Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Article in English | MEDLINE | ID: mdl-35217606

ABSTRACT

Mutations in the gene coding for leucine-rich repeat kinase 2 (LRRK2) are a leading cause of the inherited form of Parkinson's disease (PD), while LRRK2 overactivation is also associated with the more common idiopathic form of PD. LRRK2 is a large multidomain protein, including a GTPase as well as a Ser/Thr protein kinase domain. Common, disease-causing mutations increase LRRK2 kinase activity, presenting LRRK2 as an attractive target for drug discovery. Currently, drug development has mainly focused on ATP-competitive kinase inhibitors. Here, we report the identification and characterization of a variety of nanobodies that bind to different LRRK2 domains and inhibit or activate LRRK2 in cells and in in vitro. Importantly, nanobodies were identified that inhibit LRRK2 kinase activity while binding to a site that is topographically distinct from the active site and thus act through an allosteric inhibitory mechanism that does not involve binding to the ATP pocket or even to the kinase domain. Moreover, while certain nanobodies completely inhibit the LRRK2 kinase activity, we also identified nanobodies that specifically inhibit the phosphorylation of Rab protein substrates. Finally, in contrast to current type I kinase inhibitors, the studied kinase-inhibitory nanobodies did not induce LRRK2 microtubule association. These comprehensively characterized nanobodies represent versatile tools to study the LRRK2 function and mechanism and can pave the way toward novel diagnostic and therapeutic strategies for PD.


Subject(s)
Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Parkinson Disease/metabolism , Single-Domain Antibodies , Adenosine Triphosphate/metabolism , Allosteric Regulation , Animals , Binding Sites , Epitope Mapping , HEK293 Cells , Humans , Mice , Microtubules/metabolism , Phosphorylation , Protein Binding , RAW 264.7 Cells , rab GTP-Binding Proteins/metabolism
2.
J Pept Sci ; 30(5): e3563, 2024 May.
Article in English | MEDLINE | ID: mdl-38135900

ABSTRACT

Familial Parkinson's disease (PD) is frequently linked to multiple disease-causing mutations within Leucine-Rich Repeat Protein Kinase 2 (LRRK2), leading to aberrant kinase activity. Multiple pathogenic effects of enhanced LRRK2 activity have been identified, including loss of cilia and centrosomal cohesion defects. When phosphorylated by LRRK2, Rab8a and Rab10 bind to phospho-specific RILPL effector proteins. RILPL-mediated accumulation of pRabs proximal to the mother centriole is critical for initiating deficits in ciliogenesis and centrosome cohesion mediated by LRRK2. We hypothesized that Rab-derived phospho-mimics may serve to block phosphorylated Rab proteins from docking with RILPL in the context of hyperactive LRRK2 mutants. This would serve as an alternative strategy to downregulate pathogenic signaling mediated by LRRK2, rather than targeting LRRK2 kinase activity itself. To test this theory, we designed a series of constrained peptides mimicking phosphorylated Switch II derived from Rab8. These RILPL interacting peptides, termed RIP, were further shown to permeate cells. Further, several peptides were found to bind RILPL2 and restore ciliogenesis and centrosomal cohesion defects in cells expressing PD-associated mutant LRRK2. This research demonstrates the utility of constrained peptides as downstream inhibitors to target pathogenic LRRK2 activity and may provide an alternative approach to target specific pathways activated by LRRK2.


Subject(s)
Parkinson Disease , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Mutation , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Peptides/metabolism , Phosphorylation , Signal Transduction
3.
EMBO J ; 37(8)2018 04 13.
Article in English | MEDLINE | ID: mdl-29510984

ABSTRACT

Aurora-A regulates the recruitment of TACC3 to the mitotic spindle through a phospho-dependent interaction with clathrin heavy chain (CHC). Here, we describe the structural basis of these interactions, mediated by three motifs in a disordered region of TACC3. A hydrophobic docking motif binds to a previously uncharacterized pocket on Aurora-A that is blocked in most kinases. Abrogation of the docking motif causes a delay in late mitosis, consistent with the cellular distribution of Aurora-A complexes. Phosphorylation of Ser558 engages a conformational switch in a second motif from a disordered state, needed to bind the kinase active site, into a helical conformation. The helix extends into a third, adjacent motif that is recognized by a helical-repeat region of CHC, not a recognized phospho-reader domain. This potentially widespread mechanism of phospho-recognition provides greater flexibility to tune the molecular details of the interaction than canonical recognition motifs that are dominated by phosphate binding.


