in clinical trials as anti-cancer agents, including our first generation inhibitor NVP-CGM097. We recently described the rational design of a novel pyrazolopyrrolidinone core as a new lead structure and now we report on the synthesis and optimization of this to provide a highly potent lead compound. This new compound displayed excellent oral efficacy in our preclinical mechanistic in vivo model and marked a significant milestone towards the identification of our second generation clinical candidate NVP-HDM201.


Subject(s)
Antineoplastic Agents/pharmacology , Protein Multimerization/drug effects , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Pyrazoles/pharmacology , Pyrrolidinones/pharmacology , Tumor Suppressor Protein p53/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Dogs , Haplorhini , Humans , Male , Mice , Microsomes, Liver/metabolism , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Pyrazoles/pharmacokinetics , Pyrrolidinones/chemical synthesis , Pyrrolidinones/chemistry , Pyrrolidinones/pharmacokinetics , Rats, Sprague-Dawley , Stereoisomerism
13.
Cancer Res ; 78(21): 6257-6267, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30135191

ABSTRACT

Activation of p53 by inhibitors of the p53-MDM2 interaction is being pursued as a therapeutic strategy in p53 wild-type cancers. Here, we report distinct mechanisms by which the novel, potent, and selective inhibitor of the p53-MDM2 interaction HDM201 elicits therapeutic efficacy when applied at various doses and schedules. Continuous exposure of HDM201 led to induction of p21 and delayed accumulation of apoptotic cells. By comparison, high-dose pulses of HDM201 were associated with marked induction of PUMA and a rapid onset of apoptosis. shRNA screens identified PUMA as a mediator of the p53 response specifically in the pulsed regimen. Consistent with this, the single high-dose HDM201 regimen resulted in rapid and marked induction of PUMA expression and apoptosis together with downregulation of Bcl-xL in vivo Knockdown of Bcl-xL was identified as the top sensitizer to HDM201 in vitro, and Bcl-xL was enriched in relapsing tumors from mice treated with intermittent high doses of HDM201. These findings define a regimen-dependent mechanism by which disruption of MDM2-p53 elicits therapeutic efficacy when given with infrequent dosing. In an ongoing HDM201 trial, the observed exposure-response relationship indicates that the molecular mechanism elicited by pulse dosing is likely reproducible in patients. These data support the clinical comparison of daily and intermittent regimens of p53-MDM2 inhibitors.Significance: Pulsed high doses versus sustained low doses of the p53-MDM2 inhibitor HDM201 elicit a proapoptotic response from wild-type p53 cancer cells, offering guidance to current clinical trials with this and other drugs that exploit the activity of p53. Cancer Res; 78(21); 6257-67. ©2018 AACR.


Subject(s)
Antineoplastic Agents/administration & dosage , Imidazoles/administration & dosage , Neoplasms/drug therapy , Neoplasms/metabolism , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Pyrimidines/administration & dosage , Pyrroles/administration & dosage , Tumor Suppressor Protein p53/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Area Under Curve , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Drug Screening Assays, Antitumor , Humans , Imidazoles/pharmacology , Kaplan-Meier Estimate , Maximum Tolerated Dose , Mice , Neoplasm Transplantation , Pyrimidines/pharmacology , Pyrroles/pharmacology , RNA, Small Interfering/metabolism , Time Factors , bcl-X Protein/metabolism
14.
J Med Chem ; 61(15): 6724-6735, 2018 08 09.
Article in English | MEDLINE | ID: mdl-29990434

ABSTRACT

The transcription factor RORγt is an attractive drug-target due to its role in the differentiation of IL-17 producing Th17 cells that play a critical role in the etiopathology of several autoimmune diseases. Identification of starting points for RORγt inverse agonists with good properties has been a challenge. We report the identification of a fragment hit and its conversion into a potent inverse agonist through fragment optimization, growing and merging efforts. Further analysis of the binding mode revealed that inverse agonism was achieved by an unusual mechanism. In contrast to other reported inverse agonists, there is no direct interaction or displacement of helix 12 observed in the crystal structure. Nevertheless, compound 9 proved to be efficacious in a delayed-type hypersensitivity (DTH) inflammation model in rats.


Subject(s)
Drug Discovery , Drug Inverse Agonism , Nuclear Receptor Subfamily 1, Group F, Member 3/agonists , Animals , Catalytic Domain , Disease Models, Animal , Female , Inflammation/metabolism , Models, Molecular , Rats
15.
ChemMedChem ; 13(18): 1997-2007, 2018 09 19.
Article in English | MEDLINE | ID: mdl-29985556

ABSTRACT

CLK2 inhibition has been proposed as a potential mechanism to improve autism and neuronal functions in Phelan-McDermid syndrome (PMDS). Herein, the discovery of a very potent indazole CLK inhibitor series and the CLK2 X-ray structure of the most potent analogue are reported. This new indazole series was identified through a biochemical CLK2 Caliper assay screen with 30k compounds selected by an in silico approach. Novel high-resolution X-ray structures of all CLKs, including the first CLK4 X-ray structure, bound to known CLK2 inhibitor tool compounds (e.g., TG003, CX-4945), are also shown and yield insight into inhibitor selectivity in the CLK family. The efficacy of the new CLK2 inhibitors from the indazole series was demonstrated in the mouse brain slice assay, and potential safety concerns were investigated. Genotoxicity findings in the human lymphocyte micronucleus test (MNT) assay are shown by using two structurally different CLK inhibitors to reveal a major concern for pan-CLK inhibition in PMDS.


Subject(s)
Chromosome Disorders/drug therapy , Indazoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Chromosome Deletion , Chromosome Disorders/metabolism , Chromosomes, Human, Pair 22/metabolism , Crystallography, X-Ray , Dose-Response Relationship, Drug , Drug Discovery , Humans , Indazoles/chemical synthesis , Indazoles/chemistry , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Structure-Activity Relationship
16.
Protein Sci ; 27(10): 1810-1820, 2018 10.
Article in English | MEDLINE | ID: mdl-30058229

ABSTRACT

Many interactions between proteins are mediated by intrinsically disordered regions (IDRs). Intrinsically disordered proteins (IDPs) do not adopt a stable three-dimensional structure in their unbound form, but they become more structured upon binding to their partners. In this communication, we study how a bound IDR adapts to mutations, preventing the formation of hydrogen bonds at the binding interface that needs a precise positioning of the interacting residues to be formed. We use as a model the YAP:TEAD interface, where one YAP (IDP) and two TEAD residues form hydrogen bonds via their side chain. Our study shows that the conformational flexibility of bound YAP and the reorganization of water molecules at the interface help to reduce the energetic constraints created by the loss of H-bonds at the interface. The residual flexibility/dynamic of bound IDRs and water might, therefore, be a key for the adaptation of IDPs to different interface landscapes and to mutations occurring at binding interfaces.


Subject(s)
Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/genetics , Mutation , Protein Binding , Protein Conformation , Thermodynamics , Water/chemistry
17.
PLoS One ; 12(11): e0188391, 2017.
Article in English | MEDLINE | ID: mdl-29155882

ABSTRACT

Retinoic acid receptor-related-orphan-receptor-C (RORγt) is the key transcription factor that is driving the differentiation of IL-17 producing T-helper 17 (Th17) cells that are implicated in the pathology of various autoimmune and inflammatory diseases. Based on the importance of RORγt in promoting Th17-driven pathology, there is considerable interest to develop low-molecular-weight compounds with the aim of inhibiting the transcriptional activity of this nuclear hormone receptor. In this article, we describe the in vitro and in vivo pharmacology of a potent and selective small-molecular-weight RORγt inverse agonist. The compound binds to the ligand binding domain (LBD) of RORγt leading to displacement of a co-activator peptide. We show for the first time that a RORγt inverse agonist down-regulates permissive histone H3 acetylation and methylation at the IL17A and IL23R promoter regions, thereby providing insight into the transcriptional inhibition of RORγt-dependent genes. Consistent with this, the compound effectively reduced IL-17A production by polarized human T-cells and γδT-cells and attenuated transcription of RORγt target genes. The inhibitor showed good in vivo efficacy in an antigen-induced arthritis model in rats and reduced the frequencies of IL-17A producing cells in ex vivo recall assays. In summary, we demonstrate that inhibiting RORγt by a low-molecular-weight inhibitor results in efficient and selective blockade of the pro-inflammatory Th17/IL-17A pathway making it an attractive target for Th17-mediated disorders.


Subject(s)
Arthritis, Experimental/drug therapy , Imidazoles/pharmacology , Interleukin-17/antagonists & inhibitors , Intraepithelial Lymphocytes/drug effects , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Pyridines/pharmacology , Pyrimidines/pharmacology , Th17 Cells/drug effects , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Cell Line, Tumor , Female , Gene Expression Regulation , HEK293 Cells , Humans , Imidazoles/chemical synthesis , Interleukin-17/genetics , Interleukin-17/immunology , Intraepithelial Lymphocytes/immunology , Intraepithelial Lymphocytes/pathology , Kinetics , Male , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Promoter Regions, Genetic , Protein Binding , Pyridines/chemical synthesis , Pyrimidines/chemical synthesis , Rats , Rats, Inbred Lew , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/immunology , Receptors, Interleukin/genetics , Receptors, Interleukin/immunology , Signal Transduction , Th17 Cells/immunology , Th17 Cells/pathology
18.
Protein Sci ; 26(12): 2399-2409, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28960584

ABSTRACT

The Hippo pathway is deregulated in various cancers, and the discovery of molecules that modulate this pathway may open new therapeutic avenues in oncology. TEA/ATTS domain (TEAD) transcription factors are the most distal elements of the Hippo pathway and their transcriptional activity is regulated by the Yes-associated protein (YAP). Amongst the various possibilities for targeting this pathway, inhibition of the YAP:TEAD interaction is an attractive strategy. It has been shown recently that TEAD proteins are covalently linked via a conserved cysteine to a fatty acid molecule (palmitate) that binds to a deep hydrophobic cavity present in these proteins. This acylation of TEAD seems to be required for efficient binding to YAP, and understanding how it modulates the YAP:TEAD interaction may provide useful information on the regulation of TEAD function. In this report we have studied the effect of TEAD4 acylation on its interaction with YAP and the other co-activator transcriptional co-activator with PDZ-binding motif (TAZ). We show in our biochemical and cellular assays that YAP and TAZ bind in a similar manner to acylated and non-acylated TEAD4. This indicates that TEAD4 acylation is not a prerequisite for its interaction with YAP or TAZ. However, we observed that TEAD4 acylation significantly enhances its stability, suggesting that it may help this transcription factor to acquire and/or maintain its active conformation.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Muscle Proteins/chemistry , Muscle Proteins/metabolism , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism , Acylation/physiology , Humans , Models, Molecular , Signal Transduction/physiology , TEA Domain Transcription Factors , Trans-Activators , Transcriptional Coactivator with PDZ-Binding Motif Proteins , YAP-Signaling Proteins
19.
ChemMedChem ; 12(13): 1014-1021, 2017 07 06.
Article in English | MEDLINE | ID: mdl-28590087

ABSTRACT

The T-cell-specific retinoic acid receptor (RAR)-related orphan receptor-γ (RORγt) is a key transcription factor for the production of pro-inflammatory Th17 cytokines, which are implicated in the pathogenesis of autoimmune diseases. Over the years, several structurally diverse RORγt inverse agonists have been reported, but combining high potency and good physicochemical properties has remained a challenging task. We recently reported a new series of inverse agonists based on an imidazopyridine core with good physicochemical properties and excellent selectivity. Herein we report eight new X-ray crystal structures for different classes of natural and synthetic compounds, including examples selected from the patent literature. Analysis of their respective binding modes revealed insight into the molecular mechanisms that lead to agonism, antagonism, or inverse agonism. We report new molecular mechanisms for RORγt agonism and propose a separation of the inverse agonists into two classes: those that act via steric clash and those that act via other mechanisms (for the latter, co-crystallization with a co-activator peptide and helix 12 in the agonist position is still possible). For the non-steric clash inverse agonists, we propose a new mechanism ("water trapping") which can be combined with other mechanisms (e.g., close contacts with H479). In addition, we compare the interactions made for selected compounds in the "back pocket" near S404 and in the "sulfate pocket" near R364 and R367. Taken together, these new mechanistic insights should prove useful for the design and optimization of further RORγt modulators.


Subject(s)
Nuclear Receptor Subfamily 1, Group F, Member 3/chemistry , Adaptor Proteins, Signal Transducing/chemistry , Binding Sites , Cholesterol Esters/chemistry , Crystallography, X-Ray , Humans , Hydrocarbons, Fluorinated/chemistry , Imidazoles/chemistry , Models, Chemical , Nuclear Proteins/chemistry , Nuclear Receptor Interacting Protein 1 , Nuclear Receptor Subfamily 1, Group F, Member 3/agonists , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Pyridines/chemistry , Sulfonamides/chemistry , Sulfones/chemistry , Water/chemistry
20.
ChemMedChem ; 12(3): 197-201, 2017 02 03.
Article in English | MEDLINE | ID: mdl-27863026

ABSTRACT

A series of potent quinazolinedione sulfonamide antagonists of the α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid (AMPA) receptor were designed and synthesized. The structure-activity relationships (SAR) and in vivo activity of the series were investigated. In particular, compound 1 S (selurampanel; N-[7-isopropyl-6-(2-methylpyrazol-3-yl)-2,4-dioxo-1H-quinazolin-3-yl]methanesulfonamide) has shown excellent oral potency against maximal electroshock seizure (MES)-induced generalized tonic-clonic seizures in rodents as well as significant activity in patients suffering from various forms of epilepsy. The X-ray crystal structure of selurampanel bound to the AMPA receptor hGluA was also obtained.


Subject(s)
Drug Design , Quinazolinones/chemistry , Receptors, AMPA/antagonists & inhibitors , Administration, Oral , Animals , Binding Sites , Binding, Competitive , Crystallography, X-Ray , Disease Models, Animal , Electroshock , Mice , Molecular Dynamics Simulation , Protein Structure, Tertiary , Quinazolinones/administration & dosage , Quinazolinones/chemical synthesis , Quinazolinones/metabolism , Receptors, AMPA/metabolism , Seizures/drug therapy , Structure-Activity Relationship , Sulfonamides/administration & dosage , Sulfonamides/chemical synthesis , Sulfonamides/chemistry , Sulfonamides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL