Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
2.
PLoS Genet ; 12(2): e1005885, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26925779

ABSTRACT

Protein tyrosine kinases (PTKs) are a group of closely related enzymes that have evolutionarily diverged from serine/threonine kinases (STKs) to regulate pathways associated with multi-cellularity. Evolutionary divergence of PTKs from STKs has occurred through accumulation of mutations in the active site as well as in the commonly conserved hydrophobic core. While the functional significance of active site variations is well understood, relatively little is known about how hydrophobic core variations contribute to PTK evolutionary divergence. Here, using a combination of statistical sequence comparisons, molecular dynamics simulations, mutational analysis and in vitro thermostability and kinase assays, we investigate the structural and functional significance of key PTK-specific variations in the kinase core. We find that the nature of residues and interactions in the hydrophobic core of PTKs is strikingly different from other protein kinases, and PTK-specific variations in the core contribute to functional divergence by altering the stability and dynamics of the kinase domain. In particular, a functionally critical STK-conserved histidine that stabilizes the regulatory spine in STKs is selectively mutated to an alanine, serine or glutamate in PTKs, and this loss-of-function mutation is accommodated, in part, through compensatory PTK-specific interactions in the core. In particular, a PTK-conserved phenylalanine in the I-helix appears to structurally and functionally compensate for the loss of STK-histidine by interacting with the regulatory spine, which has far-reaching effects on enzyme activity, inhibitor sensing, and stability. We propose that hydrophobic core variations provide a selective advantage during PTK evolution by increasing the conformational flexibility, and therefore the allosteric potential of the kinase domain. Our studies also suggest that Tyrosine Kinase Like kinases such as RAF are intermediates in PTK evolutionary divergence inasmuch as they share features of both PTKs and STKs in the core. Finally, our studies provide an evolutionary framework for identifying and characterizing disease and drug resistance mutations in the kinase core.


Subject(s)
Evolution, Molecular , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/metabolism , Amino Acid Sequence , Aurora Kinase A/chemistry , Aurora Kinase A/genetics , Aurora Kinase A/metabolism , Catalytic Domain , Conserved Sequence , Hydrophobic and Hydrophilic Interactions , Molecular Dynamics Simulation , Molecular Sequence Data , Mutation , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, EphA3 , Structure-Activity Relationship
3.
Bioorg Med Chem ; 26(6): 1167-1173, 2018 03 15.
Article in English | MEDLINE | ID: mdl-28911855

ABSTRACT

Although EGFR is a highly sought-after drug target, inhibitor resistance remains a challenge. As an alternative strategy for kinase inhibition, we sought to explore whether allosteric activation mechanisms could effectively be disrupted. The kinase domain of EGFR forms an atypical asymmetric dimer via head-to-tail interactions and serves as a requisite for kinase activation. The kinase dimer interface is primarily formed by the H-helix derived from one kinase monomer and the small lobe of the second monomer. We hypothesized that a peptide designed to resemble the binding surface of the H-helix may serve as an effective disruptor of EGFR dimerization and activation. A library of constrained peptides was designed to mimic the H-helix of the kinase domain and interface side chains were optimized using molecular modeling. Peptides were constrained using peptide "stapling" to structurally reinforce an alpha-helical conformation. Peptide stapling was demonstrated to notably enhance cell permeation of an H-helix derived peptide termed EHBI2. Using cell-based assays, EHBI2 was further shown to significantly reduce EGFR activity as measured by EGFR phosphorylation and phosphorylation of the downstream signaling substrate Akt. To our knowledge, this is the first H-helix-based compound targeting the asymmetric interface of the kinase domain that can successfully inhibit EGFR activation and signaling. This study presents a novel, alternative targeting site for allosteric inhibition of EGFR.


