Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 18 de 18
1.
Neuropharmacology ; 121: 204-218, 2017 Jul 15.
Article En | MEDLINE | ID: mdl-28457974

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.


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
2.
ACS Med Chem Lett ; 8(1): 84-89, 2017 Jan 12.
Article En | MEDLINE | ID: mdl-28105280

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.

3.
Structure ; 24(6): 897-905, 2016 06 07.
Article En | MEDLINE | ID: mdl-27133025

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.


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
4.
J Med Chem ; 59(6): 2760-79, 2016 Mar 24.
Article En | MEDLINE | ID: mdl-26919761

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.


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
5.
Neuron ; 89(5): 983-99, 2016 Mar 02.
Article En | MEDLINE | ID: mdl-26875626

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.


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
6.
Nat Struct Mol Biol ; 21(5): 443-8, 2014 May.
Article En | MEDLINE | ID: mdl-24704786

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.


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
7.
Structure ; 20(10): 1704-14, 2012 Oct 10.
Article En | MEDLINE | ID: mdl-22921830

The NF-κB inducing kinase (NIK) regulates the non-canonical NF-κB pathway downstream of important clinical targets including BAFF, RANKL, and LTß. Despite numerous genetic studies associating dysregulation of this pathway with autoimmune diseases and hematological cancers, detailed molecular characterization of this central signaling node has been lacking. We undertook a systematic cloning and expression effort to generate soluble, well-behaved proteins encompassing the kinase domains of human and murine NIK. Structures of the apo NIK kinase domain from both species reveal an active-like conformation in the absence of phosphorylation. ATP consumption and peptide phosphorylation assays confirm that phosphorylation of NIK does not increase enzymatic activity. Structures of murine NIK bound to inhibitors possessing two different chemotypes reveal conformational flexibility in the gatekeeper residue controlling access to a hydrophobic pocket. Finally, a single amino acid difference affects the ability of some inhibitors to bind murine and human NIK with the same affinity.


Protein Serine-Threonine Kinases/chemistry , Amino Acid Sequence , Amino Acid Substitution , Animals , Catalytic Domain , Conserved Sequence , Crystallography, X-Ray , Humans , Hydrogen Bonding , Kinetics , Mice , Models, Molecular , Molecular Sequence Data , Peptide Fragments/chemistry , Phosphorylation , Protein Kinase Inhibitors/chemistry , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Structure, Secondary , Structural Homology, Protein , NF-kappaB-Inducing Kinase
8.
J Med Chem ; 55(9): 4101-13, 2012 May 10.
Article En | MEDLINE | ID: mdl-22413863

A series of compounds were designed and synthesized as antagonists of cIAP1/2, ML-IAP, and XIAP based on the N-terminus, AVPI, of mature Smac. Compound 1 (GDC-0152) has the best profile of these compounds; it binds to the XIAP BIR3 domain, the BIR domain of ML-IAP, and the BIR3 domains of cIAP1 and cIAP2 with K(i) values of 28, 14, 17, and 43 nM, respectively. These compounds promote degradation of cIAP1, induce activation of caspase-3/7, and lead to decreased viability of breast cancer cells without affecting normal mammary epithelial cells. Compound 1 inhibits tumor growth when dosed orally in the MDA-MB-231 breast cancer xenograft model. Compound 1 was advanced to human clinical trials, and it exhibited linear pharmacokinetics over the dose range (0.049 to 1.48 mg/kg) tested. Mean plasma clearance in humans was 9 ± 3 mL/min/kg, and the volume of distribution was 0.6 ± 0.2 L/kg.


Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Thiadiazoles/chemical synthesis , Thiadiazoles/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Baculoviral IAP Repeat-Containing 3 Protein , Binding, Competitive , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Caspases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Clinical Trials, Phase I as Topic , Female , Humans , Inhibitor of Apoptosis Proteins/metabolism , Male , Thiadiazoles/chemistry , Thiadiazoles/pharmacokinetics , Ubiquitin-Protein Ligases
9.
Science ; 334(6054): 376-80, 2011 Oct 21.
Article En | MEDLINE | ID: mdl-22021857

Inhibitor of apoptosis (IAP) proteins are negative regulators of cell death. IAP family members contain RING domains that impart E3 ubiquitin ligase activity. Binding of endogenous or small-molecule antagonists to select baculovirus IAP repeat (BIR) domains within cellular IAP (cIAP) proteins promotes autoubiquitination and proteasomal degradation and so releases inhibition of apoptosis mediated by cIAP. Although the molecular details of antagonist-BIR domain interactions are well understood, it is not clear how this binding event influences the activity of the RING domain. Here biochemical and structural studies reveal that the unliganded, multidomain cIAP1 sequesters the RING domain within a compact, monomeric structure that prevents RING dimerization. Antagonist binding induces conformational rearrangements that enable RING dimerization and formation of the active E3 ligase.


Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Inhibitor of Apoptosis Proteins/chemistry , Amino Acid Sequence , Animals , Cell Line , Cell Line, Tumor , Cloning, Molecular , Humans , Hydrophobic and Hydrophilic Interactions , Inhibitor of Apoptosis Proteins/metabolism , Mice , Models, Biological , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Proteasome Endopeptidase Complex/metabolism , Protein Conformation , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Structure, Secondary , Scattering, Small Angle , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism , Ubiquitinated Proteins/chemistry , Ubiquitinated Proteins/metabolism , Ubiquitination
10.
J Mol Biol ; 396(1): 166-77, 2010 Feb 12.
Article En | MEDLINE | ID: mdl-19945466

A proliferation-inducing ligand (APRIL) is a member of the tumor necrosis factor (TNF) ligand superfamily and has a proliferative effect on both normal and tumor cells. The TNF family receptors (B-cell maturation antigen (BCMA), transmembrane activator and CAML-interactor (TACI), and BAFF receptor-3 (BR3)) for APRIL and the closely related ligand, B-cell activating factor of the TNF family (BAFF), bind these ligands through a highly conserved six residue DXL motif ((F/Y/W)-D-X-L-(V/T)-(R/G)). Panning peptide phage display libraries led to the identification of several novel classes of APRIL-binding peptides, which could be grouped by their common sequence motifs. Interestingly, only one of these ten classes consisted of peptides containing the DXL motif. Nevertheless, all classes of peptides prevented APRIL, but not BAFF, from binding BCMA, their shared receptor. Synthetic peptides based on selected sequences inhibited APRIL binding to BCMA with IC(50) values of 0.49-27 microM. An X-ray crystallographic structure of APRIL bound to one of the phage-derived peptides showed that the peptide, lacking the DXL motif, was nevertheless bound in the DXL pocket on APRIL. Our results demonstrate that even though a focused, highly conserved motif is required for APRIL-receptor interaction, remarkably, many novel and distinct classes of peptides are also capable of binding APRIL at the ligand receptor interface.


Peptide Library , Peptides/classification , Peptides/isolation & purification , Tumor Necrosis Factor Ligand Superfamily Member 13/antagonists & inhibitors , Alanine/metabolism , Amino Acid Sequence , Animals , B-Cell Maturation Antigen/chemistry , B-Cell Maturation Antigen/metabolism , Immobilized Proteins/metabolism , Mice , Models, Molecular , Molecular Sequence Data , Mutagenesis/genetics , Peptides/chemistry , Protein Binding , Protein Structure, Secondary , Solubility , Tumor Necrosis Factor Ligand Superfamily Member 13/chemistry , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism
11.
J Med Chem ; 51(18): 5522-32, 2008 Sep 25.
Article En | MEDLINE | ID: mdl-18754654

Phosphatidylinositol-3-kinase (PI3K) is an important target in cancer due to the deregulation of the PI3K/ Akt signaling pathway in a wide variety of tumors. A series of thieno[3,2-d]pyrimidine derivatives were prepared and evaluated as inhibitors of PI3 kinase p110alpha. The synthesis, biological activity, and further profiling of these compounds are described. This work resulted in the discovery of 17, GDC-0941, which is a potent, selective, orally bioavailable inhibitor of PI3K and is currently being evaluated in human clinical trials for the treatment of cancer.


Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Indazoles/pharmacology , Neoplasms/drug therapy , Phosphoinositide-3 Kinase Inhibitors , Sulfonamides/pharmacology , Administration, Oral , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Biological Availability , Cell Line, Tumor , Drug Screening Assays, Antitumor , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/therapeutic use , Humans , Indazoles/administration & dosage , Indazoles/pharmacokinetics , Indazoles/therapeutic use , Magnetic Resonance Spectroscopy , Molecular Structure , Sulfonamides/administration & dosage , Sulfonamides/pharmacokinetics , Sulfonamides/therapeutic use
12.
Cell ; 131(4): 669-81, 2007 Nov 16.
Article En | MEDLINE | ID: mdl-18022362

Inhibitor of apoptosis (IAP) proteins are antiapoptotic regulators that block cell death in response to diverse stimuli. They are expressed at elevated levels in human malignancies and are attractive targets for the development of novel cancer therapeutics. Herein, we demonstrate that small-molecule IAP antagonists bind to select baculovirus IAP repeat (BIR) domains resulting in dramatic induction of auto-ubiquitination activity and rapid proteasomal degradation of c-IAPs. The IAP antagonists also induce cell death that is dependent on TNF signaling and de novo protein biosynthesis. Additionally, the c-IAP proteins were found to function as regulators of NF-kappaB signaling. Through their ubiquitin E3 ligase activities c-IAP1 and c-IAP2 promote proteasomal degradation of NIK, the central ser/thr kinase in the noncanonical NF-kappaB pathway.


Apoptosis/physiology , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Inhibitor of Apoptosis Proteins/metabolism , NF-kappa B/metabolism , Polyubiquitin/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Line , Humans , Inhibitor of Apoptosis Proteins/genetics , Mice , Molecular Structure , Neoplasms/metabolism , Neoplasms/pathology , Proteasome Endopeptidase Complex/metabolism , Protein Structure, Tertiary , Signal Transduction/physiology , Ubiquitination
13.
ACS Chem Biol ; 1(8): 525-33, 2006 Sep 19.
Article En | MEDLINE | ID: mdl-17168540

Designed second mitochondrial activator of caspases (Smac) mimetics based on an accessible [7,5]-bicyclic scaffold bind to and antagonize protein interactions involving the inhibitor of apoptosis (IAP) proteins, X-chromosome-linked IAP (XIAP), melanoma IAP (ML-IAP), and c-IAPs 1 and 2 (cIAP1 and cIAP2). The design rationale is based on a combination of phage-panning data, peptide binding studies, and a survey of potential isosteres. The synthesis of two scaffolds is described. These compounds bind the XIAP-baculoviral IAP repeat 3 (BIR3), cIAP1-BIR3, cIAP2-BIR3, and ML-IAP-BIR domains with submicromolar affinities. The most potent Smac mimetic binds the cIAP1-BIR3 and ML-IAP-BIR domains with a K i of 50 nM. The X-ray crystal structure of this compound bound to an ML-IAP/XIAP chimeric BIR domain protein is compared with that of a complex with a phage-derived tetrapeptide, AVPW. The structures show that these compounds bind to the Smac-binding site on ML-IAP with identical hydrogen-bonding patterns and similar hydrophobic interactions. Consistent with the structural data, coimmunoprecipitation experiments demonstrate that the compounds can effectively block Smac interactions with ML-IAP. The compounds are further demonstrated to activate caspase-3 and -7, to reduce cell viability in assays using MDA-MB-231 breast cancer cells and A2058 melanoma cells, and to enhance doxorubicin-induced apoptosis in MDA-MB-231 cells.


Apoptosis/drug effects , Biomimetic Materials/pharmacology , Drug Design , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/chemistry , Neoplasms/metabolism , Neoplasms/pathology , Binding Sites , Biomimetic Materials/chemical synthesis , Biomimetic Materials/chemistry , Caspases/metabolism , Cell Line , Crystallography, X-Ray , Enzyme Activation/drug effects , Humans , Inhibitor of Apoptosis Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Models, Molecular , Neoplasms/genetics , Protein Binding , Protein Structure, Tertiary
14.
J Biol Chem ; 280(8): 7218-27, 2005 Feb 25.
Article En | MEDLINE | ID: mdl-15542592

TACI is a member of the tumor necrosis factor receptor superfamily and serves as a key regulator of B cell function. TACI binds two ligands, APRIL and BAFF, with high affinity and contains two cysteine-rich domains (CRDs) in its extracellular region; in contrast, BCMA and BR3, the other known high affinity receptors for APRIL and BAFF, respectively, contain only a single or partial CRD. However, another form of TACI exists wherein the N-terminal CRD is removed by alternative splicing. We find that this shorter form is capable of ligand-induced cell signaling and that the second CRD alone (TACI_d2) contains full affinity for both ligands. Furthermore, we report the solution structure and alanine-scanning mutagenesis of TACI_d2 along with co-crystal structures of APRIL.TACI_d2 and APRIL.BCMA complexes that together reveal the mechanism by which TACI engages high affinity ligand binding through a single CRD, and we highlight sources of ligand-receptor specificity within the APRIL/BAFF system.


Cysteine , Membrane Proteins/chemistry , Receptors, Tumor Necrosis Factor/chemistry , Tumor Necrosis Factor-alpha/chemistry , Alternative Splicing , Animals , B-Cell Activating Factor , B-Cell Maturation Antigen , Crystallization , Crystallography, X-Ray , Humans , Ligands , Membrane Proteins/genetics , Mice , Mutagenesis , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Structure, Tertiary , Receptors, Tumor Necrosis Factor/genetics , Signal Transduction , Solutions , Transmembrane Activator and CAML Interactor Protein , Tumor Necrosis Factor Ligand Superfamily Member 13
15.
Biochem J ; 385(Pt 1): 11-20, 2005 Jan 01.
Article En | MEDLINE | ID: mdl-15485396

ML-IAP (melanoma inhibitor of apoptosis) is a potent anti-apoptotic protein that is strongly up-regulated in melanoma and confers protection against a variety of pro-apoptotic stimuli. The mechanism by which ML-IAP regulates apoptosis is unclear, although weak inhibition of caspases 3 and 9 has been reported. Here, the binding to and inhibition of caspase 9 by the single BIR (baculovirus IAP repeat) domain of ML-IAP has been investigated and found to be significantly less potent than the ubiquitously expressed XIAP (X-linked IAP). Engineering of the ML-IAP-BIR domain, based on comparisons with the third BIR domain of XIAP, resulted in a chimeric BIR domain that binds to and inhibits caspase 9 significantly better than either ML-IAP-BIR or XIAP-BIR3. Mutational analysis of the ML-IAP-BIR domain demonstrated that similar enhancements in caspase 9 affinity can be achieved with only three amino acid substitutions. However, none of these modifications affected binding of the ML-IAP-BIR domain to the IAP antagonist Smac (second mitochondrial activator of caspases). ML-IAP-BIR was found to bind mature Smac with low nanomolar affinity, similar to that of XIAP-BIR2-BIR3. Correspondingly, increased expression of ML-IAP results in formation of a ML-IAP-Smac complex and disruption of the endogenous interaction between XIAP and mature Smac. These results suggest that ML-IAP might regulate apoptosis by sequestering Smac and preventing it from antagonizing XIAP-mediated inhibition of caspases, rather than by direct inhibition of caspases.


Adaptor Proteins, Signal Transducing/metabolism , Apoptosis , Carrier Proteins/metabolism , Caspase Inhibitors , Cysteine Proteinase Inhibitors/chemistry , Cysteine Proteinase Inhibitors/metabolism , Mitochondrial Proteins/metabolism , Neoplasm Proteins/metabolism , Protein Engineering , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Sequence , Apoptosis Regulatory Proteins , Binding Sites , Carrier Proteins/chemistry , Carrier Proteins/genetics , Caspase 9 , Caspases/metabolism , Cell Line , Cell Line, Tumor , Crystallography, X-Ray , Cysteine Proteinase Inhibitors/genetics , Electron Spin Resonance Spectroscopy , Humans , Inhibitor of Apoptosis Proteins , Intracellular Signaling Peptides and Proteins , Kinetics , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/genetics , Models, Molecular , Molecular Sequence Data , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Protein Binding , Protein Structure, Tertiary , Proteins/chemistry , Proteins/genetics , Proteins/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , X-Linked Inhibitor of Apoptosis Protein
16.
J Mol Biol ; 343(2): 283-90, 2004 Oct 15.
Article En | MEDLINE | ID: mdl-15451660

A proliferation-inducing ligand (APRIL) is a TNF-like cytokine that stimulates tumor cell growth. Within the TNF ligand superfamily, APRIL is most similar to B-cell activation factor (BAFF) with which it shares 30% sequence identity. APRIL binds the receptors B-cell maturation antigen (BCMA) and TACI with high affinity; both of these receptors have also been shown to bind BAFF, although BCMA has significantly higher affinity for APRIL than BAFF. Determination of the crystal structure of APRIL from three crystallization conditions at resolutions of 1.8-2.4A over a pH range from 5.0 to 8.5 reveals a compact trimeric ligand with a backbone fold very similar to that of BAFF (1.1A RMSD over 122 structurally equivalent Calpha atoms), with the exception of differences in the AA' and DE loop regions. Whereas BAFF has been shown to form 20-trimer assemblies under certain conditions, the molecular determinants required for BAFF-like assemblies are absent in the APRIL structure. No crystal packing suggestive of the formation of higher-order assemblies is seen in any of the crystal forms nor does the structure vary significantly between pH 5.0 and 8.5. Modeling of the APRIL-BCMA complex shows the resulting interface is in agreement with mutagenesis data.


Membrane Proteins/chemistry , Protein Structure, Tertiary , Tumor Necrosis Factor-alpha/chemistry , Amino Acid Sequence , Animals , B-Cell Activating Factor , B-Cell Maturation Antigen , Crystallography, X-Ray , Humans , Membrane Proteins/genetics , Mice , Models, Molecular , Molecular Sequence Data , Mutation , Protein Structure, Quaternary , Receptors, Tumor Necrosis Factor/chemistry , Receptors, Tumor Necrosis Factor/genetics , Sequence Alignment , Tumor Necrosis Factor Ligand Superfamily Member 13 , Tumor Necrosis Factor-alpha/genetics
17.
J Biol Chem ; 279(16): 16727-35, 2004 Apr 16.
Article En | MEDLINE | ID: mdl-14764606

B cell maturation antigen (BCMA) is a tumor necrosis factor receptor family member whose physiological role remains unclear. BCMA has been implicated as a receptor for both a proliferation-inducing ligand (APRIL) and B cell-activating factor (BAFF), tumor necrosis factor ligands that bind to multiple tumor necrosis factor receptor and have been reported to play a role in autoimmune disease and cancer. The results presented herein provide a dual perspective analysis of BCMA binding to both APRIL and BAFF. First, we characterized the binding affinity of monomeric BCMA for its ligands; BAFF binding affinity (IC50 = 8 +/- 5 microm) is about 1000-fold reduced compared with the high affinity interaction of APRIL (IC50 = 11 +/- 3 nm). Second, shotgun alanine scanning of BCMA was used to map critical residues for either APRIL or BAFF binding. In addition to a previously described "DXL" motif (Gordon, N. C., Pan, B., Hymowitz, S. G., Yin, J., Kelley, R. F., Cochran, A. G., Yan, M., Dixit, V. M., Fairbrother, W. J., and Starovasnik, M. A. (2003) Biochemistry 42, 5977-5983), the alanine scanning results predicted four amino acid positions in BCMA (Tyr13, Ile22, Gln25, and Arg27) that could impart ligand specificity. Substitution of Tyr13 was tolerated for BAFF binding but not APRIL binding. Arg27 was required for high affinity binding to APRIL, whereas substitutions of this residue had minimal effect on affinity for BAFF. Further phage display experiments suggested the single mutations of I22K, Q25D, and R27Y as providing the greatest difference in APRIL versus BAFF binding affinity. Incorporation of the Q25D and R27Y substitutions into BCMA produced a dual specificity variant, since it has comparable binding affinity for both APRIL and BAFF, IC50 = 350 and 700 nm, respectively. Binding of the I22K mutant of monomeric BCMA to BAFF was undetectable (IC50 > 100 microm), but affinity for binding to APRIL was similar to wild-type BCMA. Based on these results, a BCMA-Fc fusion with the single I22K mutation was produced that binds APRIL, IC50 = 12 nm, and has no measurable affinity for BAFF. These results suggest that APRIL is the preferred ligand for BCMA and show that specificity can be further modified through amino acid substitutions.


Membrane Proteins/metabolism , Protein Engineering , Receptors, Tumor Necrosis Factor , Tumor Necrosis Factor-alpha/metabolism , Amino Acid Substitution , B-Cell Activating Factor , B-Cell Maturation Antigen , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Binding Sites/genetics , Humans , Ligands , Membrane Proteins/immunology , Mutation , Peptide Mapping , Protein Binding , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/immunology , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 13 , Tumor Necrosis Factor-alpha/immunology
18.
Structure ; 11(12): 1513-20, 2003 Dec.
Article En | MEDLINE | ID: mdl-14656435

EDA is a tumor necrosis factor family member involved in ectodermal development. Splice variants EDA-A1 and EDA-A2 differ only by the presence of Glu 308 and Val 309 in the expected receptor binding region of EDA-A1 but not EDA-A2. This two amino acid difference functions as a switch controlling receptor specificity. EDA-A1 binds only to EDAR, while EDA-A2 is specific for XEDAR. In order to understand the structural basis of this switch, we determined the X-ray crystal structures of the TNF domain of both EDA-A1 and EDA-A2 at 2.3 A and 2.2 A, respectively. While the backbone conformation around the splice difference is similar in both isoforms, the conformation of the following loop, the surface charge, and the shape of the expected receptor binding site differ significantly.


Alternative Splicing , Membrane Proteins/chemistry , Binding Sites , Crystallography, X-Ray , Ectodysplasins , Escherichia coli/metabolism , Humans , Ligands , Membrane Proteins/genetics , Models, Biological , Models, Molecular , Mutation , Protein Binding , Protein Conformation , Protein Isoforms , Protein Structure, Secondary , Protein Structure, Tertiary , Static Electricity
...