Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
J Clin Invest ; 127(4): 1438-1450, 2017 Apr 03.
Article in English | MEDLINE | ID: mdl-28263187

ABSTRACT

Huntington's disease (HD) is a polyglutamine (polyQ) disease caused by aberrant expansion of the polyQ tract in Huntingtin (HTT). While motor impairment mediated by polyQ-expanded HTT has been intensively studied, molecular mechanisms for nonmotor symptoms in HD, such as psychiatric manifestations, remain elusive. Here we have demonstrated that HTT forms a ternary protein complex with the scaffolding protein DISC1 and cAMP-degrading phosphodiesterase 4 (PDE4) to regulate PDE4 activity. We observed pathological cross-seeding between DISC1 and mutant HTT aggregates in the brains of HD patients as well as in a murine model that recapitulates the polyQ pathology of HD (R6/2 mice). In R6/2 mice, consequent reductions in soluble DISC1 led to dysregulation of DISC1-PDE4 complexes, aberrantly increasing the activity of PDE4. Importantly, exogenous expression of a modified DISC1, which binds to PDE4 but not mutant HTT, normalized PDE4 activity and ameliorated anhedonia in the R6/2 mice. We propose that cross-seeding of mutant HTT and DISC1 and the resultant changes in PDE4 activity may underlie the pathology of a specific subset of mental manifestations of HD, which may provide an insight into molecular signaling in mental illness in general.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Huntington Disease/enzymology , Nerve Tissue Proteins/metabolism , Protein Aggregation, Pathological/enzymology , Animals , Female , HEK293 Cells , Humans , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Mice, Transgenic , Mutation
2.
Cell Signal ; 26(9): 1958-74, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24815749

ABSTRACT

In Rat-1 cells, the dramatic decrease in the levels of both intracellular cyclic 3'5' adenosine monophosphate (cyclic AMP; cAMP) and in the activity of cAMP-activated protein kinase A (PKA) observed in mitosis was paralleled by a profound increase in cAMP hydrolyzing phosphodiesterase-4 (PDE4) activity. The decrease in PKA activity, which occurs during mitosis, was attributable to PDE4 activation as the PDE4 selective inhibitor, rolipram, but not the phosphodiesterase-3 (PDE3) inhibitor, cilostamide, specifically ablated this cell cycle-dependent effect. PDE4 inhibition caused Rat-1 cells to move from S phase into G2/M more rapidly, to transit through G2/M more quickly and to remain in G1 for a longer period. Inhibition of PDE3 elicited no observable effects on cell cycle dynamics. Selective immunopurification of each of the four PDE4 sub-families identified PDE4D as being selectively activated in mitosis. Subsequent analysis uncovered PDE4D9, an isoform whose expression can be regulated by Disrupted-In-Schizophrenia 1 (DISC1)/activating transcription factor 4 (ATF4) complex, as the sole PDE4 species activated during mitosis in Rat-1 cells. PDE4D9 becomes activated in mitosis through dual phosphorylation at Ser585 and Ser245, involving the combined action of ERK and an unidentified 'switch' kinase that has previously been shown to be activated by H2O2. Additionally, in mitosis, PDE4D9 also becomes phosphorylated at Ser67 and Ser81, through the action of MK2 (MAPKAPK2) and AMP kinase (AMPK), respectively. The multisite phosphorylation of PDE4D9 by all four of these protein kinases leads to decreased mobility (band-shift) of PDE4D9 on SDS-PAGE. PDE4D9 is predominantly concentrated in the perinuclear region of Rat-1 cells but with a fraction distributed asymmetrically at the cell margins. Our investigations demonstrate that the diminished levels of cAMP and PKA activity that characterise mitosis are due to enhanced cAMP degradation by PDE4D9. PDE4D9, was found to locate primarily not only in the perinuclear region of Rat-1 cells but also at the cell margins. We propose that the sequestration of PDE4D9 in a specific complex together with AMPK, ERK, MK2 and the H2O2-activatable 'switch' kinase allows for its selective multi-site phosphorylation, activation and regulation in mitosis.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Mitosis , Nerve Tissue Proteins/metabolism , Activating Transcription Factor 4/metabolism , Amino Acid Sequence , Animals , Cell Line , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation , Interphase , Intracellular Signaling Peptides and Proteins/metabolism , Molecular Sequence Data , Phosphorylation , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases/metabolism , Rats
3.
Proc Natl Acad Sci U S A ; 110(16): E1533-42, 2013 Apr 16.
Article in English | MEDLINE | ID: mdl-23509299