Subject(s)
Aurora Kinase A/metabolism , Microtubule-Associated Proteins/metabolism , Spindle Apparatus/metabolism , Cell Line , Humans , Microtubule-Associated Proteins/genetics , Protein Conformation, alpha-Helical
4.
Pharmacol Res ; 182: 106302, 2022 08.
Article in English | MEDLINE | ID: mdl-35691539

ABSTRACT

Wiskott-Aldrich syndrome protein family members (WASF) regulate the dynamics of the actin cytoskeleton, which plays an instrumental role in cancer metastasis and invasion. WASF1/2/3 forms a hetero-pentameric complex with CYFIP1/2, NCKAP1/1 L, Abi1/2/3 and BRK1 called the WASF Regulatory Complex (WRC), which cooperatively regulates actin nucleation by WASF1/2/3. Activation of the WRC enables actin networking and provides the mechanical force required for the formation of lamellipodia and invadopodia. Although the WRC drives cell motility essential for several routine physiological functions, its aberrant deployment is observed in cancer metastasis and invasion. WASF3 expression is correlated with metastatic potential in several cancers and inversely correlates with overall progression-free survival. Therefore, disruption of the WRC may serve as a novel strategy for targeting metastasis. Given the complexity involved in the formation of the WRC which is largely comprised of large protein-protein interfaces, there are currently no inhibitors for WASF3. However, several constrained peptide mimics of the various protein-protein interaction interfaces within the WRC were found to successfully disrupt WASF3-mediated migration and invasion. This review explores the role of the WASF3 WRC in driving metastasis and how it may be selectively targeted for suppression of metastasis.


Subject(s)
Actins , Neoplasm Metastasis , Neoplasms , Wiskott-Aldrich Syndrome Protein Family , Actins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Cell Movement/physiology , Cytoskeletal Proteins , Humans , Neoplasm Metastasis/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Wiskott-Aldrich Syndrome Protein Family/metabolism
5.
Traffic ; 20(5): 357-368, 2019 05.
Article in English | MEDLINE | ID: mdl-30941853

ABSTRACT

The classic mode of G protein-coupled receptor (GPCR)-mediated transactivation of the receptor tyrosine kinase epidermal growth factor receptor (EGFR) transactivation occurs via matrix metalloprotease (MMP)-mediated cleavage of plasma membrane-anchored EGFR ligands. Herein, we show that the Gαs-activating GPCR ligands vasoactive intestinal peptide (VIP) and prostaglandin E2 (PGE2 ) transactivate EGFR through increased cell-surface delivery of the EGFR ligand transforming growth factor-α (TGFα) in polarizing madin-darby canine kidney (MDCK) and Caco-2 cells. This is achieved by PKA-mediated phosphorylation of naked cuticle homolog 2 (NKD2), previously shown to bind TGFα and direct delivery of TGFα-containing vesicles to the basolateral surface of polarized epithelial cells. VIP and PGE2 rapidly activate protein kinase A (PKA) that then phosphorylates NKD2 at Ser-223, a process that is facilitated by the molecular scaffold A-kinase anchoring protein 12 (AKAP12). This phosphorylation stabilized NKD2, ensuring efficient cell-surface delivery of TGFα and increased EGFR activation. Thus, GPCR-triggered, PKA/AKAP12/NKD2-regulated targeting of TGFα to the cell surface represents a new mode of EGFR transactivation that occurs proximal to ligand cleavage by MMPs.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Calcium-Binding Proteins/metabolism , Cell Membrane/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Transforming Growth Factor alpha/metabolism , A Kinase Anchor Proteins/metabolism , Animals , Caco-2 Cells , Cell Cycle Proteins/metabolism , Dinoprostone/metabolism , Dogs , ErbB Receptors/metabolism , HEK293 Cells , Humans , Madin Darby Canine Kidney Cells , Protein Transport , Signal Transduction , Vasoactive Intestinal Peptide/metabolism
6.
Nat Methods ; 14(4): 427-434, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28288122

ABSTRACT

Compartmentalized biochemical activities are essential to all cellular processes, but there is no generalizable method to visualize dynamic protein activities in living cells at a resolution commensurate with cellular compartmentalization. Here, we introduce a new class of fluorescent biosensors that detect biochemical activities in living cells at a resolution up to threefold better than the diffraction limit. These 'FLINC' biosensors use binding-induced changes in protein fluorescence dynamics to translate kinase activities or protein-protein interactions into changes in fluorescence fluctuations, which are quantifiable through stochastic optical fluctuation imaging. A protein kinase A (PKA) biosensor allowed us to resolve minute PKA activity microdomains on the plasma membranes of living cells and to uncover the role of clustered anchoring proteins in organizing these activity microdomains. Together, these findings suggest that biochemical activities of the cell are spatially organized into an activity architecture whose structural and functional characteristics can be revealed by these new biosensors.


Subject(s)
Biosensing Techniques/methods , Cyclic AMP-Dependent Protein Kinases/metabolism , Cell Membrane/metabolism , Cyclic AMP-Dependent Protein Kinases/analysis , Escherichia coli/genetics , Fluorescence Resonance Energy Transfer/methods , Fluorescent Dyes , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Microscopy/instrumentation , Microscopy/methods , Molecular Imaging/methods , Mutagenesis, Site-Directed , Protein Interaction Mapping/methods , Stochastic Processes
7.
IUBMB Life ; 72(6): 1103-1114, 2020 06.
Article in English | MEDLINE | ID: mdl-32037730

ABSTRACT

Malaria remains a worldwide health concern with an estimated quarter of a billion people infected and nearly half a million deaths annually. Malaria is caused by a parasite infection from Plasmodium strains which are transmitted from mosquitoes into the human host. Although several small molecule inhibitors have been found to target the early stages of transmission and prevent parasite proliferation, multiple drug resistant parasite strains have emerged and drug resistance remains a major hurdle. As an alternative to small molecule inhibition, several peptide-based therapeutics have been explored for their potential as antimalarial compounds. Chemically constrained peptides or peptidomimetics were developed to target large binding interfaces of parasite-based proteins that have historically been difficult to selectively inhibit using small molecules. Here, we review ongoing research aimed at developing constrained peptides targeting protein-protein interactions pertinent to malaria pathogenesis. These targets include Falcipain-2, the J domain of CDPK1, myosin A tail domain interacting protein, the PKA signaling pathway, and an unclear signaling pathway involving angiotensin-derived peptides. Diverse synthetic methods were also used for each target. Merging parasite biology with synthetic strategies may provide new opportunities to develop alternative methods for uncovering novel antimalarials and may offer an alternate source for targeting drug-resistant parasite strains.


Subject(s)
Antimalarials/pharmacology , Peptides/pharmacology , Peptidomimetics/pharmacology , Plasmodium/drug effects , Angiotensin II/chemistry , Antimalarials/chemistry , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/metabolism , Humans , Peptides/chemistry , Peptidomimetics/chemistry , Plasmodium/metabolism , Protein Conformation , Protein Kinases/metabolism , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism
8.
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
10.
Molecules ; 24(8)2019 Apr 20.
Article in English | MEDLINE | ID: mdl-31009996

ABSTRACT

Kinases regulate multiple and diverse signaling pathways and misregulation is implicated in a multitude of diseases. Although significant efforts have been put forth to develop kinase-specific inhibitors, specificity remains a challenge. As an alternative to catalytic inhibition, allosteric inhibitors can target areas on the surface of an enzyme, thereby providing additional target diversity. Using cAMP-dependent protein kinase A (PKA) as a model system, we sought to develop a hydrocarbon-stapled peptide targeting the pseudosubstrate domain of the kinase. A library of peptides was designed from a Protein Kinase Inhibitor (PKI), a naturally encoded protein that serves as a pseudosubstrate inhibitor for PKA. The binding properties of these peptide analogs were characterized by fluorescence polarization and surface plasmon resonance, and two compounds were identified with KD values in the 500-600 pM range. In kinase activity assays, both compounds demonstrated inhibition with 25-35 nM IC50 values. They were also found to permeate cells and localize within the cytoplasm and inhibited PKA activity within the cellular environment. To the best of our knowledge, these stapled peptide inhibitors represent some of the highest affinity binders reported to date for hydrocarbon stapled peptides.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/chemistry , Peptides/chemistry , Peptides/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Binding Sites , Cell Line , Dose-Response Relationship, Drug , Humans , Kinetics , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding , Structure-Activity Relationship , Substrate Specificity , Surface Plasmon Resonance
11.
Bioorg Med Chem ; 26(6): 1127-1134, 2018 03 15.
Article in English | MEDLINE | ID: mdl-28917447

ABSTRACT

One powerful application of cell penetrating peptides is the delivery into cells of molecules that function as specific competitors or inhibitors of protein-protein interactions. Ablating defined protein-protein interactions is a refined way to explore their contribution to a particular cellular phenotype in a given disease context. Cell-penetrating peptides can be synthetically constrained through various chemical modifications that stabilize a given structural fold with the potential to improve competitive binding to specific targets. Theileria-transformed leukocytes display high PKA activity, but PKA is an enzyme that plays key roles in multiple cellular processes; consequently genetic ablation of kinase activity gives rise to a myriad of confounding phenotypes. By contrast, ablation of a specific kinase-substrate interaction has the potential to give more refined information and we illustrate this here by describing how surgically ablating PKA interactions with BAD gives precise information on the type of glycolysis performed by Theileria-transformed leukocytes. In addition, we provide two other examples of how ablating specific protein-protein interactions in Theileria-infected leukocytes leads to precise phenotypes and argue that constrained penetrating peptides have great therapeutic potential to combat infectious diseases in general.


Subject(s)
Cell-Penetrating Peptides/metabolism , Theileria/pathogenicity , Cell-Penetrating Peptides/chemical synthesis , Cell-Penetrating Peptides/chemistry , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/metabolism , GRB2 Adaptor Protein/chemistry , GRB2 Adaptor Protein/metabolism , Hexokinase/chemistry , Hexokinase/metabolism , Humans , Leukocytes/cytology , Leukocytes/metabolism , Leukocytes/parasitology , Oxidative Phosphorylation , Protein Interaction Maps
12.
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
13.
Bioorg Med Chem ; 26(6): 1174-1178, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29449124

ABSTRACT

Generation of the second messenger molecule cAMP mediates a variety of cellular responses which are essential for critical cellular processes. In response to elevated cAMP levels, cAMP dependent protein kinase (PKA) phosphorylates serine and threonine residues on a wide variety of target substrates. In order to enhance the precision and directionality of these signaling events, PKA is localized to discrete locations within the cell by A-kinase anchoring proteins (AKAPs). The interaction between PKA and AKAPs is mediated via an amphipathic α-helix derived from AKAPs which binds to a stable hydrophobic groove formed in the dimerization/docking (D/D) domain of PKA-R in an isoform-specific fashion. Although numerous AKAP disruptors have previously been identified that can inhibit either RI- or RII-selective AKAPs, no AKAP disruptors have been identified that have isoform specificity for RIα versus RIß or RIIα versus RIIß. As a strategy to identify isoform-specific AKAP inhibitors, a library of chemically stapled protein-protein interaction (PPI) disruptors was developed based on the RII-selective AKAP disruptor, STAD-2. An alanine was substituted at each position in the sequence, and from this library it was possible to delineate the importance of longer aliphatic residues in the formation of a region which complements the hydrophobic cleft formed by the D/D domain. Interestingly, lysine residues that were added to both terminal ends of the peptide sequence to facilitate water solubility appear to contribute to isoform specificity for RIIα over RIIß while having only weak interaction with RI. This work supports current hypotheses on the mechanisms of AKAP binding and highlights the significance of particular residue positions that aid in distinguishing between the RII isoforms and may provide insight into future design of isoform-selective AKAP disruptors.


Subject(s)
A Kinase Anchor Proteins/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Peptides/metabolism , A Kinase Anchor Proteins/antagonists & inhibitors , Amino Acid Sequence , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/genetics , Fluorescence Polarization , Humans , Kinetics , Peptides/chemical synthesis , Peptides/chemistry , Protein Binding , Protein Interaction Maps , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Protein Structure, Secondary , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
14.
J Neurosci ; 36(5): 1660-8, 2016 Feb 03.
Article in English | MEDLINE | ID: mdl-26843647

ABSTRACT

Little is known about intracellular signaling mechanisms that persistently excite neurons in pain pathways. Persistent spontaneous activity (SA) generated in the cell bodies of primary nociceptors within dorsal root ganglia (DRG) has been found to make major contributions to chronic pain in a rat model of spinal cord injury (SCI) (Bedi et al., 2010; Yang et al., 2014). The occurrence of SCI-induced SA in a large fraction of DRG neurons and the persistence of this SA long after dissociation of the neurons provide an opportunity to define intrinsic cell signaling mechanisms that chronically drive SA in pain pathways. The present study demonstrates that SCI-induced SA requires continuing activity of adenylyl cyclase (AC) and cAMP-dependent protein kinase (PKA), as well as a scaffolded complex containing AC5/6, A-kinase anchoring protein 150 (AKAP150), and PKA. SCI caused a small but significant increase in the expression of AKAP150 but not other AKAPs. DRG membranes isolated from SCI animals revealed a novel alteration in the regulation of AC. AC activity stimulated by Ca(2+)-calmodulin increased, while the inhibition of AC activity by Gαi showed an unexpected and dramatic decrease after SCI. Localized enhancement of the activity of AC within scaffolded complexes containing PKA is likely to contribute to chronic pathophysiological consequences of SCI, including pain, that are promoted by persistent hyperactivity in DRG neurons. SIGNIFICANCE STATEMENT: Chronic neuropathic pain is a major clinical problem with poorly understood mechanisms and inadequate treatments. Recent findings indicate that chronic pain in a rat SCI model depends upon hyperactivity in dorsal root ganglia (DRG) neurons. Although cAMP signaling is involved in many forms of neural plasticity, including hypersensitivity of nociceptors in the presence of inflammatory mediators, our finding that continuing cAMP-PKA signaling is required for persistent SA months after SCI and long after isolation of nociceptors is surprising. The dependence of ongoing SA upon AKAP150 and AC5/6 was unknown. The discovery of a dramatic decrease in Gαi inhibition of AC activity after SCI is novel for any physiological system and potentially has broad implications for understanding chronic pain mechanisms.


Subject(s)
Action Potentials/physiology , Adenylyl Cyclases/physiology , Cyclic AMP-Dependent Protein Kinases/physiology , Ganglia, Spinal/enzymology , Nociceptors/physiology , Spinal Cord Injuries/enzymology , A Kinase Anchor Proteins/pharmacology , Action Potentials/drug effects , Animals , Cells, Cultured , Ganglia, Spinal/drug effects , Male , Rats , Spinal Cord Injuries/pathology , Tissue Scaffolds
15.
Biochem J ; 473(13): 1881-94, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27102985

ABSTRACT

A-kinase anchoring proteins (AKAPs) interact with the dimerization/docking (D/D) domains of regulatory subunits of the ubiquitous protein kinase A (PKA). AKAPs tether PKA to defined cellular compartments establishing distinct pools to increase the specificity of PKA signalling. Here, we elucidated the structure of an extended PKA-binding domain of AKAP18ß bound to the D/D domain of the regulatory RIIα subunits of PKA. We identified three hydrophilic anchor points in AKAP18ß outside the core PKA-binding domain, which mediate contacts with the D/D domain. Such anchor points are conserved within AKAPs that bind regulatory RII subunits of PKA. We derived a different set of anchor points in AKAPs binding regulatory RI subunits of PKA. In vitro and cell-based experiments confirm the relevance of these sites for the interaction of RII subunits with AKAP18 and of RI subunits with the RI-specific smAKAP. Thus we report a novel mechanism governing interactions of AKAPs with PKA. The sequence specificity of each AKAP around the anchor points and the requirement of these points for the tight binding of PKA allow the development of selective inhibitors to unequivocally ascribe cellular functions to the AKAP18-PKA and other AKAP-PKA interactions.


Subject(s)
A Kinase Anchor Proteins/chemistry , A Kinase Anchor Proteins/metabolism , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/metabolism , Amino Acid Sequence , Binding Sites , Calorimetry , HEK293 Cells , Humans , Immunoprecipitation , Molecular Sequence Data , Protein Binding , Protein Conformation , Protein Subunits/chemistry , Protein Subunits/metabolism , Signal Transduction , Surface Plasmon Resonance
16.
Biochim Biophys Acta ; 1854(10 Pt B): 1575-85, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25847873

ABSTRACT

Many cellular functions in eukaryotic pathogens are mediated by the cyclic nucleotide binding (CNB) domain, which senses second messengers such as cyclic AMP and cyclic GMP. Although CNB domain-containing proteins have been identified in many pathogenic organisms, an incomplete understanding of how CNB domains in pathogens differ from other eukaryotic hosts has hindered the development of selective inhibitors for CNB domains associated with infectious diseases. Here, we identify and classify CNB domain-containing proteins in eukaryotic genomes to understand the evolutionary basis for CNB domain functional divergence in pathogens. We identify 359 CNB domain-containing proteins in 31 pathogenic organisms and classify them into distinct subfamilies based on sequence similarity within the CNB domain as well as functional domains associated with the CNB domain. Our study reveals novel subfamilies with pathogen-specific variations in the phosphate-binding cassette. Analyzing these variations in light of existing structural and functional data provides new insights into ligand specificity and promiscuity and clues for drug design. This article is part of a Special Issue entitled: Inhibitors of Protein Kinases.


Subject(s)
Evolution, Molecular , Host-Pathogen Interactions/genetics , Protein Kinases/genetics , Protein Structure, Tertiary/genetics , Cyclic AMP/chemistry , Cyclic AMP/genetics , Cyclic GMP/chemistry , Cyclic GMP/genetics , Drug Design , Genome , Humans , Infections/genetics , Infections/pathology , Phylogeny , Protein Binding , Protein Kinases/chemistry , Protein Structure, Tertiary/drug effects , Signal Transduction
17.
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
18.
Chembiochem ; 17(8): 693-697, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26611881

ABSTRACT

A-Kinase anchoring proteins (AKAPs) act as spatial and temporal regulators of protein kinase A (PKA) by localizing PKA along with multiple proteins into discrete signaling complexes. AKAPs interact with the PKA holoenzyme through an α-helix that docks into a groove formed on the dimerization/docking domain of PKA-R in an isoform-dependent fashion. In an effort to understand isoform selectivity at the molecular level, a library of protein-protein interaction (PPI) disruptors was designed to systematically probe the significance of an aromatic residue on the AKAP docking sequence for RI selectivity. The stapled peptide library was designed based on a high affinity, RI-selective disruptor of AKAP binding, RI-STAD-2. Phe, Trp and Leu were all found to maintain RI selectivity, whereas multiple intermediate-sized hydrophobic substitutions at this position either resulted in loss of isoform selectivity (Ile) or a reversal of selectivity (Val). As a limited number of RI-selective sequences are currently known, this study aids in our understanding of isoform selectivity and establishing parameters for discovering additional RI-selective AKAPs.


Subject(s)
A Kinase Anchor Proteins/chemistry , Cyclic AMP-Dependent Protein Kinase Type I/chemistry , Dimerization , Humans , Hydrophobic and Hydrophilic Interactions , Isoenzymes/chemistry , Molecular Docking Simulation , Protein Binding
19.
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
20.
Chembiochem ; 15(18): 2783-7, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25318996

ABSTRACT

Around 300 people from 18 countries took part in the fourth biennial Chemical Biology conference at The European Molecular Biology Laboratory (EMBL) in Heidelberg, from August 20 to 23, 2014. Many advances in the field of chemical biology were presented in talks and poster sessions. Picture: Petra Riedinger (EMBL).


Subject(s)
Computational Biology , Drug Discovery , Computational Biology/methods , Drug Discovery/methods , Germany , Humans , Models, Molecular , Protein Engineering/methods , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL