Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Science ; 376(6589): 163-169, 2022 04 08.
Article in English | MEDLINE | ID: mdl-35271300

ABSTRACT

Cytokines signal through cell surface receptor dimers to initiate activation of intracellular Janus kinases (JAKs). We report the 3.6-angstrom-resolution cryo-electron microscopy structure of full-length JAK1 complexed with a cytokine receptor intracellular domain Box1 and Box2 regions captured as an activated homodimer bearing the valine→phenylalanine (VF) mutation prevalent in myeloproliferative neoplasms. The seven domains of JAK1 form an extended structural unit, the dimerization of which is mediated by close-packing of the pseudokinase (PK) domains from the monomeric subunits. The oncogenic VF mutation lies within the core of the JAK1 PK interdimer interface, enhancing packing complementarity to facilitate ligand-independent activation. The carboxy-terminal tyrosine kinase domains are poised for transactivation and to phosphorylate the receptor STAT (signal transducer and activator of transcription)-recruiting motifs projecting from the overhanging FERM (four-point-one, ezrin, radixin, moesin)-SH2 (Src homology 2)-domains. Mapping of constitutively active JAK mutants supports a two-step allosteric activation mechanism and reveals opportunities for selective therapeutic targeting of oncogenic JAK signaling.


Subject(s)
Janus Kinase 1 , Receptors, Cytokine , src Homology Domains , Allosteric Regulation , Cryoelectron Microscopy , Enzyme Activation , Humans , Janus Kinase 1/chemistry , Janus Kinase 1/metabolism , Mutation , Myeloproliferative Disorders/enzymology , Myeloproliferative Disorders/genetics , Phosphorylation , Protein Multimerization , Receptors, Cytokine/chemistry , STAT Transcription Factors/metabolism
2.
Sci Transl Med ; 13(625): eabg7565, 2021 12 22.
Article in English | MEDLINE | ID: mdl-34936383

ABSTRACT

Chimeric antigen receptor (CAR) T cells induce durable responses in patients with refractory hematological tumors. However, low CAR T cell activity, poor engraftment, or short in-patient persistence can lead to tumor progression or relapse. Furthermore, excessive CAR T cell expansion and activation can result in life-threatening cytokine release syndrome (CRS). Thus, in-patient control of the CAR T cell population is essential. Interleukin-2 (IL-2) is a critical cytokine for T cell proliferation and effector function, but its clinical use is limited by immune-mediated toxicity. Here, we report on an orthogonal IL-2 receptor and ligand system that enables specific in vivo control of CAR T cell expansion and activation, wherein an orthogonal human IL-2 (STK-009) selectively pairs with an orthogonal human IL-2Rß (hoRb) expressed on CAR T cells. STK-009 expands hoRb-expressing CAR T cells in the presence and absence of tumor antigen and maintains the presence of stem cell memory T cells (TSCM) and effector T cells. In preclinical models of human CAR-refractory lymphoma, STK-009 treatment resulted in systemic and intratumoral expansion and activation of hoRb-expressing anti­CD19-CD28ζ CAR T cells (SYNCAR). The orthogonal IL-2 receptor/ligand system delivers complete responses in large subcutaneous lymphomas, even with substantially reduced CAR T cell doses, by selectively expanding and activating CAR T cells in vivo. STK-009 withdrawal allowed normal CAR T cell contraction, thereby limiting CRS induced by tumor antigen­specific T cell activation. These data suggest that the orthogonal IL-2 receptor/ligand system provides the in vivo control necessary to maximize efficacy of CAR T therapies.


Subject(s)
Interleukin-2 , Lymphoma , Antigens, CD19 , Humans , Immunotherapy, Adoptive , Lymphoma/therapy , Neoplasm Recurrence, Local/therapy , Receptors, Antigen, T-Cell , T-Lymphocytes
3.
Cell ; 184(4): 983-999.e24, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33606986

ABSTRACT

Interleukin-12 (IL-12) and IL-23 are heterodimeric cytokines that are produced by antigen-presenting cells to regulate the activation and differentiation of lymphocytes, and they share IL-12Rß1 as a receptor signaling subunit. We present a crystal structure of the quaternary IL-23 (IL-23p19/p40)/IL-23R/IL-12Rß1 complex, together with cryoelectron microscopy (cryo-EM) maps of the complete IL-12 (IL-12p35/p40)/IL-12Rß2/IL-12Rß1 and IL-23 receptor (IL-23R) complexes, which reveal "non-canonical" topologies where IL-12Rß1 directly engages the common p40 subunit. We targeted the shared IL-12Rß1/p40 interface to design a panel of IL-12 partial agonists that preserved interferon gamma (IFNγ) induction by CD8+ T cells but impaired cytokine production from natural killer (NK) cells in vitro. These cell-biased properties were recapitulated in vivo, where IL-12 partial agonists elicited anti-tumor immunity to MC-38 murine adenocarcinoma absent the NK-cell-mediated toxicity seen with wild-type IL-12. Thus, the structural mechanism of receptor sharing used by IL-12 family cytokines provides a protein interface blueprint for tuning this cytokine axis for therapeutics.


Subject(s)
Interleukin-12/chemistry , Interleukin-12/metabolism , Killer Cells, Natural/metabolism , Receptors, Interleukin/chemistry , Receptors, Interleukin/metabolism , T-Lymphocytes/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cryoelectron Microscopy , Crystallography, X-Ray , Epitopes/immunology , Female , HEK293 Cells , Humans , Immunity , Interleukin-12/agonists , Interleukin-12 Subunit p40/chemistry , Interleukin-12 Subunit p40/metabolism , Mice, Inbred C57BL , Models, Molecular , Neoplasms/immunology , Neoplasms/pathology , Protein Structure, Quaternary , Receptors, Interleukin/ultrastructure , Receptors, Interleukin-12/metabolism , Signal Transduction , Structure-Activity Relationship
4.
ACS Med Chem Lett ; 11(3): 327-333, 2020 Mar 12.
Article in English | MEDLINE | ID: mdl-32184965

ABSTRACT

IRAK4 kinase activity transduces signaling from multiple IL-1Rs and TLRs to regulate cytokines and chemokines implicated in inflammatory diseases. As such, there is high interest in identifying selective IRAK4 inhibitors for the treatment of these disorders. We previously reported the discovery of potent and selective dihydrobenzofuran inhibitors of IRAK4. Subsequent studies, however, showed inconsistent inhibition in disease-relevant pharmacodynamic models. Herein, we describe application of a human whole blood assay to the discovery of a series of benzolactam IRAK4 inhibitors. We identified potent molecule 19 that achieves robust in vivo inhibition of cytokines relevant to human disease.

5.
J Med Chem ; 62(13): 6223-6240, 2019 07 11.
Article in English | MEDLINE | ID: mdl-31082230

ABSTRACT

A series of pyrazolopyrimidine inhibitors of IRAK4 were developed from a high-throughput screen (HTS). Modification of an HTS hit led to a series of bicyclic heterocycles with improved potency and kinase selectivity but lacking sufficient solubility to progress in vivo. Structure-based drug design, informed by cocrystal structures with the protein and small-molecule crystal structures, yielded a series of dihydrobenzofurans. This semisaturated bicycle provided superior druglike properties while maintaining excellent potency and selectivity. Improved physicochemical properties allowed for progression into in vivo experiments, where lead molecules exhibited low clearance and showed target-based inhibition of IRAK4 signaling in an inflammation-mediated PK/PD mouse model.


Subject(s)
Interleukin-1 Receptor-Associated Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Aminoquinolines/chemical synthesis , Aminoquinolines/metabolism , Aminoquinolines/pharmacology , Animals , Benzofurans/chemical synthesis , Benzofurans/metabolism , Benzofurans/pharmacology , Catalytic Domain , Female , Humans , Interleukin-1 Receptor-Associated Kinases/metabolism , Mice, Inbred C57BL , Molecular Structure , Protein Binding , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/metabolism , Pyrazoles/chemical synthesis , Pyrazoles/metabolism , Pyrimidines/chemical synthesis , Pyrimidines/metabolism , Rats , Signal Transduction/drug effects , Structure-Activity Relationship
6.
Bioorg Med Chem Lett ; 29(12): 1497-1501, 2019 06 15.
Article in English | MEDLINE | ID: mdl-31000154

ABSTRACT

Receptor-interacting protein kinase 1 (RIPK1), a key component of the cellular necroptosis pathway, has gained recognition as an important therapeutic target. Pharmacologic inhibition or genetic inactivation of RIPK1 has shown promise in animal models of disease ranging from acute ischemic conditions, chronic inflammation, and neurodegeneration. We present here a class of RIPK1 inhibitors that is distinguished by a lack of a lipophilic aromatic group present in most literature inhibitors that typically occupies a hydrophobic back pocket of the protein active site. Despite not having this ubiquitous feature of many known RIPK1 inhibitors, we were able to obtain compounds with good potency, kinase selectivity, and pharmacokinetic properties in rats. The use of the lipophilic yet metabolically stable pentafluoroethyl group was critical to balancing the potency and properties of optimized analogs.


Subject(s)
Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Humans , Necrosis , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Structure-Activity Relationship
7.
MAbs ; 10(7): 979-991, 2018 10.
Article in English | MEDLINE | ID: mdl-30102105

ABSTRACT

Infections caused by methicillin-resistant Staphylococcus aureus (MRSA) are a growing health threat worldwide. Efforts to identify novel antibodies that target S. aureus cell surface antigens are a promising direction in the development of antibiotics that can halt MRSA infection. We biochemically and structurally characterized three patient-derived MRSA-targeting antibodies that bind to wall teichoic acid (WTA), which is a polyanionic surface glycopolymer. In S. aureus, WTA exists in both α- and ß-forms, based on the stereochemistry of attachment of a N-acetylglucosamine residue to the repeating phosphoribitol sugar unit. We identified a panel of antibodies cloned from human patients that specifically recognize the α or ß form of WTA, and can bind with high affinity to pathogenic wild-type strains of S. aureus bacteria. To investigate how the ß-WTA specific antibodies interact with their target epitope, we determined the X-ray crystal structures of the three ß-WTA specific antibodies, 4462, 4497, and 6078 (Protein Data Bank IDs 6DWI, 6DWA, and 6DW2, respectively), bound to a synthetic WTA epitope. These structures reveal that all three of these antibodies, while utilizing distinct antibody complementarity-determining region sequences and conformations to interact with ß-WTA, fulfill two recognition principles: binding to the ß-GlcNAc pyranose core and triangulation of WTA phosphate residues with polar contacts. These studies reveal the molecular basis for targeting a unique S. aureus cell surface epitope and highlight the power of human patient-based antibody discovery techniques for finding novel pathogen-targeting therapeutics.


Subject(s)
Anti-Bacterial Agents/chemistry , Antibodies, Bacterial/chemistry , Antigen-Antibody Complex/metabolism , Cell Wall/chemistry , Complementarity Determining Regions/chemistry , Methicillin-Resistant Staphylococcus aureus/physiology , Staphylococcal Infections/immunology , Teichoic Acids/chemistry , Anti-Bacterial Agents/blood , Antibodies, Bacterial/blood , Antigen-Antibody Complex/genetics , Cell Wall/metabolism , Crystallography, X-Ray/methods , Humans , Immunity, Humoral , Protein Binding , Protein Conformation , Staphylococcal Infections/therapy , Structure-Activity Relationship , Teichoic Acids/metabolism , Transgenes/genetics
8.
Elife ; 72018 07 25.
Article in English | MEDLINE | ID: mdl-30044226

ABSTRACT

Cytokines and interferons initiate intracellular signaling via receptor dimerization and activation of Janus kinases (JAKs). How JAKs structurally respond to changes in receptor conformation induced by ligand binding is not known. Here, we present two crystal structures of the human JAK2 FERM and SH2 domains bound to Leptin receptor (LEPR) and Erythropoietin receptor (EPOR), which identify a novel dimeric conformation for JAK2. This 2:2 JAK2/receptor dimer, observed in both structures, identifies a previously uncharacterized receptor interaction essential to dimer formation that is mediated by a membrane-proximal peptide motif called the 'switch' region. Mutation of the receptor switch region disrupts STAT phosphorylation but does not affect JAK2 binding, indicating that receptor-mediated formation of the JAK2 FERM dimer is required for kinase activation. These data uncover the structural and molecular basis for how a cytokine-bound active receptor dimer brings together two JAK2 molecules to stimulate JAK2 kinase activity.


Subject(s)
Janus Kinase 2/chemistry , Peptide Fragments/chemistry , Protein Conformation , Receptors, Erythropoietin/chemistry , Receptors, Leptin/chemistry , Crystallography, X-Ray , Dimerization , FERM Domains/genetics , Humans , Janus Kinase 2/genetics , Mutation , Peptide Fragments/genetics , Phosphorylation/genetics , Protein Binding/genetics , Receptors, Erythropoietin/genetics , Receptors, Leptin/genetics , STAT Transcription Factors/chemistry , STAT Transcription Factors/genetics , Signal Transduction/genetics , src Homology Domains/genetics
9.
Neuropharmacology ; 121: 204-218, 2017 Jul 15.
Article in English | MEDLINE | ID: mdl-28457974

ABSTRACT

Ionotropic glutamate receptors (iGluRs) mediate fast excitatory neurotransmission and are key nervous system drug targets. While diverse pharmacological tools have yielded insight into iGluR extracellular domain function, less is known about molecular mechanisms underlying the ion conduction gating process within the transmembrane domain (TMD). We have discovered a novel NMDAR positive allosteric modulator (PAM), GNE-9278, with a unique binding site on the extracellular surface of the TMD. Mutation of a single residue near the Lurcher motif on GluN1 M3 can convert GNE-9278 modulation from positive to negative, and replacing three AMPAR pre-M1 residues with corresponding NMDAR residues can confer GNE-9278 sensitivity to AMPARs. Modulation by GNE-9278 is state-dependent and significantly alters extracellular domain pharmacology. The unique properties and structural determinants of GNE-9278 reveal new modulatory potential of the iGluR TMD.


Subject(s)
Receptors, N-Methyl-D-Aspartate/metabolism , Synaptic Transmission/physiology , Allosteric Regulation/drug effects , Allosteric Regulation/genetics , Binding Sites/drug effects , Binding Sites/genetics , Calcium/metabolism , Dose-Response Relationship, Drug , Doxycycline/pharmacology , Electric Stimulation , Excitatory Amino Acid Agents/chemistry , Excitatory Amino Acid Agents/pharmacology , Glutamic Acid/pharmacology , Glycine/metabolism , HEK293 Cells , Humans , Membrane Potentials/drug effects , Membrane Potentials/genetics , Patch-Clamp Techniques , Protein Domains/drug effects , Protein Domains/genetics , Pyrimidinones/chemistry , Pyrimidinones/pharmacology , Receptors, N-Methyl-D-Aspartate/genetics , Sulfonamides/chemistry , Sulfonamides/pharmacology , Synaptic Transmission/drug effects , Synaptic Transmission/genetics , Transfection
10.
Article in English | MEDLINE | ID: mdl-28458652

ABSTRACT

The Janus kinases (JAKs) are non-receptor tyrosine kinases essential for signaling in response to cytokines and interferons and thereby control many essential functions in growth, development, and immune regulation. JAKs are unique among tyrosine kinases for their constitutive yet non-covalent association with class I and II cytokine receptors, which upon cytokine binding bring together two JAKs to create an active signaling complex. JAK association with cytokine receptors is facilitated by N-terminal FERM and SH2 domains, both of which are classical mediators of peptide interactions. Together, the JAK FERM and SH2 domains mediate a bipartite interaction with two distinct receptor peptide motifs, the proline-rich "Box1" and hydrophobic "Box2," which are present in the intracellular domain of cytokine receptors. While the general sidechain chemistry of Box1 and Box2 peptides is conserved between receptors, they share very weak primary sequence homology, making it impossible to posit why certain JAKs preferentially interact with and signal through specific subsets of cytokine receptors. Here, we review the structure and function of the JAK FERM and SH2 domains in light of several recent studies that reveal their atomic structure and elucidate interaction mechanisms with both the Box1 and Box2 receptor motifs. These crystal structures demonstrate how evolution has repurposed the JAK FERM and SH2 domains into a receptor-binding module that facilitates interactions with multiple receptors possessing diverse primary sequences.

11.
ACS Med Chem Lett ; 8(1): 84-89, 2017 Jan 12.
Article in English | MEDLINE | ID: mdl-28105280

ABSTRACT

The N-methyl-d-aspartate receptor (NMDAR) is an ionotropic glutamate receptor, gated by the endogenous coagonists glutamate and glycine, permeable to Ca2+ and Na+. NMDAR dysfunction is associated with numerous neurological and psychiatric disorders, including schizophrenia, depression, and Alzheimer's disease. Recently, we have disclosed GNE-0723 (1), a GluN2A subunit-selective and brain-penetrant positive allosteric modulator (PAM) of NMDARs. This work highlights the discovery of a related pyridopyrimidinone core with distinct structure-activity relationships, despite the structural similarity to GNE-0723. GNE-5729 (13), a pyridopyrimidinone-based NMDAR PAM, was identified with both an improved pharmacokinetic profile and increased selectivity against AMPARs. We also include X-ray structure analysis and modeling to propose hypotheses for the activity and selectivity differences.

12.
Structure ; 24(6): 897-905, 2016 06 07.
Article in English | MEDLINE | ID: mdl-27133025

ABSTRACT

JAK1 is a member of the Janus kinase (JAK) family of non-receptor tyrosine kinases that are activated in response to cytokines and interferons. Here, we present two crystal structures of the human JAK1 FERM and SH2 domains bound to peptides derived from the class II cytokine receptors IFN-λ receptor 1 and IL-10 receptor 1 (IFNLR1 and IL10RA). These structures reveal an interaction site in the JAK1 FERM that accommodates the so-called "box1" membrane-proximal receptor peptide motif. Biophysical analysis of the JAK1-IFNLR1 interaction indicates that the receptor box1 is the primary driver of the JAK1 interaction, and identifies residues conserved among class II receptors as important for binding. In addition, we demonstrate that a second "box2" receptor motif further stabilizes the JAK1-IFNLR1 complex. Together, these data identify a conserved JAK binding site for receptor peptides and elucidate the mechanism by which class II cytokine receptors interact with JAK1.


Subject(s)
Interleukin-10 Receptor alpha Subunit/chemistry , Janus Kinase 1/chemistry , Janus Kinase 1/metabolism , Receptors, Cytokine/chemistry , Amino Acid Motifs , Binding Sites , Crystallography, X-Ray , Humans , Interleukin-10 Receptor alpha Subunit/metabolism , Models, Molecular , Protein Binding , Protein Conformation , Protein Domains , Receptors, Cytokine/metabolism , Receptors, Interferon
13.
J Med Chem ; 59(6): 2760-79, 2016 Mar 24.
Article in English | MEDLINE | ID: mdl-26919761

ABSTRACT

The N-methyl-D-aspartate receptor (NMDAR) is a Na(+) and Ca(2+) permeable ionotropic glutamate receptor that is activated by the coagonists glycine and glutamate. NMDARs are critical to synaptic signaling and plasticity, and their dysfunction has been implicated in a number of neurological disorders, including schizophrenia, depression, and Alzheimer's disease. Herein we describe the discovery of potent GluN2A-selective NMDAR positive allosteric modulators (PAMs) starting from a high-throughput screening hit. Using structure-based design, we sought to increase potency at the GluN2A subtype, while improving selectivity against related α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs). The structure-activity relationship of channel deactivation kinetics was studied using a combination of electrophysiology and protein crystallography. Effective incorporation of these strategies resulted in the discovery of GNE-0723 (46), a highly potent and brain penetrant GluN2A-selective NMDAR PAM suitable for in vivo characterization.


Subject(s)
Excitatory Amino Acid Antagonists/chemical synthesis , Excitatory Amino Acid Antagonists/pharmacology , Receptors, N-Methyl-D-Aspartate/drug effects , Animals , CHO Cells , Calcium/metabolism , Cricetinae , Cricetulus , Crystallography, X-Ray , Drug Discovery , HEK293 Cells , High-Throughput Screening Assays , Humans , Kinetics , Models, Molecular , Patch-Clamp Techniques , Receptors, AMPA/drug effects , Structure-Activity Relationship
14.
Neuron ; 89(5): 983-99, 2016 Mar 02.
Article in English | MEDLINE | ID: mdl-26875626

ABSTRACT

To enhance physiological function of NMDA receptors (NMDARs), we identified positive allosteric modulators (PAMs) of NMDARs with selectivity for GluN2A subunit-containing receptors. X-ray crystallography revealed a binding site at the GluN1-GluN2A dimer interface of the extracellular ligand-binding domains (LBDs). Despite the similarity between the LBDs of NMDARs and AMPA receptors (AMPARs), GluN2A PAMs with good selectivity against AMPARs were identified. Potentiation was observed with recombinant triheteromeric GluN1/GluN2A/GluN2B NMDARs and with synaptically activated NMDARs in brain slices from wild-type (WT), but not GluN2A knockout (KO), mice. Individual GluN2A PAMs exhibited variable degrees of glutamate (Glu) dependence, impact on NMDAR Glu EC50, and slowing of channel deactivation. These distinct PAMs also exhibited differential impacts during synaptic plasticity induction. The identification of a new NMDAR modulatory site and characterization of GluN2A-selective PAMs provide powerful molecular tools to dissect NMDAR function and demonstrate the feasibility of a therapeutically desirable type of NMDAR enhancement.


Subject(s)
Models, Molecular , Nerve Net/physiology , Neurons/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Allosteric Regulation , Animals , Binding Sites/genetics , CHO Cells , Calcium/metabolism , Cricetulus , Crystallography, X-Ray , Excitatory Amino Acid Agents/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , HEK293 Cells , Hippocampus/cytology , Humans , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/drug effects , Receptors, N-Methyl-D-Aspartate/genetics
15.
Proc Natl Acad Sci U S A ; 111(22): 8025-30, 2014 Jun 03.
Article in English | MEDLINE | ID: mdl-24843152

ABSTRACT

Janus kinases (JAKs) are receptor-associated multidomain tyrosine kinases that act downstream of many cytokines and interferons. JAK kinase activity is regulated by the adjacent pseudokinase domain via an unknown mechanism. Here, we report the 2.8-Å structure of the two-domain pseudokinase-kinase module from the JAK family member TYK2 in its autoinhibited form. We find that the pseudokinase and kinase interact near the kinase active site and that most reported mutations in cancer-associated JAK alleles cluster in or near this interface. Mutation of residues near the TYK2 interface that are analogous to those in cancer-associated JAK alleles, including the V617F and "exon 12" JAK2 mutations, results in increased kinase activity in vitro. These data indicate that JAK pseudokinases are autoinhibitory domains that hold the kinase domain inactive until receptor dimerization stimulates transition to an active state.


Subject(s)
Janus Kinase 1/metabolism , Janus Kinase 2/metabolism , Janus Kinase 3/metabolism , Neoplasms/enzymology , Signal Transduction/physiology , TYK2 Kinase/metabolism , Animals , Cell Line , Crystallography, X-Ray , Dimerization , Enzyme Activation/physiology , Humans , Insecta/cytology , Janus Kinase 1/genetics , Janus Kinase 2/genetics , Janus Kinase 3/genetics , Mutation , Neoplasms/genetics , Protein Structure, Tertiary , Structure-Activity Relationship , TYK2 Kinase/chemistry , TYK2 Kinase/genetics
16.
Nat Struct Mol Biol ; 21(5): 443-8, 2014 May.
Article in English | MEDLINE | ID: mdl-24704786

ABSTRACT

Tyrosine kinase 2 (TYK2) is a member of the Janus kinase (JAK) family of nonreceptor tyrosine kinases, which are essential for proper signaling in immune responses and development. Here we present a 2.0-Å-resolution crystal structure of a receptor-binding fragment of human TYK2, encompassing the FERM and SH2 domains, in complex with a so-called 'box2'-containing intracellular peptide motif from the interferon-α receptor chain 1 (IFNAR1). The TYK2-IFNAR1 interface reveals an unexpected receptor-binding mode that mimics a SH2 domain-phosphopeptide interaction, with a glutamate replacing the canonical phosphotyrosine residue. This structure provides the first view, to our knowledge, of a JAK in complex with its cognate receptor and defines the molecular logic through which JAKs have evolved to interact with divergent receptor sequences.


Subject(s)
Interferon-alpha/chemistry , TYK2 Kinase/chemistry , Amino Acid Sequence , Binding Sites , Conserved Sequence , Crystallography, X-Ray , Humans , Interferon-alpha/metabolism , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary , TYK2 Kinase/metabolism
17.
Bioorg Med Chem Lett ; 23(21): 5923-30, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24042009

ABSTRACT

A highly ligand efficient, novel 8-oxo-pyridopyrimidine containing inhibitor of Jak1 and Jak2 isoforms with a pyridone moiety as the hinge-binding motif was discovered. Structure-based design strategies were applied to significantly improve enzyme potency and the polarity of the molecule was adjusted to gain cellular activity. The crystal structures of two representative inhibitors bound to Jak1 were obtained to enable SAR exploration.


Subject(s)
Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Humans , Janus Kinase 1/chemistry , Janus Kinase 1/metabolism , Janus Kinase 2/chemistry , Janus Kinase 2/metabolism , Molecular Docking Simulation , Structure-Activity Relationship
18.
J Biol Chem ; 288(37): 26926-43, 2013 Sep 13.
Article in English | MEDLINE | ID: mdl-23897821

ABSTRACT

Histone deacetylases (HDACs) are critical in the control of gene expression, and dysregulation of their activity has been implicated in a broad range of diseases, including cancer, cardiovascular, and neurological diseases. HDAC inhibitors (HDACi) employing different zinc chelating functionalities such as hydroxamic acids and benzamides have shown promising results in cancer therapy. Although it has also been suggested that HDACi with increased isozyme selectivity and potency may broaden their clinical utility and minimize side effects, the translation of this idea to the clinic remains to be investigated. Moreover, a detailed understanding of how HDACi with different pharmacological properties affect biological functions in vitro and in vivo is still missing. Here, we show that a panel of benzamide-containing HDACi are slow tight-binding inhibitors with long residence times unlike the hydroxamate-containing HDACi vorinostat and trichostatin-A. Characterization of changes in H2BK5 and H4K14 acetylation following HDACi treatment in the neuroblastoma cell line SH-SY5Y revealed that the timing and magnitude of histone acetylation mirrored both the association and dissociation kinetic rates of the inhibitors. In contrast, cell viability and microarray gene expression analysis indicated that cell death induction and changes in transcriptional regulation do not correlate with the dissociation kinetic rates of the HDACi. Therefore, our study suggests that determining how the selective and kinetic inhibition properties of HDACi affect cell function will help to evaluate their therapeutic utility.


Subject(s)
Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Histone Deacetylase Inhibitors/chemistry , Histones/chemistry , Acetylation , Benzamides/chemistry , Binding, Competitive , Cell Line, Tumor , Cell Survival/drug effects , Humans , Hydroxamic Acids/chemistry , Inhibitory Concentration 50 , Kinetics , Oligonucleotide Array Sequence Analysis , Protein Binding , Pyridines/chemistry , Transcription, Genetic , Vorinostat
19.
Bioorg Med Chem Lett ; 23(12): 3592-8, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23642482

ABSTRACT

The identification of a novel fused triazolo-pyrrolopyridine scaffold, optimized derivatives of which display nanomolar inhibition of Janus kinase 1, is described. Prototypical example 3 demonstrated lower cell potency shift, better permeability in cells and higher oral exposure in rat than the corresponding, previously reported, imidazo-pyrrolopyridine analogue 2. Examples 6, 7 and 18 were subsequently identified from an optimization campaign and demonstrated modest selectivity over JAK2, moderate to good oral bioavailability in rat with overall pharmacokinetic profiles comparable to that reported for an approved pan-JAK inhibitor (tofacitinib).


Subject(s)
Janus Kinase 1/antagonists & inhibitors , Pyridines/pharmacology , Animals , Crystallography, X-Ray , Janus Kinase 1/chemistry , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/chemistry , Kinetics , Models, Molecular , Pyridines/chemistry , Pyrroles/chemistry , Pyrroles/pharmacology , Rats
20.
Bioorg Med Chem Lett ; 22(24): 7627-33, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23107482

ABSTRACT

Herein we describe our successful efforts in obtaining C-2 substituted imidazo-pyrrolopyridines with improved JAK1 selectivity relative to JAK2 by targeting an amino acid residue that differs between the two isoforms (JAK1: E966; JAK2: D939). Efforts to improve cellular potency by reducing the polarity of the inhibitors are also detailed. The X-ray crystal structure of a representative inhibitor in complex with the JAK1 enzyme is also disclosed.


Subject(s)
Drug Discovery , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Pyrroles/pharmacology , Animals , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , Imidazoles/chemistry , Janus Kinase 1/metabolism , Janus Kinase 2/metabolism , Male , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry , Pyridines/administration & dosage , Pyridines/chemistry , Pyrroles/administration & dosage , Pyrroles/chemistry , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...