ABSTRACT

V-raf-1 murine leukemia viral oncogene homolog 1 (Raf-1) is a key activator of the ERK pathway and is a target for cross-regulation of this pathway by the cAMP signaling system. The cAMP-activated protein kinase, PKA, inhibits Raf-1 by phosphorylation on S259. Here, we show that the cAMP-degrading phosphodiesterase-8A (PDE8A) associates with Raf-1 to protect it from inhibitory phosphorylation by PKA, thereby enhancing Raf-1's ability to stimulate ERK signaling. PDE8A binds to Raf-1 with high (picomolar) affinity. Mapping of the interaction domain on PDE8A using peptide array technology identified amino acids 454-465 as the main binding site, which could be disrupted by mutation. A cell-permeable peptide corresponding to this region disrupted the PDE8A/Raf-1 interaction in cells, thereby reducing ERK activation and the cellular response to EGF. Overexpression of a catalytically inactive PDE8A in cells displayed a dominant negative phenotype on ERK activation. These effects were recapitulated at the organism level in genetically modified (PDE8A(-/-)) mice. Similarly, PDE8 deletion in Drosophila melanogaster reduced basal ERK activation and sensitized flies to stress-induced death. We propose that PDE8A is a physiological regulator of Raf-1 signaling in some cells.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/metabolism , MAP Kinase Signaling System/physiology , Proto-Oncogene Proteins c-raf/metabolism , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , Animals , Blotting, Western , DNA Primers/genetics , Drosophila melanogaster , Gene Deletion , HEK293 Cells , HeLa Cells , Humans , Immunoprecipitation , MAP Kinase Signaling System/genetics , Mass Spectrometry , Mice , Mice, Knockout , Mutagenesis, Site-Directed , Phosphorylation , Surface Plasmon Resonance
4.
J Med Chem ; 54(9): 3331-47, 2011 May 12.
Article in English | MEDLINE | ID: mdl-21456625

ABSTRACT

A survey of PDE4 inhibitors reveals that some compounds trigger intracellular aggregation of PDE4A4 into accretion foci through association with the ubiquitin-binding scaffold protein p62 (SQSTM1). We show that this effect is driven by inhibitor occupancy of the catalytic pocket and stabilization of a "capped state" in which a sequence within the enzyme's upstream conserved region 2 (UCR2) module folds across the catalytic pocket. Only certain inhibitors cause PDE4A4 foci formation, and the structural features responsible for driving the process are defined. Switching to the UCR2-capped state induces conformational transition in the enzyme's regulatory N-terminal portion, facilitating protein association events responsible for reversible aggregate assembly. PDE4-selective inhibitors able to trigger relocalization of PDE4A4 into foci can therefore be expected to exert actions on cells that extend beyond simple inhibition of PDE4 catalytic activity and that may arise from reconfiguring the enzyme's protein association partnerships.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Phosphodiesterase 4 Inhibitors/pharmacology , Animals , CHO Cells , Catalytic Domain , Cricetinae , Cricetulus , Crystallography, X-Ray , Cyclic Nucleotide Phosphodiesterases, Type 4/chemistry , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/metabolism , Models, Molecular , Phosphodiesterase 4 Inhibitors/chemistry , Pyridines/chemistry , Pyridines/pharmacology , Rolipram/chemistry , Rolipram/pharmacology , Sequestosome-1 Protein , Spiro Compounds/chemistry , Spiro Compounds/pharmacology , Stereoisomerism , Structure-Activity Relationship , Xanthines/chemistry , Xanthines/pharmacology
5.
Mol Cell Biol ; 30(22): 5406-20, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20837708

