Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Elife ; 82019 05 07.
Article in English | MEDLINE | ID: mdl-31063128

ABSTRACT

Fibrolamellar carcinoma (FLC) is a rare liver cancer. FLCs uniquely produce DNAJ-PKAc, a chimeric enzyme consisting of a chaperonin-binding domain fused to the Cα subunit of protein kinase A. Biochemical analyses of clinical samples reveal that a unique property of this fusion enzyme is the ability to recruit heat shock protein 70 (Hsp70). This cellular chaperonin is frequently up-regulated in cancers. Gene-editing of mouse hepatocytes generated disease-relevant AML12DNAJ-PKAc cell lines. Further analyses indicate that the proto-oncogene A-kinase anchoring protein-Lbc is up-regulated in FLC and functions to cluster DNAJ-PKAc/Hsp70 sub-complexes with a RAF-MEK-ERK kinase module. Drug screening reveals Hsp70 and MEK inhibitor combinations that selectively block proliferation of AML12DNAJ-PKAc cells. Phosphoproteomic profiling demonstrates that DNAJ-PKAc biases the signaling landscape toward ERK activation and engages downstream kinase cascades. Thus, the oncogenic action of DNAJ-PKAc involves an acquired scaffolding function that permits recruitment of Hsp70 and mobilization of local ERK signaling.


Subject(s)
Carcinoma, Hepatocellular/physiopathology , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/metabolism , Fetal Proteins/metabolism , Liver Neoplasms/physiopathology , Molecular Chaperones/metabolism , Recombinant Fusion Proteins/metabolism , A Kinase Anchor Proteins/metabolism , Animals , Cell Line , Cell Proliferation , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/genetics , Fetal Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Hepatocytes/pathology , Humans , Mice , Models, Theoretical , Molecular Chaperones/genetics , Protein Binding , Proto-Oncogene Mas , Recombinant Fusion Proteins/genetics , Signal Transduction
2.
J Biol Chem ; 294(9): 3152-3168, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30598507

ABSTRACT

Breast cancer screening and new precision therapies have led to improved patient outcomes. Yet, a positive prognosis is less certain when primary tumors metastasize. Metastasis requires a coordinated program of cellular changes that promote increased survival, migration, and energy consumption. These pathways converge on mitochondrial function, where distinct signaling networks of kinases, phosphatases, and metabolic enzymes regulate these processes. The protein kinase A-anchoring protein dAKAP1 compartmentalizes protein kinase A (PKA) and other signaling enzymes at the outer mitochondrial membrane and thereby controls mitochondrial function and dynamics. Modulation of these processes occurs in part through regulation of dynamin-related protein 1 (Drp1). Here, we report an inverse relationship between the expression of dAKAP1 and mesenchymal markers in breast cancer. Molecular, cellular, and in silico analyses of breast cancer cell lines confirmed that dAKAP1 depletion is associated with impaired mitochondrial function and dynamics, as well as with increased glycolytic potential and invasiveness. Furthermore, disruption of dAKAP1-PKA complexes affected cell motility and mitochondrial movement toward the leading edge in invasive breast cancer cells. We therefore propose that depletion of dAKAP1-PKA "signaling islands" from the outer mitochondrial membrane augments progression toward metastatic breast cancer.


Subject(s)
A Kinase Anchor Proteins/metabolism , Breast Neoplasms/pathology , Cell Movement , Mitochondrial Membranes/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Mesoderm/metabolism , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Dynamics , Neoplasm Invasiveness
3.
Proc Natl Acad Sci U S A ; 115(49): E11465-E11474, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30455320

ABSTRACT

A-kinase anchoring proteins (AKAPs) shape second-messenger signaling responses by constraining protein kinase A (PKA) at precise intracellular locations. A defining feature of AKAPs is a helical region that binds to regulatory subunits (RII) of PKA. Mining patient-derived databases has identified 42 nonsynonymous SNPs in the PKA-anchoring helices of five AKAPs. Solid-phase RII binding assays confirmed that 21 of these amino acid substitutions disrupt PKA anchoring. The most deleterious side-chain modifications are situated toward C-termini of AKAP helices. More extensive analysis was conducted on a valine-to-methionine variant in the PKA-anchoring helix of AKAP18. Molecular modeling indicates that additional density provided by methionine at position 282 in the AKAP18γ isoform deflects the pitch of the helical anchoring surface outward by 6.6°. Fluorescence polarization measurements show that this subtle topological change reduces RII-binding affinity 8.8-fold and impairs cAMP responsive potentiation of L-type Ca2+ currents in situ. Live-cell imaging of AKAP18γ V282M-GFP adducts led to the unexpected discovery that loss of PKA anchoring promotes nuclear accumulation of this polymorphic variant. Targeting proceeds via a mechanism whereby association with the PKA holoenzyme masks a polybasic nuclear localization signal on the anchoring protein. This led to the discovery of AKAP18ε: an exclusively nuclear isoform that lacks a PKA-anchoring helix. Enzyme-mediated proximity-proteomics reveal that compartment-selective variants of AKAP18 associate with distinct binding partners. Thus, naturally occurring PKA-anchoring-defective AKAP variants not only perturb dissemination of local second-messenger responses, but also may influence the intracellular distribution of certain AKAP18 isoforms.


Subject(s)
A Kinase Anchor Proteins/metabolism , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/genetics , Membrane Proteins/metabolism , A Kinase Anchor Proteins/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation, Enzymologic , Genome-Wide Association Study , Humans , Membrane Proteins/genetics , Models, Molecular , Polymorphism, Single Nucleotide , Protein Binding , Protein Conformation , Protein Isoforms , Protein Transport
4.
Elife ; 62017 10 02.
Article in English | MEDLINE | ID: mdl-28967377

ABSTRACT

Scaffolding the calcium/calmodulin-dependent phosphatase 2B (PP2B, calcineurin) focuses and insulates termination of local second messenger responses. Conformational flexibility in regions of intrinsic disorder within A-kinase anchoring protein 79 (AKAP79) delineates PP2B access to phosphoproteins. Structural analysis by negative-stain electron microscopy (EM) reveals an ensemble of dormant AKAP79-PP2B configurations varying in particle length from 160 to 240 Å. A short-linear interaction motif between residues 337-343 of AKAP79 is the sole PP2B-anchoring determinant sustaining these diverse topologies. Activation with Ca2+/calmodulin engages additional interactive surfaces and condenses these conformational variants into a uniform population with mean length 178 ± 17 Å. This includes a Leu-Lys-Ile-Pro sequence (residues 125-128 of AKAP79) that occupies a binding pocket on PP2B utilized by the immunosuppressive drug cyclosporin. Live-cell imaging with fluorescent activity-sensors infers that this region fine-tunes calcium responsiveness and drug sensitivity of the anchored phosphatase.


Subject(s)
A Kinase Anchor Proteins/chemistry , A Kinase Anchor Proteins/metabolism , Calcineurin/chemistry , Calcineurin/metabolism , Calcium/metabolism , Calmodulin/metabolism , Humans , Microscopy, Electron , Protein Binding , Protein Conformation , Protein Interaction Maps
5.
Science ; 356(6344): 1288-1293, 2017 06 23.
Article in English | MEDLINE | ID: mdl-28642438

ABSTRACT

Hormones can transmit signals through adenosine 3',5'-monophosphate (cAMP) to precise intracellular locations. The fidelity of these responses relies on the activation of localized protein kinase A (PKA) holoenzymes. Association of PKA regulatory type II (RII) subunits with A-kinase-anchoring proteins (AKAPs) confers location, and catalytic (C) subunits phosphorylate substrates. Single-particle electron microscopy demonstrated that AKAP79 constrains RII-C subassemblies within 150 to 250 angstroms of its targets. Native mass spectrometry established that these macromolecular assemblies incorporated stoichiometric amounts of cAMP. Chemical-biology- and live cell-imaging techniques revealed that catalytically active PKA holoenzymes remained intact within the cytoplasm. These findings indicate that the parameters of anchored PKA holoenzyme action are much more restricted than originally anticipated.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Holoenzymes/metabolism , Signal Transduction , A Kinase Anchor Proteins/metabolism , Animals , Cell Line, Tumor , Cyclic AMP/chemistry , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/genetics , Holoenzymes/chemistry , Humans , Mice , Microscopy, Electron , Mitochondria/enzymology , Phosphorylation , Protein Binding , Protein Stability , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
6.
Proc Natl Acad Sci U S A ; 113(30): E4328-37, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27402760

ABSTRACT

Filtration through the kidney eliminates toxins, manages electrolyte balance, and controls water homeostasis. Reabsorption of water from the luminal fluid of the nephron occurs through aquaporin-2 (AQP2) water pores in principal cells that line the kidney-collecting duct. This vital process is impeded by formation of an "actin barrier" that obstructs the passive transit of AQP2 to the plasma membrane. Bidirectional control of AQP2 trafficking is managed by hormones and signaling enzymes. We have discovered that vasopressin-independent facets of this homeostatic mechanism are under the control of A-Kinase Anchoring Protein 220 (AKAP220; product of the Akap11 gene). CRISPR/Cas9 gene editing and imaging approaches show that loss of AKAP220 disrupts apical actin networks in organoid cultures. Similar defects are evident in tissue sections from AKAP220-KO mice. Biochemical analysis of AKAP220-null kidney extracts detected reduced levels of active RhoA GTPase, a well-known modulator of the actin cytoskeleton. Fluorescent imaging of kidney sections from these genetically modified mice revealed that RhoA and AQP2 accumulate at the apical surface of the collecting duct. Consequently, these animals are unable to appropriately dilute urine in response to overhydration. We propose that membrane-proximal signaling complexes constrained by AKAP220 impact the actin barrier dynamics and AQP2 trafficking to ensure water homeostasis.


Subject(s)
A Kinase Anchor Proteins/metabolism , Actins/metabolism , Aquaporin 2/metabolism , Kidney/metabolism , Renal Reabsorption , A Kinase Anchor Proteins/genetics , Animals , Female , Homeostasis , Kidney Tubules, Collecting/metabolism , Male , Mice, Knockout , Organ Culture Techniques , Water/metabolism , rhoA GTP-Binding Protein/metabolism
7.
Cell Signal ; 28(7): 733-40, 2016 07.
Article in English | MEDLINE | ID: mdl-26724383

ABSTRACT

The Ca(2+)-responsive phosphatase calcineurin/protein phosphatase 2B dephosphorylates the transcription factor NFATc3. In the myocardium activation of NFATc3 down-regulates the expression of voltage-gated K(+) (Kv) channels after myocardial infarction (MI). This prolongs action potential duration and increases the probability of arrhythmias. Although recent studies infer that calcineurin is activated by local and transient Ca(2+) signals the molecular mechanism that underlies the process is unclear in ventricular myocytes. Here we test the hypothesis that sequestering of calcineurin to the sarcolemma of ventricular myocytes by the anchoring protein AKAP150 is required for acute activation of NFATc3 and the concomitant down-regulation of Kv channels following MI. Biochemical and cell based measurements resolve that approximately 0.2% of the total calcineurin activity in cardiomyocytes is associated with AKAP150. Electrophysiological analyses establish that formation of this AKAP150-calcineurin signaling dyad is essential for the activation of the phosphatase and the subsequent down-regulation of Kv channel currents following MI. Thus AKAP150-mediated targeting of calcineurin to sarcolemmal micro-domains in ventricular myocytes contributes to the local and acute gene remodeling events that lead to the down-regulation of Kv currents.


Subject(s)
A Kinase Anchor Proteins/metabolism , Calcineurin/metabolism , Down-Regulation , Heart Ventricles/pathology , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , NFATC Transcription Factors/metabolism , Potassium Channels, Voltage-Gated/metabolism , Aging , Animals , Animals, Newborn , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Down-Regulation/drug effects , Gene Expression Regulation/drug effects , Mice , Myocardial Infarction/pathology , Myocytes, Cardiac/drug effects , Phenylephrine/pharmacology , Protein Transport/drug effects
8.
Elife ; 4: e09384, 2015 Sep 25.
Article in English | MEDLINE | ID: mdl-26406118

ABSTRACT

Correct orientation of the mitotic spindle in stem cells underlies organogenesis. Spindle abnormalities correlate with cancer progression in germ line-derived tumors. We discover a macromolecular complex between the scaffolding protein Gravin/AKAP12 and the mitotic kinases, Aurora A and Plk1, that is down regulated in human seminoma. Depletion of Gravin correlates with an increased mitotic index and disorganization of seminiferous tubules. Biochemical, super-resolution imaging, and enzymology approaches establish that this Gravin scaffold accumulates at the mother spindle pole during metaphase. Manipulating elements of the Gravin-Aurora A-Plk1 axis prompts mitotic delay and prevents appropriate assembly of astral microtubules to promote spindle misorientation. These pathological responses are conserved in seminiferous tubules from Gravin(-/-) mice where an overabundance of Oct3/4 positive germ line stem cells displays randomized orientation of mitotic spindles. Thus, we propose that Gravin-mediated recruitment of Aurora A and Plk1 to the mother (oldest) spindle pole contributes to the fidelity of symmetric cell division.


Subject(s)
A Kinase Anchor Proteins/metabolism , Aurora Kinase A/metabolism , Cell Cycle Proteins/metabolism , Cell Division , Germ Cells/physiology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Spindle Apparatus/metabolism , Stem Cells/physiology , Animals , Humans , Mice , Mice, Knockout , Seminoma/pathology , Polo-Like Kinase 1
9.
J Biol Chem ; 290(32): 19445-57, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-26088133

ABSTRACT

The proximity of an enzyme to its substrate can influence rate and magnitude of catalysis. A-kinase anchoring protein 220 (AKAP220) is a multivalent anchoring protein that can sequester a variety of signal transduction enzymes. These include protein kinase A (PKA) and glycogen synthase kinase 3ß (GSK3ß). Using a combination of molecular and cellular approaches we show that GSK3ß phosphorylation of Thr-1132 on AKAP220 initiates recruitment of this kinase into the enzyme scaffold. We also find that AKAP220 anchors GSK3ß and its substrate ß-catenin in membrane ruffles. Interestingly, GSK3ß can be released from the multienzyme complex in response to PKA phosphorylation on serine 9, which suppresses GSK3ß activity. The signaling scaffold may enhance this regulatory mechanism, as AKAP220 has the capacity to anchor two PKA holoenzymes. Site 1 on AKAP220 (residues 610-623) preferentially interacts with RII, whereas site 2 (residues 1633-1646) exhibits a dual specificity for RI and RII. In vitro affinity measurements revealed that site 2 on AKAP220 binds RII with ∼10-fold higher affinity than site 1. Occupancy of both R subunit binding sites on AKAP220 could provide a mechanism to amplify local cAMP responses and enable cross-talk between PKA and GSK3ß.


Subject(s)
A Kinase Anchor Proteins/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Epithelial Cells/enzymology , Glycogen Synthase Kinase 3/metabolism , Protein Subunits/metabolism , A Kinase Anchor Proteins/genetics , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Cyclic AMP-Dependent Protein Kinases/genetics , Epithelial Cells/cytology , Gene Expression Regulation/drug effects , Genetic Engineering , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , HEK293 Cells , Holoenzymes/genetics , Holoenzymes/metabolism , Humans , Mice , Molecular Sequence Data , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Protein Subunits/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Signal Transduction
10.
J Biol Chem ; 290(22): 14107-19, 2015 May 29.
Article in English | MEDLINE | ID: mdl-25882844

ABSTRACT

Anchoring proteins direct protein kinases and phosphoprotein phosphatases toward selected substrates to control the efficacy, context, and duration of neuronal phosphorylation events. The A-kinase anchoring protein AKAP79/150 interacts with protein kinase A (PKA), protein kinase C (PKC), and protein phosphatase 2B (calcineurin) to modulate second messenger signaling events. In a mass spectrometry-based screen for additional AKAP79/150 binding partners, we have identified the Roundabout axonal guidance receptor Robo2 and its ligands Slit2 and Slit3. Biochemical and cellular approaches confirm that a linear sequence located in the cytoplasmic tail of Robo2 (residues 991-1070) interfaces directly with sites on the anchoring protein. Parallel studies show that AKAP79/150 interacts with the Robo3 receptor in a similar manner. Immunofluorescent staining detects overlapping expression patterns for murine AKAP150, Robo2, and Robo3 in a variety of brain regions, including hippocampal region CA1 and the islands of Calleja. In vitro kinase assays, peptide spot array mapping, and proximity ligation assay staining approaches establish that human AKAP79-anchored PKC selectively phosphorylates the Robo3.1 receptor subtype on serine 1330. These findings imply that anchored PKC locally modulates the phosphorylation status of Robo3.1 in brain regions governing learning and memory and reward.


Subject(s)
A Kinase Anchor Proteins/metabolism , Protein Kinase C/metabolism , Receptors, Immunologic/metabolism , Animals , Brain/metabolism , Cytoplasm/metabolism , Gene Silencing , Glutathione Transferase/metabolism , HEK293 Cells , Hippocampus/metabolism , Humans , Ligands , Macromolecular Substances , Mass Spectrometry , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Microscopy, Fluorescence , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Phosphorylation , Protein Interaction Mapping , Protein Isoforms , RNA, Small Interfering/metabolism , Receptors, Cell Surface , Signal Transduction
11.
Nat Rev Mol Cell Biol ; 16(4): 232-44, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25785716

ABSTRACT

Cellular responses to environmental cues involve the mobilization of GTPases, protein kinases and phosphoprotein phosphatases. The spatial organization of these signalling enzymes by scaffold proteins helps to guide the flow of molecular information. Allosteric modulation of scaffolded enzymes can alter their catalytic activity or sensitivity to second messengers in a manner that augments, insulates or terminates local cellular events. This Review examines the features of scaffold proteins and highlights examples of locally organized groups of signalling enzymes that drive essential physiological processes, including hormone action, heart rate, cell division, organelle movement and synaptic transmission.


Subject(s)
Cell Physiological Phenomena , Nuclear Matrix-Associated Proteins/metabolism , Signal Transduction , Animals , Cells/enzymology , Genes, Switch/genetics , Humans , Models, Biological
12.
Elife ; 2: e01319, 2013 Nov 05.
Article in English | MEDLINE | ID: mdl-24192038

ABSTRACT

Anchoring proteins sequester kinases with their substrates to locally disseminate intracellular signals and avert indiscriminate transmission of these responses throughout the cell. Mechanistic understanding of this process is hampered by limited structural information on these macromolecular complexes. A-kinase anchoring proteins (AKAPs) spatially constrain phosphorylation by cAMP-dependent protein kinases (PKA). Electron microscopy and three-dimensional reconstructions of type-II PKA-AKAP18γ complexes reveal hetero-pentameric assemblies that adopt a range of flexible tripartite configurations. Intrinsically disordered regions within each PKA regulatory subunit impart the molecular plasticity that affords an ∼16 nanometer radius of motion to the associated catalytic subunits. Manipulating flexibility within the PKA holoenzyme augmented basal and cAMP responsive phosphorylation of AKAP-associated substrates. Cell-based analyses suggest that the catalytic subunit remains within type-II PKA-AKAP18γ complexes upon cAMP elevation. We propose that the dynamic movement of kinase sub-structures, in concert with the static AKAP-regulatory subunit interface, generates a solid-state signaling microenvironment for substrate phosphorylation. DOI: http://dx.doi.org/10.7554/eLife.01319.001.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Chromatography, Gel , Microscopy, Electron , Phosphorylation , Substrate Specificity
13.
J Clin Invest ; 123(7): 3014-24, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23934128

ABSTRACT

Clearance of invading pathogens is essential to preventing overwhelming inflammation and sepsis that are symptomatic of bacterial peritonitis. Macrophages participate in this innate immune response by engulfing and digesting pathogens, a process called phagocytosis. Oxidized phospholipids (OxPL) are danger-associated molecular patterns (DAMPs) generated in response to infection that can prevent the phagocytic clearance of bacteria. We investigated the mechanism underlying OxPL action in macrophages. Exposure to OxPL induced alterations in actin polymerization, resulting in spreading of peritoneal macrophages and diminished uptake of E. coli. Pharmacological and cell-based studies showed that an anchored pool of PKA mediates the effects of OxPL. Gene silencing approaches identified the A-kinase anchoring protein (AKAP) WAVE1 as an effector of OxPL action in vitro. Chimeric Wave1(-/-) mice survived significantly longer after infection with E. coli and OxPL treatment in vivo. Moreover, we found that endogenously generated OxPL in human peritoneal dialysis fluid from end-stage renal failure patients inhibited phagocytosis via WAVE1. Collectively, these data uncover an unanticipated role for WAVE1 as a critical modulator of the innate immune response to severe bacterial infections.


Subject(s)
Escherichia coli Infections/immunology , Macrophages, Peritoneal/immunology , Peritonitis/immunology , Phagocytosis , Phospholipids/physiology , Wiskott-Aldrich Syndrome Protein Family/metabolism , Animals , Cell Line , Cyclic AMP-Dependent Protein Kinases/metabolism , Dimyristoylphosphatidylcholine/pharmacology , Enzyme Activation , Escherichia coli/immunology , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Humans , Immunity, Innate , Kidney Failure, Chronic/immunology , Kidney Failure, Chronic/metabolism , Kidney Failure, Chronic/therapy , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidation-Reduction , Peritoneal Dialysis , Peritonitis/metabolism , Peritonitis/microbiology , Phosphatidylcholines/pharmacology , Phosphatidylcholines/physiology , Wiskott-Aldrich Syndrome Protein Family/genetics
14.
J Biol Chem ; 288(24): 17111-21, 2013 Jun 14.
Article in English | MEDLINE | ID: mdl-23625929

ABSTRACT

PKA is retained within distinct subcellular environments by the association of its regulatory type II (RII) subunits with A-kinase anchoring proteins (AKAPs). Conventional reagents that universally disrupt PKA anchoring are patterned after a conserved AKAP motif. We introduce a phage selection procedure that exploits high-resolution structural information to engineer RII mutants that are selective for a particular AKAP. Selective RII (RSelect) sequences were obtained for eight AKAPs following competitive selection screening. Biochemical and cell-based experiments validated the efficacy of RSelect proteins for AKAP2 and AKAP18. These engineered proteins represent a new class of reagents that can be used to dissect the contributions of different AKAP-targeted pools of PKA. Molecular modeling and high-throughput sequencing analyses revealed the molecular basis of AKAP-selective interactions and shed new light on native RII-AKAP interactions. We propose that this structure-directed evolution strategy might be generally applicable for the investigation of other protein interaction surfaces.


Subject(s)
A Kinase Anchor Proteins/chemistry , Cell Surface Display Techniques , A Kinase Anchor Proteins/genetics , A Kinase Anchor Proteins/metabolism , Amino Acid Sequence , Amino Acid Substitution , Binding Sites , Consensus Sequence , HEK293 Cells , High-Throughput Nucleotide Sequencing , Humans , Membrane Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Protein Transport , Sequence Analysis, DNA
15.
Mol Cell ; 48(4): 547-59, 2012 Nov 30.
Article in English | MEDLINE | ID: mdl-23063527

ABSTRACT

The mitogenic and second-messenger signals that promote cell proliferation often proceed through multienzyme complexes. The kinase-anchoring protein Gravin integrates cAMP and calcium/phospholipid signals at the plasma membrane by sequestering protein kinases A and C with G protein-coupled receptors. In this report we define a role for Gravin as a temporal organizer of phosphorylation-dependent protein-protein interactions during mitosis. Mass spectrometry, molecular, and cellular approaches show that CDK1/Cyclin B1 phosphorylates Gravin on threonine 766 to prime the recruitment of the polo-like kinase Plk1 at defined phases of mitosis. Fluorescent live-cell imaging reveals that cells depleted of Gravin exhibit mitotic defects that include protracted prometaphase and misalignment of chromosomes. Moreover, a Gravin T766A phosphosite mutant that is unable to interact with Plk1 negatively impacts cell proliferation. In situ detection of phospho-T766 Gravin in biopsy sections of human glioblastomas suggests that this phosphorylation event might identify malignant neoplasms.


Subject(s)
A Kinase Anchor Proteins/metabolism , Cell Cycle Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , A Kinase Anchor Proteins/genetics , Animals , Cell Cycle Proteins/genetics , Cell Division , Cell Proliferation , Humans , Mice , Mitosis , Phosphorylation , Protein Binding , Tumor Cells, Cultured , Polo-Like Kinase 1
16.
EMBO J ; 31(20): 3991-4004, 2012 Oct 17.
Article in English | MEDLINE | ID: mdl-22940692

ABSTRACT

Endocrine release of insulin principally controls glucose homeostasis. Nutrient-induced exocytosis of insulin granules from pancreatic ß-cells involves ion channels and mobilization of Ca(2+) and cyclic AMP (cAMP) signalling pathways. Whole-animal physiology, islet studies and live-ß-cell imaging approaches reveal that ablation of the kinase/phosphatase anchoring protein AKAP150 impairs insulin secretion in mice. Loss of AKAP150 impacts L-type Ca(2+) currents, and attenuates cytoplasmic accumulation of Ca(2+) and cAMP in ß-cells. Yet surprisingly AKAP150 null animals display improved glucose handling and heightened insulin sensitivity in skeletal muscle. More refined analyses of AKAP150 knock-in mice unable to anchor protein kinase A or protein phosphatase 2B uncover an unexpected observation that tethering of phosphatases to a seven-residue sequence of the anchoring protein is the predominant molecular event underlying these metabolic phenotypes. Thus anchored signalling events that facilitate insulin secretion and glucose homeostasis may be set by AKAP150 associated phosphatase activity.


Subject(s)
A Kinase Anchor Proteins/physiology , Glucose/metabolism , Homeostasis/physiology , Insulin Resistance/genetics , Membrane Proteins/physiology , Phosphoprotein Phosphatases/physiology , A Kinase Anchor Proteins/chemistry , A Kinase Anchor Proteins/deficiency , A Kinase Anchor Proteins/genetics , Amino Acid Motifs , Animals , Calcineurin/metabolism , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cyclic AMP/physiology , Glucose/pharmacology , Homeostasis/drug effects , Insulin/metabolism , Insulin/pharmacology , Insulin Secretion , Insulinoma/pathology , Islets of Langerhans/drug effects , Islets of Langerhans/enzymology , Islets of Langerhans/metabolism , Liver/enzymology , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Molecular , Muscle, Skeletal/enzymology , Pancreatic Neoplasms/pathology , Protein Interaction Mapping , Protein Kinases/metabolism , Second Messenger Systems/drug effects , Second Messenger Systems/physiology , Sequence Deletion , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism
17.
EMBO J ; 31(14): 3147-56, 2012 May 29.
Article in English | MEDLINE | ID: mdl-22643219

ABSTRACT

Several neurotransmitters, including acetylcholine, regulate neuronal tone by suppressing a non-inactivating low-threshold voltage-gated potassium current generated by the M-channel. Agonist dependent control of the M-channel is mediated by calmodulin, activation of anchored protein kinase C (PKC), and depletion of the phospholipid messenger phosphatidylinositol 4,5-bisphosphate (PIP2). In this report, we show how this trio of second messenger responsive events acts synergistically and in a stepwise manner to suppress activity of the M-current. PKC phosphorylation of the KCNQ2 channel subunit induces dissociation of calmodulin from the M-channel complex. The calmodulin-deficient channel has a reduced affinity towards PIP2. This pathway enhances the effect of concomitant reduction of PIP2, which leads to disruption of the M-channel function. These findings clarify how a common lipid cofactor, such as PIP2, can selectively regulate ion channels.


Subject(s)
Ion Channel Gating/physiology , KCNQ2 Potassium Channel/metabolism , Receptors, Muscarinic/metabolism , Second Messenger Systems/physiology , Animals , CHO Cells , Cricetinae , Cricetulus , HEK293 Cells , Humans , KCNQ2 Potassium Channel/genetics , Phosphatidylinositol 4,5-Diphosphate/genetics , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphorylation/physiology , Protein Kinase C/genetics , Protein Kinase C/metabolism , Rats , Receptors, Muscarinic/genetics
18.
EMBO Mol Med ; 4(1): 15-26, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22095752

ABSTRACT

A decline in ocular lens transparency known as cataract afflicts 90% of individuals by the age 70. Chronic deterioration of lens tissue occurs as a pathophysiological consequence of defective water and nutrient circulation through channel and transporter proteins. A key component is the aquaporin-0 (AQP0) water channel whose permeability is tightly regulated in healthy lenses. Using a variety of cellular and biochemical approaches we have discovered that products of the A-kinase anchoring protein 2 gene (AKAP2/AKAP-KL) form a stable complex with AQP0 to sequester protein kinase A (PKA) with the channel. This permits PKA phosphorylation of serine 235 within a calmodulin (CaM)-binding domain of AQP0. The additional negative charge introduced by phosphoserine 235 perturbs electrostatic interactions between AQP0 and CaM to favour water influx through the channel. In isolated mouse lenses, displacement of PKA from the AKAP2-AQP0 channel complex promotes cortical cataracts as characterized by severe opacities and cellular damage. Thus, anchored PKA modulation of AQP0 is a homeostatic mechanism that must be physically intact to preserve lens transparency.