Subject(s)
ErbB Receptors/metabolism , Peptides/chemistry , Protein Kinase Inhibitors/chemistry , Allosteric Regulation , Cell Line, Tumor , Dimerization , ErbB Receptors/chemistry , Humans , Microscopy, Fluorescence , Peptides/chemical synthesis , Peptides/metabolism , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/metabolism , Protein Structure, Secondary
4.
Biochim Biophys Acta ; 1854(10 Pt B): 1667-75, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25916936

ABSTRACT

Protein kinase activity is regulated not only by direct strategies affecting activity but also by spatial and temporal regulatory mechanisms. Kinase signaling pathways are coordinated by scaffolding proteins that orchestrate the assembly of multi-protein complexes. One family of such scaffolding proteins are the A-kinase anchoring proteins (AKAPs). AKAPs share the commonality of binding cAMP-dependent protein kinase (PKA). In addition, they bind further signaling proteins and kinase substrates and tether such multi-protein complexes to subcellular locations. The A-kinase binding (AKB) domain of AKAPs typically contains a conserved helical motif that interacts directly with the dimerization/docking (D/D) domain of the regulatory subunits of PKA. Based on a pull-down proteomics approach, we identified neurochondrin (neurite-outgrowth promoting protein) as a previously unidentified AKAP. Here, we show that neurochondrin interacts directly with PKA through a novel mechanism that involves two distinct binding regions. In addition, we demonstrate that neurochondrin has strong isoform selectivity towards the RIIα subunit of PKA with nanomolar affinity. This article is part of a Special Issue entitled: Inhibitors of Protein Kinases.


Subject(s)
A Kinase Anchor Proteins/metabolism , Cyclic AMP-Dependent Protein Kinase RIIalpha Subunit/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Nerve Tissue Proteins/metabolism , A Kinase Anchor Proteins/chemistry , Amino Acid Sequence , Binding Sites , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinase RIIalpha Subunit/chemistry , Cyclic AMP-Dependent Protein Kinases/chemistry , Humans , Multiprotein Complexes , Nerve Tissue Proteins/chemistry , Protein Binding , Signal Transduction
5.
Bioorg Med Chem ; 23(12): 2761-6, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25840798

ABSTRACT

The epidermal growth factor receptor (EGFR) dimerization arm is a key feature that stabilizes dimerization of the extracellular receptor, thereby mediating activation of the tyrosine kinase domain. Peptides mimicking this ß-loop feature can disrupt dimer formation and kinase activation, yet these peptides lack structural constraints or contain redox sensitive disulfide bonds which may limit their stability in physiological environments. Selenylsulfide bonds are a promising alternative to disulfide bonds as they maintain much of the same structural and chemical behavior, yet they are inherently less prone to reduction. Herein, we describe the synthesis, stability and activity of selenylsulfide-bridged dimerization arm mimics. The synthesis was accomplished using an Fmoc-based strategy along with C-terminal labeling for improved overall yield. This selenylsulfide-bridged peptide displayed both proteolytic stability and structural stability even under reducing conditions, demonstrating the potential application of the selenylsulfide bond to generate redox stable ß-loop peptides for disruption of protein-protein interactions.


Subject(s)
ErbB Receptors/metabolism , Peptides/chemistry , Peptidomimetics/chemistry , Protein Multimerization/drug effects , Selenium/chemistry , Sulfides/chemistry , Amino Acid Sequence , Animals , Cell Line , Drug Design , ErbB Receptors/chemistry , Humans , Mice , Models, Molecular , Molecular Sequence Data , Peptides/chemical synthesis , Peptides/pharmacology , Peptidomimetics/chemical synthesis , Peptidomimetics/pharmacology , Protein Conformation/drug effects , Protein Interaction Maps/drug effects , Protein Stability , Selenium/pharmacology , Sulfides/chemical synthesis , Sulfides/pharmacology
6.
Pharmacol Ther ; 173: 159-170, 2017 May.
Article in English | MEDLINE | ID: mdl-28185915

ABSTRACT

Kinases are amongst the largest families in the human proteome and serve as critical mediators of a myriad of cell signaling pathways. Since altered kinase activity is implicated in a variety of pathological diseases, kinases have become a prominent class of proteins for targeted inhibition. Although numerous small molecule and antibody-based inhibitors have already received clinical approval, several challenges may still exist with these strategies including resistance, target selection, inhibitor potency and in vivo activity profiles. Constrained peptide inhibitors have emerged as an alternative strategy for kinase inhibition. Distinct from small molecule inhibitors, peptides can provide a large binding surface area that allows them to bind shallow protein surfaces rather than defined pockets within the target protein structure. By including chemical constraints within the peptide sequence, additional benefits can be bestowed onto the peptide scaffold such as improved target affinity and target selectivity, cell permeability and proteolytic resistance. In this review, we highlight examples of diverse chemistries that are being employed to constrain kinase-targeting peptide scaffolds and highlight their application to modulate kinase signaling as well as their potential clinical implications.


Subject(s)
Peptides/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinases/drug effects , Animals , Binding Sites , Drug Design , Humans , Molecular Targeted Therapy , Protein Binding , Protein Kinases/metabolism , Signal Transduction/drug effects
7.
Cancer Biol Ther ; 18(8): 571-583, 2017 Aug 03.
Article in English | MEDLINE | ID: mdl-28692379

ABSTRACT

The oncoprotein c-Myc is often overexpressed in cancer cells, and the stability of this protein has major significance in deciding the fate of a cell. Thus, targeting c-Myc levels is an attractive approach for developing therapeutic agents for cancer treatment. In this study, we report the anti-cancer activity of the macrocyclic peptides [D-Trp]CJ-15,208 (cyclo[Phe-D-Pro-Phe-D-Trp]) and the natural product CJ-15,208 (cyclo[Phe-D-Pro-Phe-Trp]). [D-Trp]CJ-15,208 reduced c-Myc protein levels in prostate cancer cells and decreased cell proliferation with IC50 values ranging from 2.0 to 16 µM in multiple PC cell lines. [D-Trp]CJ-15,208 induced early and late apoptosis in PC-3 cells following 48 hours treatment, and growth arrest in the G2 cell cycle phase following both 24 and 48 hours treatment. Down regulation of c-Myc in PC-3 cells resulted in loss of sensitivity to [D-Trp]CJ-15,208 treatment, while overexpression of c-Myc in HEK-293 cells imparted sensitivity of these cells to [D-Trp]CJ-15,208 treatment. This macrocyclic tetrapeptide also regulated PP2A by reducing the levels of its phosphorylated form which regulates the stability of cellular c-Myc protein. Thus [D-Trp]CJ-15,208 represents a new lead compound for the potential development of an effective treatment of prostate cancer.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Peptides, Cyclic/pharmacology , Prostatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-myc/metabolism , Down-Regulation , HEK293 Cells , Humans , Male , Peptides, Cyclic/chemistry , Peptides, Cyclic/therapeutic use , Phosphorylation , Prostate/cytology , Prostate/pathology , Prostatic Neoplasms/pathology , Stereoisomerism , Tryptophan/chemistry , Tryptophan/pharmacology , Tryptophan/therapeutic use
8.
Cancer Res ; 76(4): 965-73, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26676744

ABSTRACT

Activation of the WASF3 protein by extracellular stimuli promotes actin cytoskeleton reorganization and facilitates cancer cell invasion, whereas WASF3 depletion suppresses invasion and metastasis. In quiescent cells, the interaction between WASF3 and a complex of proteins, including CYFIP1, acts as a conformational restraint to prevent WASF3 activation. Therefore, we took advantage of this endogenous regulatory mechanism to investigate potential sites that disrupt WASF3 function. Here, we show that genetic knockdown of CYFIP1 in cancer cells led to the destabilization of the WASF3 complex, loss of WASF3 function, and suppressed invasion. Based on existing crystallographic data, we developed stapled peptides, referred to as WASF Helix Mimics (WAHM), that target an α-helical interface between WASF3 and CYFIP1. Treatment of highly invasive breast and prostate cancer cells with WAHM inhibitor peptides significantly reduced motility and invasion in vitro. Mechanistic investigations revealed that these inhibitors suppressed the interaction between Rac and the WASF3 complex, which has been shown to promote cell migration. Furthermore, peptide-mediated inhibition of WASF3 also resulted in the dysregulation of known downstream targets such as MMP-9 and KISS1. Finally, we demonstrate that this invasive phenotype is specific to WASF3 as depletion of WASF1 and WASF2, which can also bind to CYFIP1, did not affect invasion. Collectively, our findings suggest that targeting WASF3 function with WAHM peptides could represent a promising therapeutic strategy for preventing tumor invasion and metastasis.