ABSTRACT

The mammalian target of rapamycin complex 1 (mTORC1) is a molecular hub that regulates protein synthesis in response to a number of extracellular stimuli. Cyclic AMP (cAMP) is considered to be an important second messenger that controls mTOR; however, the signaling components of this pathway have not yet been elucidated. Here, we identify cAMP phosphodiesterase 4D (PDE4D) as a binding partner of Rheb that acts as a cAMP-specific negative regulator of mTORC1. Under basal conditions, PDE4D binds Rheb in a noncatalytic manner that does not require its cAMP-hydrolyzing activity and thereby inhibits the ability of Rheb to activate mTORC1. However, elevated cAMP levels disrupt the interaction of PDE4D with Rheb and increase the interaction between Rheb and mTOR. This enhanced Rheb-mTOR interaction induces the activation of mTORC1 and cap-dependent translation, a cellular function of mTORC1. Taken together, our results suggest a novel regulatory mechanism for mTORC1 in which the cAMP-determined dynamic interaction between Rheb and PDE4D provides a key, unique regulatory event. We also propose a new role for PDE4 as a molecular transducer for cAMP signaling.


Subject(s)
Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Monomeric GTP-Binding Proteins/metabolism , Neuropeptides/metabolism , Second Messenger Systems/physiology , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Line , Cells, Cultured , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Fibroblasts/cytology , Fibroblasts/physiology , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Monomeric GTP-Binding Proteins/genetics , Multiprotein Complexes , Neuropeptides/genetics , Protein Binding , Proteins , Ras Homolog Enriched in Brain Protein , TOR Serine-Threonine Kinases/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
6.
J Biol Chem ; 285(44): 33614-22, 2010 Oct 29.
Article in English | MEDLINE | ID: mdl-20732872

ABSTRACT

Vascular endothelial cell (VEC) permeability is largely dependent on the integrity of vascular endothelial cadherin (VE-cadherin or VE-Cad)-based intercellular adhesions. Activators of protein kinase A (PKA) or of exchange protein activated by cAMP (EPAC) reduce VEC permeability largely by stabilizing VE-Cad-based intercellular adhesions. Currently, little is known concerning the nature and composition of the signaling complexes that allow PKA or EPAC to regulate VE-Cad-based structures and through these actions control permeability. Using pharmacological, biochemical, and cell biological approaches we identified and determined the composition and functionality of a signaling complex that coordinates cAMP-mediated control of VE-Cad-based adhesions and VEC permeability. Thus, we report that PKA, EPAC1, and cyclic nucleotide phosphodiesterase 4D (PDE4D) enzymes integrate into VE-Cad-based signaling complexes in human arterial endothelial cells. Importantly, we show that protein-protein interactions between EPAC1 and PDE4D serve to foster their integration into VE-Cad-based complexes and allow robust local regulation of EPAC1-based stabilization of VE-Cad-based adhesions. Of potential translational importance, we mapped the EPAC1 peptide motif involved in binding PDE4D and show that a cell-permeable variant of this peptide antagonizes EPAC1-PDE4D binding and directly alters VEC permeability. Collectively, our data indicate that PDE4D regulates both the activity and subcellular localization of EPAC1 and identify a novel mechanism for regulated EPAC1 signaling in these cells.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Endothelium, Vascular/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Amino Acid Motifs , Atherosclerosis/metabolism , Cells, Cultured , Cyclic Nucleotide Phosphodiesterases, Type 4 , Humans , Intercellular Junctions/metabolism , Macromolecular Substances , Peptides/chemistry , Permeability , Signal Transduction , beta Catenin/metabolism
7.
Mol Cell Biol ; 30(18): 4379-90, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20647544

