Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters











Database
Language
Publication year range
1.
J Med Chem ; 67(2): 1544-1562, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38175811

ABSTRACT

NLRP3 is a molecular sensor recognizing a wide range of danger signals. Its activation leads to the assembly of an inflammasome that allows for activation of caspase-1 and subsequent maturation of IL-1ß and IL-18, as well as cleavage of Gasdermin-d and pyroptotic cell death. The NLRP3 inflammasome has been implicated in a plethora of diseases including gout, type 2 diabetes, atherosclerosis, Alzheimer's disease, and cancer. In this publication, we describe the discovery of a novel, tricyclic, NLRP3-binding scaffold by high-throughput screening. The hit (1) could be optimized into an advanced compound NP3-562 demonstrating excellent potency in human whole blood and full inhibition of IL-1ß release in a mouse acute peritonitis model at 30 mg/kg po dose. An X-ray structure of NP3-562 bound to the NLRP3 NACHT domain revealed a unique binding mode as compared to the known sulfonylurea-based inhibitors. In addition, NP3-562 shows also a good overall development profile.


Subject(s)
Diabetes Mellitus, Type 2 , Gout , Mice , Animals , Humans , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Inflammasomes/metabolism , Diabetes Mellitus, Type 2/metabolism , Macrophages/metabolism , Interleukin-1beta/metabolism , Caspase 1/metabolism
2.
J Mol Biol ; 433(24): 167309, 2021 12 03.
Article in English | MEDLINE | ID: mdl-34687713

ABSTRACT

The NLRP3 inflammasome assembles in response to a variety of pathogenic and sterile danger signals, resulting in the production of interleukin-1ß and interleukin-18. NLRP3 is a key component of the innate immune system and has been implicated as a driver of a number of acute and chronic diseases. We report the 2.8 Å crystal structure of the NLRP3 NACHT domain in complex with an inhibitor. The structure defines a binding pocket formed by the four subdomains of the NACHT domain, and shows the inhibitor acts as an intramolecular glue, which locks the protein in an inactive conformation. It provides further molecular insight into our understanding of NLRP3 activation, helps to detail the residues involved in subdomain coordination within the NLRP3 NACHT domain, and gives molecular insights into how gain-of-function mutations de-stabilize the inactive conformation of NLRP3. Finally, it suggests stabilizing the auto-inhibited form of the NACHT domain is an effective way to inhibit NLRP3, and will aid the structure-based development of NLRP3 inhibitors for a range of inflammatory diseases.


Subject(s)
Inflammasomes/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/chemistry , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Furans/chemistry , Furans/pharmacology , Humans , Indenes/chemistry , Indenes/pharmacology , Inflammasomes/metabolism , Protein Domains , Sulfonamides/chemistry , Sulfonamides/pharmacology
3.
Nat Commun ; 12(1): 2442, 2021 04 26.
Article in English | MEDLINE | ID: mdl-33903593

ABSTRACT

The transcription factor PAX8 is critical for the development of the thyroid and urogenital system. Comprehensive genomic screens furthermore indicate an additional oncogenic role for PAX8 in renal and ovarian cancers. While a plethora of PAX8-regulated genes in different contexts have been proposed, we still lack a mechanistic understanding of how PAX8 engages molecular complexes to drive disease-relevant oncogenic transcriptional programs. Here we show that protein isoforms originating from the MECOM locus form a complex with PAX8. These include MDS1-EVI1 (also called PRDM3) for which we map its interaction with PAX8 in vitro and in vivo. We show that PAX8 binds a large number of genomic sites and forms transcriptional hubs. At a subset of these, PAX8 together with PRDM3 regulates a specific gene expression module involved in adhesion and extracellular matrix. This gene module correlates with PAX8 and MECOM expression in large scale profiling of cell lines, patient-derived xenografts (PDXs) and clinical cases and stratifies gynecological cancer cases with worse prognosis. PRDM3 is amplified in ovarian cancers and we show that the MECOM locus and PAX8 sustain in vivo tumor growth, further supporting that the identified function of the MECOM locus underlies PAX8-driven oncogenic functions in ovarian cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , MDS1 and EVI1 Complex Locus Protein/genetics , Ovarian Neoplasms/genetics , PAX8 Transcription Factor/genetics , Animals , Cell Line, Tumor , Female , HEK293 Cells , Humans , MDS1 and EVI1 Complex Locus Protein/metabolism , Mice, Nude , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , PAX8 Transcription Factor/metabolism , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Tumor Burden/genetics , Xenograft Model Antitumor Assays/methods
4.
J Med Chem ; 63(10): 5102-5118, 2020 05 28.
Article in English | MEDLINE | ID: mdl-32083858

ABSTRACT

Bruton's tyrosine kinase (BTK), a cytoplasmic tyrosine kinase, plays a central role in immunity and is considered an attractive target for treating autoimmune diseases. The use of currently marketed covalent BTK inhibitors is limited to oncology indications based on their suboptimal kinase selectivity. We describe the discovery and preclinical profile of LOU064 (remibrutinib, 25), a potent, highly selective covalent BTK inhibitor. LOU064 exhibits an exquisite kinase selectivity due to binding to an inactive conformation of BTK and has the potential for a best-in-class covalent BTK inhibitor for the treatment of autoimmune diseases. It demonstrates potent in vivo target occupancy with an EC90 of 1.6 mg/kg and dose-dependent efficacy in rat collagen-induced arthritis. LOU064 is currently being tested in phase 2 clinical studies for chronic spontaneous urticaria and Sjoegren's syndrome.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Agammaglobulinaemia Tyrosine Kinase/metabolism , Drug Discovery/methods , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Agammaglobulinaemia Tyrosine Kinase/chemistry , Animals , Benzamides/chemistry , Benzamides/metabolism , Benzamides/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Bridged Bicyclo Compounds, Heterocyclic/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Crystallography, X-Ray/methods , Dogs , Dose-Response Relationship, Drug , Female , Humans , Mice , Protein Binding/physiology , Protein Kinase Inhibitors/chemistry , Protein Structure, Secondary , Protein Structure, Tertiary , Rats , Rats, Inbred Lew , Sheep
5.
ACS Med Chem Lett ; 10(10): 1467-1472, 2019 Oct 10.
Article in English | MEDLINE | ID: mdl-31620235

ABSTRACT

Bruton's tyrosine kinase (BTK) is a member of the TEC kinase family and is selectively expressed in a subset of immune cells. It is a key regulator of antigen receptor signaling in B cells and of Fc receptor signaling in mast cells and macrophages. A BTK inhibitor will likely have a positive impact on autoimmune diseases which are caused by autoreactive B cells and immune-complex driven inflammation. We report the design, optimization, and characterization of potent and selective covalent BTK inhibitors. Starting from the selective reversible inhibitor 3 binding to an inactive conformation of BTK, we designed covalent irreversible compounds by attaching an electrophilic warhead to reach Cys481. The first prototype 4 covalently modified BTK and showed an excellent kinase selectivity including several Cys-containing kinases, validating the design concept. In addition, this compound blocked FcγR-mediated hypersensitivity in vivo. Optimization of whole blood potency and metabolic stability resulted in compounds such as 8, which maintained the excellent kinase selectivity and showed improved BTK occupancy in vivo.

6.
Proc Natl Acad Sci U S A ; 116(8): 2935-2944, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30718402

ABSTRACT

Human antigen R (HuR) is a key regulator of cellular mRNAs containing adenylate/uridylate-rich elements (AU-rich elements; AREs). These are a major class of cis elements within 3' untranslated regions, targeting these mRNAs for rapid degradation. HuR contains three RNA recognition motifs (RRMs): a tandem RRM1 and 2, followed by a flexible linker and a C-terminal RRM3. While RRM1 and 2 are structurally characterized, little is known about RRM3. Here we present a 1.9-Å-resolution crystal structure of RRM3 bound to different ARE motifs. This structure together with biophysical methods and cell-culture assays revealed the mechanism of RRM3 ARE recognition and dimerization. While multiple RNA motifs can be bound, recognition of the canonical AUUUA pentameric motif is possible by binding to two registers. Additionally, RRM3 forms homodimers to increase its RNA binding affinity. Finally, although HuR stabilizes ARE-containing RNAs, we found that RRM3 counteracts this effect, as shown in a cell-based ARE reporter assay and by qPCR with native HuR mRNA targets containing multiple AUUUA motifs, possibly by competing with RRM12.


Subject(s)
ELAV Proteins/chemistry , ELAV-Like Protein 1/chemistry , RNA Recognition Motif/genetics , RNA-Binding Proteins/chemistry , 3' Untranslated Regions , AU Rich Elements/genetics , Crystallography, X-Ray , Dimerization , ELAV-Like Protein 1/genetics , Humans , Magnetic Resonance Spectroscopy , RNA-Binding Proteins/genetics , Ribonucleoside Diphosphate Reductase/chemistry , Tumor Suppressor Proteins/chemistry
7.
Chembiochem ; 16(17): 2433-6, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26472355

ABSTRACT

Malaria continues to be one of the most devastating human diseases despite many efforts to limit its spread by prevention of infection or by pharmaceutical treatment of patients. We have conducted a screen for antiplasmodial compounds by using a natural product library. Here we report on cyclomarin A as a potent growth inhibitor of Plasmodium falciparum and the identification of its molecular target, diadenosine triphosphate hydrolase (PfAp3Aase), by chemical proteomics. Using a biochemical assay, we could show that cyclomarin A is a specific inhibitor of the plasmodial enzyme but not of the closest human homologue hFHIT. Co-crystallisation experiments demonstrate a unique binding mode of the inhibitor. One molecule of cyclomarin A binds a dimeric PfAp3Aase and prevents the formation of the enzyme⋅substrate complex. These results validate PfAp3Aase as a new drug target for the treatment of malaria. We have previously elucidated the structurally unrelated regulatory subunit ClpC1 of the ClpP protease as the molecular target of cyclomarin A in Mycobacterium tuberculosis. Thus, cyclomarin A is a rare example of a natural product with two distinct and specific modes of action.


Subject(s)
Biological Products/chemistry , Oligopeptides/chemistry , Acid Anhydride Hydrolases/antagonists & inhibitors , Acid Anhydride Hydrolases/metabolism , Antimalarials/chemistry , Antimalarials/metabolism , Antimalarials/pharmacology , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Binding Sites , Biological Products/metabolism , Biological Products/pharmacology , Endopeptidase Clp/antagonists & inhibitors , Endopeptidase Clp/metabolism , Humans , Inhibitory Concentration 50 , Molecular Dynamics Simulation , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Oligopeptides/metabolism , Oligopeptides/pharmacology , Plasmodium falciparum/drug effects , Plasmodium falciparum/enzymology , Plasmodium falciparum/growth & development , Protein Binding , Protein Structure, Tertiary
8.
J Biol Chem ; 290(24): 15210-8, 2015 Jun 12.
Article in English | MEDLINE | ID: mdl-25918157

ABSTRACT

Macrophages are important cellular effectors in innate immune responses and play a major role in autoimmune diseases such as rheumatoid arthritis. Cancer Osaka thyroid (COT) kinase, also known as mitogen-activated protein kinase kinase kinase 8 (MAP3K8) and tumor progression locus 2 (Tpl-2), is a serine-threonine (ST) kinase and is a key regulator in the production of pro-inflammatory cytokines in macrophages. Due to its pivotal role in immune biology, COT kinase has been identified as an attractive target for pharmaceutical research that is directed at the discovery of orally available, selective, and potent inhibitors for the treatment of autoimmune disorders and cancer. The production of monomeric, recombinant COT kinase has proven to be very difficult, and issues with solubility and stability of the enzyme have hampered the discovery and optimization of potent and selective inhibitors. We developed a protocol for the production of recombinant human COT kinase that yields pure and highly active enzyme in sufficient yields for biochemical and structural studies. The quality of the enzyme allowed us to establish a robust in vitro phosphorylation assay for the efficient biochemical characterization of COT kinase inhibitors and to determine the x-ray co-crystal structures of the COT kinase domain in complex with two ATP-binding site inhibitors. The structures presented in this study reveal two distinct ligand binding modes and a unique kinase domain architecture that has not been observed previously. The structurally versatile active site significantly impacts the design of potent, low molecular weight COT kinase inhibitors.


Subject(s)
MAP Kinase Kinase Kinases/chemistry , Protein Folding , Proto-Oncogene Proteins/chemistry , Crystallography, X-Ray , Humans , Models, Molecular , Protein Conformation , Recombinant Proteins/chemistry
9.
J Biomol NMR ; 51(4): 449-56, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21964698

ABSTRACT

An easy to use and robust approach for amino acid type selective isotope labeling in insect cells is presented. It relies on inexpensive commercial media and can be implemented in laboratories without sophisticated infrastructure. In contrast to previous protocols, where either high protein amounts or high incorporation ratios were obtained, here we achieve both at the same time. By supplementing media with a well considered amount of yeast extract, similar protein amounts as with full media are obtained, without compromising on isotope incorporation. In single and dual amino acid labeling experiments incorporation ratios are consistently ≥90% for all amino acids tested. This enables NMR studies of eukaryotic proteins and their interactions even for proteins with low expression levels. We show applications with human kinases, where protein-ligand interactions are characterized by 2D [(15)N, (1)H]- and [(13)C, (1)H]-HSQC spectra.


Subject(s)
Isotope Labeling/methods , Proto-Oncogene Proteins c-akt/analysis , Proto-Oncogene Proteins c-akt/chemistry , Amino Acids/analysis , Amino Acids/chemistry , Amino Acids/metabolism , Animals , Baculoviridae/chemistry , Cells, Cultured , Humans , Insecta , Nuclear Magnetic Resonance, Biomolecular/methods
SELECTION OF CITATIONS
SEARCH DETAIL