Subject(s)
A Kinase Anchor Proteins/metabolism , Aquaporins/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Eye Proteins/metabolism , Lens, Crystalline/metabolism , Membrane Proteins/metabolism , Animals , Aquaporins/chemistry , Calmodulin/metabolism , Cataract/metabolism , Cataract/pathology , Eye Proteins/chemistry , Lens, Crystalline/enzymology , Mice , Mice, Inbred C57BL , Phosphopeptides/analysis , Phosphorylation , Protein Binding , Sheep , Static Electricity , Water/metabolism
19.
Proc Natl Acad Sci U S A ; 108(48): E1227-35, 2011 Nov 29.
Article in English | MEDLINE | ID: mdl-22084075

ABSTRACT

A-kinase anchoring proteins (AKAPs) tether the cAMP-dependent protein kinase (PKA) to intracellular sites where they preferentially phosphorylate target substrates. Most AKAPs exhibit nanomolar affinity for the regulatory (RII) subunit of the type II PKA holoenzyme, whereas dual-specificity anchoring proteins also bind the type I (RI) regulatory subunit of PKA with 10-100-fold lower affinity. A range of cellular, biochemical, biophysical, and genetic approaches comprehensively establish that sphingosine kinase interacting protein (SKIP) is a truly type I-specific AKAP. Mapping studies located anchoring sites between residues 925-949 and 1,140-1,175 of SKIP that bind RI with dissociation constants of 73 and 774 nM, respectively. Molecular modeling and site-directed mutagenesis approaches identify Phe 929 and Tyr 1,151 as RI-selective binding determinants in each anchoring site. SKIP complexes exist in different states of RI-occupancy as single-molecule pull-down photobleaching experiments show that 41 ± 10% of SKIP sequesters two YFP-RI dimers, whereas 59 ± 10% of the anchoring protein binds a single YFP-RI dimer. Imaging, proteomic analysis, and subcellular fractionation experiments reveal that SKIP is enriched at the inner mitochondrial membrane where it associates with a prominent PKA substrate, the coiled-coil helix protein ChChd3.


Subject(s)
A Kinase Anchor Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Models, Molecular , Protein Conformation , A Kinase Anchor Proteins/genetics , Adaptor Proteins, Signal Transducing/genetics , Analysis of Variance , Animals , Blotting, Western , Cell Line , Cloning, Molecular , Humans , Immunoprecipitation , Mass Spectrometry , Mice , Mutagenesis, Site-Directed , Protein Binding/genetics , Surface Plasmon Resonance , Transfection
20.
J Biol Chem ; 286(45): 39269-81, 2011 Nov 11.
Article in English | MEDLINE | ID: mdl-21890631

ABSTRACT

Cell movement requires the coordinated reception, integration, and processing of intracellular signals. We have discovered that the protein kinase A anchoring protein AKAP220 interacts with the cytoskeletal scaffolding protein IQGAP1 to influence cell motility. AKAP220/IQGAP1 networks receive and integrate calcium and cAMP second messenger signals and position signaling enzymes near their intended substrates at leading edges of migrating cells. IQGAP1 supports calcium/calmodulin-dependent association of factors that modulate microtubule dynamics. AKAP220 suppresses GSK-3ß and positions this kinase to allow recruitment of the plus-end microtubule tracking protein CLASP2. Gene silencing of AKAP220 alters the rate of microtubule polymerization and the lateral tracking of growing microtubules and retards cell migration in metastatic human cancer cells. This reveals an unappreciated role for this anchored kinase/microtubule effector protein network in the propagation of cell motility.


Subject(s)
A Kinase Anchor Proteins/metabolism , Cell Movement/physiology , Second Messenger Systems/physiology , A Kinase Anchor Proteins/genetics , Calcium/metabolism , Calmodulin/genetics , Calmodulin/metabolism , Cell Line, Tumor , Cyclic AMP/genetics , Cyclic AMP/metabolism , Gene Silencing , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Microtubules/genetics , Microtubules/metabolism , ras GTPase-Activating Proteins/genetics , ras GTPase-Activating Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...