Subject(s)
Wiskott-Aldrich Syndrome Protein Family/genetics , Wiskott-Aldrich Syndrome Protein Family/metabolism , Cell Movement , Humans , Neoplasm Invasiveness , Neoplasm Metastasis , Peptides , Signal Transduction
9.
PLoS One ; 10(3): e0118796, 2015.
Article in English | MEDLINE | ID: mdl-25790232

ABSTRACT

The epidermal growth factor receptor (EGFR) is overexpressed in multiple carcinomas and is the focus of a variety of targeted therapies. Here we report the design of peptide-based compounds that mimic the EGFR dimerization arm and inhibit allosteric activation of EGFR. These peptides are modified to contain a triazolyl bridge between the peptide strands to constrain the EGFR dimerization arm ß-loop. In this study, we demonstrate that these peptides have significantly improved proteolytic stability over the non-modified peptide sequence, and their inhibitory effects are dependent on the number of the methylene units and orientation of the introduced triazolyl bridge. We identified a peptide, EDA2, which downregulates receptor phosphorylation and dimerization and reduces cell viability. This is the first example of a biologically active triazolyl-bridged peptide targeting the EGFR dimerization interface that effectively downregulates EGFR activation.


Subject(s)
Allosteric Regulation/physiology , Carcinoma/metabolism , Ectodysplasins/biosynthesis , ErbB Receptors/chemistry , Models, Molecular , Protein Engineering/methods , Dimerization , Ectodysplasins/metabolism , Humans , Molecular Dynamics Simulation , Triazoles/chemistry
10.
ACS Chem Biol ; 10(6): 1502-10, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-25765284

ABSTRACT

A-Kinase Anchoring Proteins (AKAPs) coordinate complex signaling events by serving as spatiotemporal modulators of cAMP-dependent protein kinase activity in cells. Although AKAPs organize a plethora of diverse pathways, their cellular roles are often elusive due to the dynamic nature of these signaling complexes. AKAPs can interact with the type I or type II PKA holoenzymes by virtue of high-affinity interactions with the R-subunits. As a means to delineate AKAP-mediated PKA signaling in cells, we sought to develop isoform-selective disruptors of AKAP signaling. Here, we report the development of conformationally constrained peptides named RI-STapled Anchoring Disruptors (RI-STADs) that target the docking/dimerization domain of the type 1 regulatory subunit of PKA. These high-affinity peptides are isoform-selective for the RI isoforms, can outcompete binding by the classical AKAP disruptor Ht31, and can selectively displace RIα, but not RIIα, from binding the dual-specific AKAP149 complex. Importantly, these peptides are cell-permeable and disrupt Type I PKA-mediated phosphorylation events in the context of live cells. Hence, RI-STAD peptides are versatile cellular tools to selectively probe anchored type I PKA signaling events.


Subject(s)
A Kinase Anchor Proteins/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinase Type II/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinase Type I/antagonists & inhibitors , Peptides/chemistry , Protein Kinase Inhibitors/chemistry , Protein Subunits/antagonists & inhibitors , A Kinase Anchor Proteins/chemistry , A Kinase Anchor Proteins/metabolism , Amino Acid Sequence , Binding Sites/drug effects , Cell Line, Tumor , Cell Membrane Permeability , Cyclic AMP-Dependent Protein Kinase Type I/chemistry , Cyclic AMP-Dependent Protein Kinase Type I/metabolism , Cyclic AMP-Dependent Protein Kinase Type II/chemistry , Cyclic AMP-Dependent Protein Kinase Type II/metabolism , Humans , Kinetics , Molecular Sequence Data , Peptides/pharmacology , Phosphorylation , Protein Binding/drug effects , Protein Conformation , Protein Kinase Inhibitors/pharmacology , Protein Subunits/chemistry , Protein Subunits/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL