Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters










Publication year range
1.
Bioorg Med Chem Lett ; 89: 129277, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37105490

ABSTRACT

Inhibition of NF-κB inducing kinase (NIK) has been pursued as a promising therapeutic target for autoimmune disorders due to its highly regulated role in key steps of the NF-κB signaling pathway. Previously reported NIK inhibitors from our group were shown to be potent, selective, and efficacious, but had higher human dose projections than desirable for immunology indications. Herein we report the clearance-driven optimization of a NIK inhibitor guided by metabolite identification studies and structure-based drug design. This led to the identification of an azabicyclo[3.1.0]hexanone motif that attenuated in vitro and in vivo clearance while maintaining NIK potency and increasing selectivity over other kinases, resulting in a greater than ten-fold reduction in predicted human dose.


Subject(s)
NF-kappa B , Signal Transduction , Humans , NF-kappa B/metabolism , Half-Life , Drug Design
2.
Nature ; 609(7926): 400-407, 2022 09.
Article in English | MEDLINE | ID: mdl-35768504

ABSTRACT

The RAS-RAF pathway is one of the most commonly dysregulated in human cancers1-3. Despite decades of study, understanding of the molecular mechanisms underlying dimerization and activation4 of the kinase RAF remains limited. Recent structures of inactive RAF monomer5 and active RAF dimer5-8 bound to 14-3-39,10 have revealed the mechanisms by which 14-3-3 stabilizes both RAF conformations via specific phosphoserine residues. Prior to RAF dimerization, the protein phosphatase 1 catalytic subunit (PP1C) must dephosphorylate the N-terminal phosphoserine (NTpS) of RAF11 to relieve inhibition by 14-3-3, although PP1C in isolation lacks intrinsic substrate selectivity. SHOC2 is as an essential scaffolding protein that engages both PP1C and RAS to dephosphorylate RAF NTpS11-13, but the structure of SHOC2 and the architecture of the presumptive SHOC2-PP1C-RAS complex remain unknown. Here we present a cryo-electron microscopy structure of the SHOC2-PP1C-MRAS complex to an overall resolution of 3 Å, revealing a tripartite molecular architecture in which a crescent-shaped SHOC2 acts as a cradle and brings together PP1C and MRAS. Our work demonstrates the GTP dependence of multiple RAS isoforms for complex formation, delineates the RAS-isoform preference for complex assembly, and uncovers how the SHOC2 scaffold and RAS collectively drive specificity of PP1C for RAF NTpS. Our data indicate that disease-relevant mutations affect complex assembly, reveal the simultaneous requirement of two RAS molecules for RAF activation, and establish rational avenues for discovery of new classes of inhibitors to target this pathway.


Subject(s)
Intracellular Signaling Peptides and Proteins , Protein Phosphatase 1 , Signal Transduction , ras Proteins , Cryoelectron Microscopy , Guanosine Triphosphate/metabolism , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Multiprotein Complexes/ultrastructure , Mutation , Phosphoserine , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/ultrastructure , Protein Phosphatase 1/chemistry , Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism , Protein Phosphatase 1/ultrastructure , Substrate Specificity , raf Kinases/metabolism , ras Proteins/chemistry , ras Proteins/genetics , ras Proteins/metabolism , ras Proteins/ultrastructure
3.
Biochemistry ; 59(41): 3982-3992, 2020 10 20.
Article in English | MEDLINE | ID: mdl-32970425

ABSTRACT

The Ras-RAF-MEK-ERK signaling axis, commonly mutated in human cancers, is highly regulated to prevent aberrant signaling in healthy cells. One of the pathway modulators, 14-3-3, a constitutive dimer, induces RAF dimerization and activation by binding to a phosphorylated motif C-terminal to the RAF kinase domain. Recent work has suggested that a C-terminal "DTS" region in BRAF is necessary for this 14-3-3-mediated activation. We show that the catalytic activity and ATP binding affinity of the BRAF:14-3-3 complex is insensitive to the presence or absence of the DTS, while the ATP sites of both BRAF molecules are identical and available for binding. We also present a crystal structure of the apo BRAF:14-3-3 complex showing that the DTS is not required to attain the catalytically active conformation of BRAF. Rather, BRAF dimerization induced by 14-3-3 is the key step in activation, allowing the active BRAF:14-3-3 tetramer to achieve catalytic activity comparable to the constitutively active oncogenic BRAF V600E mutant.


Subject(s)
14-3-3 Proteins/chemistry , 14-3-3 Proteins/metabolism , Proto-Oncogene Proteins B-raf/chemistry , Proto-Oncogene Proteins B-raf/metabolism , Adenosine Triphosphate/metabolism , Catalysis , Humans , Protein Binding , Protein Multimerization , Signal Transduction
4.
Nat Struct Mol Biol ; 27(2): 134-141, 2020 02.
Article in English | MEDLINE | ID: mdl-31988522

ABSTRACT

The RAS-RAF-MEK-ERK signaling axis is frequently activated in human cancers. Physiological concentrations of ATP prevent formation of RAF kinase-domain (RAFKD) dimers that are critical for activity. Here we present a 2.9-Å-resolution crystal structure of human BRAFKD in complex with MEK and the ATP analog AMP-PCP, revealing interactions between BRAF and ATP that induce an inactive, monomeric conformation of BRAFKD. We also determine how 14-3-3 relieves the negative regulatory effect of ATP through a 2.5-Å-resolution crystal structure of the BRAFKD-14-3-3 complex, in which dimeric 14-3-3 enforces a dimeric BRAFKD assembly to increase BRAF activity. Our data suggest that most oncogenic BRAF mutations alter interactions with ATP and counteract the negative effects of ATP binding by lowering the threshold for RAF dimerization and pathway activation. Our study establishes a framework for rationalizing oncogenic BRAF mutations and provides new avenues for improved RAF-inhibitor discovery.


Subject(s)
14-3-3 Proteins/metabolism , Adenosine Triphosphate/metabolism , Proto-Oncogene Proteins B-raf/metabolism , 14-3-3 Proteins/chemistry , Adenosine Triphosphate/analogs & derivatives , Ataxia Telangiectasia Mutated Proteins/chemistry , Ataxia Telangiectasia Mutated Proteins/metabolism , Crystallography, X-Ray , Humans , Molecular Dynamics Simulation , Protein Conformation , Protein Multimerization , Proto-Oncogene Proteins B-raf/chemistry
5.
MAbs ; 12(1): 1692764, 2020.
Article in English | MEDLINE | ID: mdl-31779513

ABSTRACT

Some antibodies exhibit elevated viscosity at high concentrations, making them poorly suited for therapeutic applications requiring administration by injection such as subcutaneous or ocular delivery. Here we studied an anti-IL-13/IL-17 bispecific IgG4 antibody, which has anomalously high viscosity compared to its parent monospecific antibodies. The viscosity of the bispecific IgG4 in solution was decreased by only ~30% in the presence of NaCl, suggesting electrostatic interactions are insufficient to fully explain the drivers of viscosity. Intriguingly, addition of arginine-HCl reduced the viscosity of the bispecific IgG4 by ~50% to its parent IgG level. These data suggest that beyond electrostatics, additional types of interactions such as cation-π and/or π-π may contribute to high viscosity more significantly than previously understood. Molecular dynamics simulations of antibody fragments in the mixed solution of free arginine and explicit water were conducted to identify hotspots involved in self-interactions. Exposed surface aromatic amino acids displayed an increased number of contacts with arginine. Mutagenesis of the majority of aromatic residues pinpointed by molecular dynamics simulations effectively decreased the solution's viscosity when tested experimentally. This mutational method to reduce the viscosity of a bispecific antibody was extended to a monospecific anti-GCGR IgG1 antibody with elevated viscosity. In all cases, point mutants were readily identified that both reduced viscosity and retained antigen-binding affinity. These studies demonstrate a new approach to mitigate high viscosity of some antibodies by mutagenesis of surface-exposed aromatic residues on complementarity-determining regions that may facilitate some clinical applications.


Subject(s)
Antibodies, Bispecific/chemistry , Arginine/chemistry , Complementarity Determining Regions/chemistry , Immunoglobulin G/chemistry , Animals , Humans , Interleukin-13/immunology , Interleukin-17/immunology , Mice , Mutagenesis, Site-Directed , Static Electricity , Viscosity
6.
J Med Chem ; 61(15): 6801-6813, 2018 08 09.
Article in English | MEDLINE | ID: mdl-29940120

ABSTRACT

NF-κB-inducing kinase (NIK) is a protein kinase central to the noncanonical NF-κB pathway downstream from multiple TNF receptor family members, including BAFF, which has been associated with B cell survival and maturation, dendritic cell activation, secondary lymphoid organ development, and bone metabolism. We report herein the discovery of lead chemical series of NIK inhibitors that were identified through a scaffold-hopping strategy using structure-based design. Electronic and steric properties of lead compounds were modified to address glutathione conjugation and amide hydrolysis. These highly potent compounds exhibited selective inhibition of LTßR-dependent p52 translocation and transcription of NF-κB2 related genes. Compound 4f is shown to have a favorable pharmacokinetic profile across species and to inhibit BAFF-induced B cell survival in vitro and reduce splenic marginal zone B cells in vivo.


Subject(s)
Drug Discovery , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Mice , Models, Molecular , Protein Conformation , Protein Serine-Threonine Kinases/chemistry , NF-kappaB-Inducing Kinase
7.
Structure ; 26(5): 767-777.e5, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29706531

ABSTRACT

Receptor-interacting protein kinase 4 (RIPK4) is a highly conserved regulator of epidermal differentiation. Members of the RIPK family possess a common kinase domain as well as unique accessory domains that likely dictate subcellular localization and substrate preferences. Mutations in human RIPK4 manifest as Bartsocas-Papas syndrome (BPS), a genetic disorder characterized by severe craniofacial and limb abnormalities. We describe the structure of the murine Ripk4 (MmRipk4) kinase domain, in ATP- and inhibitor-bound forms. The crystallographic dimer of MmRipk4 is similar to those of RIPK2 and BRAF, and we show that the intact dimeric entity is required for MmRipk4 catalytic activity through a series of engineered mutations and cell-based assays. We also assess the impact of BPS mutations on protein structure and activity to elucidate the molecular origins of the disease.


Subject(s)
Adenosine Triphosphate/metabolism , Mutation , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Animals , Catalytic Domain , Crystallography, X-Ray , Enzyme Activation , Mice , Models, Molecular , Protein Conformation , Protein Multimerization , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins B-raf/chemistry , Receptor-Interacting Protein Serine-Threonine Kinase 2/chemistry
8.
Article in English | MEDLINE | ID: mdl-29440321

ABSTRACT

Despite decades of extensive drug discovery efforts, there are currently no targeted therapies approved to treat KRAS mutant cancers. In this review, we highlight the challenges and opportunities in targeting KRAS mutant tumors through inhibition of mitogen-activated protein kinase (MAPK) signaling with conformation-specific kinase inhibitors. Through structural analysis and mechanistic studies with BRAF and mitogen-activated protein kinase (MEK) inhibitors, we describe how kinase-dependent and -independent functions of MAPK signaling components regulate KRAS-driven tumorigenesis and how these insights can be used to treat RAS mutant cancers with small molecule kinase inhibitors.


Subject(s)
Genes, ras/genetics , MAP Kinase Signaling System/genetics , Mitogen-Activated Protein Kinases/genetics , Mutation/genetics , Neoplasms/genetics , Antineoplastic Agents/therapeutic use , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Humans , MAP Kinase Kinase Kinases/antagonists & inhibitors , Neoplasms/drug therapy , Neoplasms/enzymology , Proto-Oncogene Proteins B-raf/genetics , raf Kinases/antagonists & inhibitors
9.
Structure ; 24(4): 498-499, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-27050685

ABSTRACT

In this issue of Structure, Castañeda et al. (2016b) use multi-disciplinary approaches including NMR techniques, small-angle neutron scattering, and docking to convincingly demonstrate that K27-linked diubiquitin is relatively rigid with unexpected similarity to the conformation of K48-linked diubiquitin bound to the UBA2 domain of hHR23a.


Subject(s)
Models, Molecular , Ubiquitin/chemistry , Magnetic Resonance Spectroscopy , Scattering, Small Angle , Ubiquitins/chemistry
10.
Cancer Cell ; 29(4): 477-493, 2016 Apr 11.
Article in English | MEDLINE | ID: mdl-26996308

ABSTRACT

Activating mutations in protein kinases drive many cancers. While how recurring point mutations affect kinase activity has been described, the effect of in-frame deletions is not well understood. We show that oncogenic deletions within the ß3-αC loop of HER2 and BRAF are analogous to the recurrent EGFR exon 19 deletions. We identify pancreatic carcinomas with BRAF deletions mutually exclusive with KRAS mutations. Crystal structures of BRAF deletions reveal the truncated loop restrains αC in an active "in" conformation, imparting resistance to inhibitors like vemurafenib that bind the αC "out" conformation. Characterization of loop length explains the prevalence of five amino acid deletions in BRAF, EGFR, and HER2 and highlights the importance of this region for kinase activity and inhibitor efficacy.


Subject(s)
Genes, erbB-1 , Genes, erbB-2 , Mutation , Neoplasm Proteins/genetics , Neoplasms/genetics , Proto-Oncogene Proteins B-raf/genetics , Amino Acid Sequence , Amino Acid Substitution , Antineoplastic Agents/pharmacology , Base Pairing/genetics , Conserved Sequence , Dimerization , Drug Resistance, Neoplasm/genetics , Enzyme Activation/genetics , ErbB Receptors/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Neoplasms/enzymology , Protein Conformation , Protein Interaction Mapping , Protein Kinase Inhibitors/pharmacology , Protein Structure, Secondary , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Structure-Activity Relationship
11.
ACS Med Chem Lett ; 6(3): 276-81, 2015 Mar 12.
Article in English | MEDLINE | ID: mdl-25815138

ABSTRACT

A minor structural change to tertiary sulfonamide RORc ligands led to distinct mechanisms of action. Co-crystal structures of two compounds revealed mechanistically consistent protein conformational changes. Optimized phenylsulfonamides were identified as RORc agonists while benzylsulfonamides exhibited potent inverse agonist activity. Compounds behaving as agonists in our biochemical assay also gave rise to an increased production of IL-17 in human PBMCs whereas inverse agonists led to significant suppression of IL-17 under the same assay conditions. The most potent inverse agonist compound showed >180-fold selectivity over the ROR isoforms as well as all other nuclear receptors that were profiled.

12.
Bioorg Med Chem Lett ; 25(6): 1333-7, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25701252

ABSTRACT

SAR studies focused on improving the pharmacokinetic (PK) properties of the previously reported potent and selective Btk inhibitor CGI-1746 (1) resulted in the clinical candidate GDC-0834 (2), which retained the potency and selectivity of CGI-1746, but with much improved PK in preclinical animal models. Structure based design efforts drove this work as modifications to 1 were investigated at both the solvent exposed region as well as 'H3 binding pocket'. However, in vitro metabolic evaluation of 2 revealed a non CYP-mediated metabolic process that was more prevalent in human than preclinical species (mouse, rat, dog, cyno), leading to a high-level of uncertainly in predicting human pharmacokinetics. Due to its promising potency, selectivity, and preclinical efficacy, a single dose IND was filed and 2 was taken in to a single dose phase I trial in healthy volunteers to quickly evaluate the human pharmacokinetics. In human, 2 was found to be highly labile at the exo-cyclic amide bond that links the tetrahydrobenzothiophene moiety to the central aniline ring, resulting in insufficient parent drug exposure. This information informed the back-up program and discovery of improved inhibitors.


Subject(s)
Protein Kinase Inhibitors/chemistry , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidinones/chemistry , Thiophenes/chemistry , Agammaglobulinaemia Tyrosine Kinase , Animals , Benzamides/chemistry , Benzamides/metabolism , Binding Sites , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Bridged Bicyclo Compounds, Heterocyclic/metabolism , Crystallography, X-Ray , Dogs , Half-Life , Humans , Mice , Microsomes, Liver/metabolism , Molecular Dynamics Simulation , Protein Binding , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacokinetics , Protein Structure, Tertiary , Protein-Tyrosine Kinases/metabolism , Pyrimidinones/chemical synthesis , Pyrimidinones/pharmacokinetics , Rats , Structure-Activity Relationship , Thiophenes/chemical synthesis , Thiophenes/pharmacokinetics
13.
ACS Med Chem Lett ; 5(10): 1088-93, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25313317

ABSTRACT

A-1155463, a highly potent and selective BCL-XL inhibitor, was discovered through nuclear magnetic resonance (NMR) fragment screening and structure-based design. This compound is substantially more potent against BCL-XL-dependent cell lines relative to our recently reported inhibitor, WEHI-539, while possessing none of its inherent pharmaceutical liabilities. A-1155463 caused a mechanism-based and reversible thrombocytopenia in mice and inhibited H146 small cell lung cancer xenograft tumor growth in vivo following multiple doses. A-1155463 thus represents an excellent tool molecule for studying BCL-XL biology as well as a productive lead structure for further optimization.

14.
Structure ; 22(9): 1221-1222, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25185824

ABSTRACT

In this issue of Structure, Liu and colleagues report the structure of the TNF superfamily member LIGHT bound to decoy receptor 3 (DcR3). Both LIGHT and DcR3 interact with multiple binding partners. The authors identify a conserved interaction important for affinity as well as additional interactions that can be targeted to introduce selectivity.


Subject(s)
Receptors, Tumor Necrosis Factor, Member 6b/chemistry , Tumor Necrosis Factor Ligand Superfamily Member 14/chemistry , Humans
15.
Cancer Cell ; 26(3): 402-413, 2014 Sep 08.
Article in English | MEDLINE | ID: mdl-25155755

ABSTRACT

Numerous oncogenic mutations occur within the BRAF kinase domain (BRAF(KD)). Here we show that stable BRAF-MEK1 complexes are enriched in BRAF(WT) and KRAS mutant (MT) cells but not in BRAF(MT) cells. The crystal structure of the BRAF(KD) in a complex with MEK1 reveals a face-to-face dimer sensitive to MEK1 phosphorylation but insensitive to BRAF dimerization. Structure-guided studies reveal that oncogenic BRAF mutations function by bypassing the requirement for BRAF dimerization for activity or weakening the interaction with MEK1. Finally, we show that conformation-specific BRAF inhibitors can sequester a dormant BRAF-MEK1 complex resulting in pathway inhibition. Taken together, these findings reveal a regulatory role for BRAF in the MAPK pathway independent of its kinase activity but dependent on interaction with MEK.


Subject(s)
MAP Kinase Kinase 1/chemistry , Proto-Oncogene Proteins B-raf/chemistry , Catalytic Domain , Crystallography, X-Ray , HCT116 Cells , HEK293 Cells , Humans , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/metabolism , Models, Molecular , Mutation, Missense , Point Mutation , Protein Structure, Quaternary , Protein Structure, Secondary , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins p21(ras) , Signal Transduction , ras Proteins/genetics
16.
Bioorg Med Chem Lett ; 24(16): 3891-7, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25017032

ABSTRACT

Using structure-based drug design principles, we identified opportunities to reduce the lipophilicity of our tertiary sulfonamide RORc inverse agonists. The new analogs possessed improved RORc cellular potencies with >77-fold selectivity for RORc over other nuclear receptors in our cell assay suite. The reduction in lipophilicity also led to an increased plasma-protein unbound fraction and improvements in cellular permeability and aqueous solubility.


Subject(s)
Blood Proteins/chemistry , Cell Membrane Permeability/drug effects , Nuclear Receptor Subfamily 1, Group F, Member 3/agonists , Sulfonamides/pharmacology , Animals , Binding Sites/drug effects , Blood Proteins/metabolism , Crystallography, X-Ray , Dogs , Dose-Response Relationship, Drug , Drug Design , Humans , Hydrophobic and Hydrophilic Interactions , Madin Darby Canine Kidney Cells , Models, Molecular , Molecular Structure , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Rats , Solubility , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/chemistry
17.
Bioorg Med Chem Lett ; 23(24): 6604-9, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24239186

ABSTRACT

The structure-activity relationships of T0901317 analogs were explored as RORc inverse agonists using the principles of property- and structure-based drug design. An X-ray co-crystal structure of T0901317 and RORc was obtained and provided molecular insight into why T0901317 functioned as an inverse agonist of RORc; whereas, the same ligand functioned as an agonist of FXR, LXR, and PXR. The structural data was also used to design inhibitors with improved RORc biochemical and cellular activities. The improved inhibitors possessed enhanced selectivity profiles (rationalized using the X-ray crystallographic data) against other nuclear receptors.


Subject(s)
Drug Design , Nuclear Receptor Subfamily 1, Group F, Member 3/agonists , Propanols/chemistry , Sulfonamides/chemistry , Sulfonamides/pharmacology , Binding Sites , Crystallography, X-Ray , Drug Inverse Agonism , Humans , Hydrocarbons, Fluorinated/chemical synthesis , Hydrocarbons, Fluorinated/chemistry , Hydrocarbons, Fluorinated/metabolism , Interferon-gamma/metabolism , Interleukin-17/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Molecular Dynamics Simulation , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Protein Binding/drug effects , Protein Structure, Tertiary , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/metabolism
18.
Proc Natl Acad Sci U S A ; 110(49): 19896-901, 2013 Dec 03.
Article in English | MEDLINE | ID: mdl-24248355

ABSTRACT

Homotrimeric TNF superfamily ligands signal by inducing trimers of their cognate receptors. As a biologically active heterotrimer, Lymphotoxin(LT)α1ß2 is unique in the TNF superfamily. How the three unique potential receptor-binding interfaces in LTα1ß2 trigger signaling via LTß Receptor (LTßR) resulting in lymphoid organogenesis and propagation of inflammatory signals is poorly understood. Here we show that LTα1ß2 possesses two binding sites for LTßR with distinct affinities and that dimerization of LTßR by LTα1ß2 is necessary and sufficient for signal transduction. The crystal structure of a complex formed by LTα1ß2, LTßR, and the fab fragment of an antibody that blocks LTßR activation reveals the lower affinity receptor-binding site. Mutations targeting each potential receptor-binding site in an engineered single-chain variant of LTα1ß2 reveal the high-affinity site. NF-κB reporter assays further validate that disruption of receptor interactions at either site is sufficient to prevent signaling via LTßR.


Subject(s)
Cytokines/chemistry , Lymphotoxin alpha1, beta2 Heterotrimer/metabolism , Lymphotoxin beta Receptor/metabolism , Multiprotein Complexes/immunology , Signal Transduction/immunology , Chromatography, Gel , Cytokines/immunology , Dimerization , Humans , Multiprotein Complexes/metabolism
19.
Science ; 339(6126): 1441-5, 2013 Mar 22.
Article in English | MEDLINE | ID: mdl-23371553

ABSTRACT

Receptor-interacting protein kinase 4 (RIPK4) is required for epidermal differentiation and is mutated in Bartsocas-Papas syndrome. RIPK4 binds to protein kinase C, but its signaling mechanisms are largely unknown. Ectopic RIPK4, but not catalytically inactive or Bartsocas-Papas RIPK4 mutants, induced accumulation of cytosolic ß-catenin and a transcriptional program similar to that caused by Wnt3a. In Xenopus embryos, Ripk4 synergized with coexpressed Xwnt8, whereas Ripk4 morpholinos or catalytic inactive Ripk4 antagonized Wnt signaling. RIPK4 interacted constitutively with the adaptor protein DVL2 and, after Wnt3a stimulation, with the co-receptor LRP6. Phosphorylation of DVL2 by RIPK4 favored canonical Wnt signaling. Wnt-dependent growth of xenografted human tumor cells was suppressed by RIPK4 knockdown, suggesting that RIPK4 overexpression may contribute to the growth of certain tumor types.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Wnt Signaling Pathway , Xenopus Proteins/metabolism , Animals , Cell Line , Cell Line, Tumor , Cytosol/metabolism , Dishevelled Proteins , Female , Gene Knockdown Techniques , HEK293 Cells , Humans , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Neoplasm Transplantation , Neoplasms/metabolism , Ovarian Neoplasms/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Transplantation, Heterologous , Wnt3A Protein/metabolism , Xenopus Proteins/genetics , Xenopus laevis/embryology , Xenopus laevis/metabolism , beta Catenin/metabolism
20.
Nat Med ; 19(2): 202-8, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23291630

ABSTRACT

Proteins in the B cell CLL/lymphoma 2 (BCL-2) family are key regulators of the apoptotic process. This family comprises proapoptotic and prosurvival proteins, and shifting the balance toward the latter is an established mechanism whereby cancer cells evade apoptosis. The therapeutic potential of directly inhibiting prosurvival proteins was unveiled with the development of navitoclax, a selective inhibitor of both BCL-2 and BCL-2-like 1 (BCL-X(L)), which has shown clinical efficacy in some BCL-2-dependent hematological cancers. However, concomitant on-target thrombocytopenia caused by BCL-X(L) inhibition limits the efficacy achievable with this agent. Here we report the re-engineering of navitoclax to create a highly potent, orally bioavailable and BCL-2-selective inhibitor, ABT-199. This compound inhibits the growth of BCL-2-dependent tumors in vivo and spares human platelets. A single dose of ABT-199 in three patients with refractory chronic lymphocytic leukemia resulted in tumor lysis within 24 h. These data indicate that selective pharmacological inhibition of BCL-2 shows promise for the treatment of BCL-2-dependent hematological cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Blood Platelets/drug effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Hematologic Neoplasms/drug therapy , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Sulfonamides/pharmacology , Aniline Compounds/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Survival/drug effects , Dogs , Female , HeLa Cells , Humans , Mice , Mice, SCID , Proto-Oncogene Proteins c-bcl-2/chemistry , Tumor Burden , Xenograft Model Antitumor Assays , bcl-X Protein/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...