ABSTRACT

Calcineurin is a widely expressed and highly conserved Ser/Thr phosphatase. Calcineurin is inhibited by the immunosuppressant drug cyclosporine A (CsA) or tacrolimus (FK506). The critical role of CsA/FK506 as an immunosuppressant following transplantation surgery provides a strong incentive to understand the phosphatase calcineurin. Here we uncover a novel regulatory pathway for cyclic AMP (cAMP) signaling by the phosphatase calcineurin which is also evolutionarily conserved in Caenorhabditis elegans. We found that calcineurin binds directly to and inhibits the proteosomal degradation of cAMP-hydrolyzing phosphodiesterase 4D (PDE4D). We show that ubiquitin conjugation and proteosomal degradation of PDE4D are controlled by a cullin 1-containing E(3) ubiquitin ligase complex upon dual phosphorylation by casein kinase 1 (CK1) and glycogen synthase kinase 3beta (GSK3beta) in a phosphodegron motif. Our findings identify a novel signaling process governing G-protein-coupled cAMP signal transduction-opposing actions of the phosphatase calcineurin and the CK1/GSK3beta protein kinases on the phosphodegron-dependent degradation of PDE4D. This novel signaling system also provides unique functional insights into the complications elicited by CsA in transplant patients.


Subject(s)
Caenorhabditis elegans Proteins , Calcineurin/genetics , Calcineurin/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Evolution, Molecular , Second Messenger Systems/physiology , Amino Acid Motifs , Animals , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Calcineurin Inhibitors , Cell Line , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Cyclosporine/metabolism , Enzyme Inhibitors/metabolism , Gene Expression Regulation, Enzymologic , Humans , Mice , Mice, Knockout , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism
8.
J Biol Chem ; 284(17): 11425-35, 2009 Apr 24.
Article in English | MEDLINE | ID: mdl-19153083

ABSTRACT

betaArrestin is a multifunctional signal scaffold protein. Using SPOT immobilized peptide arrays, coupled with scanning alanine substitution and mutagenesis, we show that the MAPK kinase, MEK1, interacts directly with betaarrestin1. Asp(26) and Asp(29) in the N-terminal domain of betaarrestin1 are critical for its binding to MEK1, whereas Arg(47) and Arg(49) in the N-terminal domain of MEK1 are critical for its binding to betaarrestin1. Wild-type FLAG-tagged betaarrestin1 co-immunopurifies with MEK1 in HEKB2 cells, whereas the D26A/D29A mutant does not. ERK-dependent phosphorylation at Ser(412) was compromised in the D26A/D29A-betaarrestin1 mutant. A cell-permeable, 25-mer N-stearoylated betaarrestin1 peptide that encompassed the N-domain MEK1 binding site blocked betaarrestin1/MEK1 association in HEK cells and recapitulated the altered phenotype seen with the D26A/D29A-betaarrestin1 in compromising the ERK-dependent phosphorylation of betaarrestin1. In addition, the MEK disruptor peptide promoted the ability of betaarrestin1 to co-immunoprecipitate with endogenous c-Src and clathrin, facilitating the isoprenaline-stimulated internalization of the beta(2)-adrenergic receptor.


Subject(s)
Arrestins/metabolism , Isoproterenol/metabolism , MAP Kinase Kinase 1/metabolism , Adrenergic beta-Agonists/pharmacology , Amino Acid Sequence , Arginine/chemistry , Aspartic Acid/chemistry , Clathrin/metabolism , Humans , Models, Biological , Molecular Sequence Data , Phosphorylation , Protein Structure, Tertiary , Sequence Homology, Amino Acid , beta-Arrestins , src-Family Kinases/metabolism
9.
Proc Natl Acad Sci U S A ; 105(35): 12791-6, 2008 Sep 02.
Article in English | MEDLINE | ID: mdl-18728186

ABSTRACT

We identify a compartmentalized signaling system that identifies a functional role for the GTP exchange factor, exchange protein activated by cAMP (EPAC) coupled to Rap2 in the nucleus. In this system, cAMP regulates the nuclear/cytoplasmic trafficking of DNA-dependent protein kinase (DNA-PK), a critical kinase that acts to repair double-stranded breaks (DSBs) in damaged DNA and to phosphorylate the cell survival kinase, PKB/Akt. Intersecting regulatory inputs for cAMP employ EPAC to transduce positive effects, namely the Rap2-dependent nuclear exit and activation of DNA-PK, whereas protein kinase A (PKA) provides the negative input by antagonizing these actions. We identify this as a compartmentalized regulatory system where modulation of cAMP input into the stimulatory, EPAC and inhibitory, PKA intersecting arms is provided by spatially discrete, cAMP degradation systems. The distribution of DNA-PK between nuclear and cytoplasmic compartments can thus potentially be influenced by relative inputs of cAMP signaling through the EPAC and PKA pathways. Through this signaling system EPAC activation can thereby impact on the Ser-473 phosphorylation status of PKB/Akt and the repair of etoposide-induced DSBs.


Subject(s)
Cell Nucleus/enzymology , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Amino Acid Sequence , DNA Breaks, Double-Stranded , Enzyme Activation , HeLa Cells , Humans , Intracellular Space/metabolism , Molecular Sequence Data , Peptides/chemistry , Phosphoric Diester Hydrolases/metabolism , Phosphorylation , Phosphoserine/metabolism , Protein Transport , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , rap GTP-Binding Proteins/metabolism
10.
J Cell Sci ; 119(Pt 18): 3799-810, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-16940352

ABSTRACT

The unique N-terminal regions of PDE4 cAMP-specific phosphodiesterases confer interaction with distinct signalling and scaffolding proteins. The PDE4A1 isoform is unique in being entirely membrane associated. Its N-terminal region is formed from two helices separated by a mobile hinge, where helix-2 contains a TAPAS1 domain that inserts into the lipid bilayer in a Ca2+-triggered fashion. Here we show that helix-1 is important for intracellular targeting of PDE4A1 in living cells, facilitating membrane association, targeting to the trans-Golgi stack and conferring Ca2+-stimulated intracellular redistribution in a manner that is dependent on the phospholipase-D-mediated generation of phosphatidic acid. The LxDFF motif within helix-1 is pivotal to this, where Leu4-Phe6-Phe7 forms a compact hydrophobic pocket on one side of helix-1 whereas Asp5, located on the opposite face of helix-1, provides the Ca2+-regulation site. Mutation of Asp5 to Ala or the release of Ca2+ from intracellular stores de-restricts trans-Golgi localisation of PDE4A1 allowing it to redistribute in cells in a phosphatidic-acid-dependent manner. This study provides the first evidence for Ca2+-triggered relocalisation of a cAMP phosphodiesterase and indicates a potential means for allowing cross-talk between the cAMP, phospholipase D and Ca2+-signalling pathways.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/chemistry , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Calcium/metabolism , Phospholipase D/metabolism , Amino Acid Sequence , Animals , Binding Sites , COS Cells , Calcium Signaling/drug effects , Cell Survival/drug effects , Chlorocebus aethiops , Cyclic Nucleotide Phosphodiesterases, Type 4 , Models, Molecular , Molecular Sequence Data , Phosphatidic Acids/biosynthesis , Protein Structure, Secondary , Protein Transport/drug effects , Recombinant Fusion Proteins/metabolism , Structure-Activity Relationship , Thapsigargin/pharmacology , trans-Golgi Network/drug effects
11.
Biochem J ; 394(Pt 2): 427-35, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16356165

ABSTRACT

Membrane-recruitment of GRK2 (G-protein receptor kinase 2) provides a fundamental step in the desensitization process controlling GPCRs (G-protein-coupled receptors), such as the beta2AR (beta2-adrenergic receptor). In the present paper, we show that challenge of HEK-293beta2 [human embryonic kidney cells stably overexpressing the FLAG-tagged beta2AR-GFP (green fluorescent protein)] cells with the beta-adrenoceptor agonist, isoprenaline, causes GRK2 to become phosphorylated by PKA (cAMP-dependent protein kinase). This action is facilitated when cAMP-specific PDE4 (phosphodiesterase-4) activity is selectively inactivated, either chemically with rolipram or by siRNA (small interfering RNA)-mediated knockdown of PDE4B and PDE4D. PDE4-selective inhibition by rolipram facilitates the isoprenaline-induced membrane translocation of GRK2, phosphorylation of the beta2AR by GRK2, membrane translocation of beta-arrestin and internalization of beta2ARs. PDE4-selective inhibition also enhances the ability of isoprenaline to trigger the PKA phosphorylation of GRK2 in cardiac myocytes. In the absence of isoprenaline, rolipram-induced inhibition of PDE4 activity in HEK-293beta2 cells acts to stimulate PKA phosphorylation of GRK2, with consequential effects on GRK2 membrane recruitment and GRK2-mediated phosphorylation of the beta2AR. We propose that a key role for PDE4 enzymes is: (i) to gate the action of PKA on GRK2, influencing the rate of GRK2 phosphorylation of the beta2AR and consequential recruitment of beta-arrestin subsequent to beta-adrenoceptor agonist challenge, and (ii) to protect GRK2 from inappropriate membrane recruitment in unstimulated cells through its phosphorylation by PKA in response to fluctuations in basal levels of cAMP.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Cell Membrane/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Myocytes, Cardiac/metabolism , beta-Adrenergic Receptor Kinases/metabolism , 3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Cell Line , Cyclic Nucleotide Phosphodiesterases, Type 3 , Cyclic Nucleotide Phosphodiesterases, Type 4 , G-Protein-Coupled Receptor Kinase 2 , Humans , Isoproterenol/pharmacology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , Phosphorylation , Protein Transport , Rolipram/pharmacology
12.
Science ; 310(5751): 1187-91, 2005 Nov 18.
Article in English | MEDLINE | ID: mdl-16293762

ABSTRACT

The disrupted in schizophrenia 1 (DISC1) gene is a candidate susceptibility factor for schizophrenia, but its mechanistic role in the disorder is unknown. Here we report that the gene encoding phosphodiesterase 4B (PDE4B) is disrupted by a balanced translocation in a subject diagnosed with schizophrenia and a relative with chronic psychiatric illness. The PDEs inactivate adenosine 3',5'-monophosphate (cAMP), a second messenger implicated in learning, memory, and mood. We show that DISC1 interacts with the UCR2 domain of PDE4B and that elevation of cellular cAMP leads to dissociation of PDE4B from DISC1 and an increase in PDE4B activity. We propose a mechanistic model whereby DISC1 sequesters PDE4B in resting cells and releases it in an activated state in response to elevated cAMP.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/genetics , Cyclic AMP/metabolism , Nerve Tissue Proteins/genetics , Schizophrenia/genetics , Signal Transduction , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Adult , Affective Disorders, Psychotic/genetics , Affective Disorders, Psychotic/metabolism , Animals , Cadherins/genetics , Cell Line , Chromosomes, Human, Pair 1 , Chromosomes, Human, Pair 16 , Cyclic Nucleotide Phosphodiesterases, Type 4 , Enzyme Activation , Genetic Predisposition to Disease , Humans , Male , Nerve Tissue Proteins/metabolism , Protein Binding , Rats , Schizophrenia/enzymology , Schizophrenia/metabolism , Translocation, Genetic
13.
Cell Signal ; 17(9): 1158-73, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15905070

ABSTRACT

We employ a novel, dominant negative approach to identify a key role for certain tethered cyclic AMP specific phosphodiesterase-4 (PDE4) isoforms in regulating cyclic AMP dependent protein kinase A (PKA) sub-populations in resting COS1 cells. A fraction of PKA is clearly active in resting COS1 cells and this activity increases when cells are treated with the selective PDE4 inhibitor, rolipram. Point mutation of a critical, conserved aspartate residue in the catalytic site of long PDE4A4, PDE4B1, PDE4C2 and PDE4D3 isoforms renders them catalytically inactive. Overexpressed in resting COS1 cells, catalytically inactive forms of PDE4C2 and PDE4D3, but not PDE4A4 and PDE4B1, are constitutively PKA phosphorylated while overexpressed active versions of all these isoforms are not. Inactive and active versions of all these isoforms are PKA phosphorylated in cells where protein kinase A is maximally activated with forskolin and IBMX. By contrast, rolipram challenge of COS1 cells selectively triggers the PKA phosphorylation of recombinant, active PDE4D3 and PDE4C2 but not recombinant, active PDE4A4 and PDE4B1. Purified, recombinant PDE4D3 and PDE4A4 show a similar dose-dependency for in vitro phosphorylation by PKA. Disruption of the tethering of PKA type-II to PKA anchor proteins (AKAPs), achieved using the peptide Ht31, prevents inactive forms of PDE4C2 and PDE4D3 being constitutively PKA phosphorylated in resting cells as does siRNA-mediated knockdown of PKA-RII, but not PKA-RI. PDE4C2 and PDE4D3 co-immunoprecipitate from COS1 cell lysates with 250 kDa and 450 kDa AKAPs that tether PKA type-II and not PKA type-I. PKA type-II co-localises with AKAP450 in the centrosomal region of COS1 cells. The perinuclear distribution of recombinant, inactive PDE4D3, but not inactive PDE4A4, overlaps with AKAP450 and PKA type-II. The distribution of PKA phosphorylated inactive PDE4D3 also overlaps with that of AKAP450 in the centrosomal region of COS1 cells. We propose that a novel role for PDE4D3 and PDE4C2 is to gate the activation of AKAP450-tethered PKA type-II localised in the perinuclear region under conditions of basal cAMP generation in resting cells.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/physiology , Adaptor Proteins, Signal Transducing/metabolism , Centrosome/enzymology , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/biosynthesis , 3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , Animals , COS Cells , Chlorocebus aethiops , Cyclic AMP-Dependent Protein Kinase Type II , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic Nucleotide Phosphodiesterases, Type 4 , Enzyme Activation , Isoenzymes/genetics , Isoenzymes/metabolism , Microscopy, Confocal , Mutation , Phosphodiesterase Inhibitors/pharmacology , RNA Interference , Rolipram/pharmacology
14.
Mol Pharmacol ; 67(6): 1920-34, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15738310

ABSTRACT

PDE4A11 is a novel cAMP-specific phosphodiesterase that is conserved in humans, mouse, rat, pig, and bat. Exon-1(4A11) encodes its unique, 81 amino acid N-terminal region. Reverse-transcriptase polymerase chain reaction performed across the splice junction, plus identification of expressed sequence tags, identifies PDE4A11 as a long isoform possessing UCR1 and UCR2 regulatory domains. Transcript analysis shows that PDE4A11 is widely expressed compared with PDE4A10 and PDE4A4B long isoforms. Truncation analysis identifies a putative promoter in a 250-base pair region located immediately upstream of the start site in Exon-1(4A11). Recombinant PDE4A11, expressed in COS-7 cells, is a 126-kDa protein localized predominantly around the nucleus and in membrane ruffles. PDE4A11 exhibits a K(m) for cAMP hydrolysis of 4 microM, with relative V(max) similar to that of PDE4A10 and PDE4A4B. PDE4A11 is dose-dependently inhibited by rolipram, 4-[(3-butoxy-4-methoxyphenyl)-methyl]-2-imidazolidinone (Ro 20-1724), cilomilast, roflumilast, and denbufylline, with IC(50) values of 0.7, 0.9, 0.03, 0.004, and 0.3 microM, respectively. Soluble and particulate PDE4A11 exhibit distinct rates of thermal inactivation (55 degrees C; T((0.5)) = 2.5 and 4.4 min, respectively). Elevating cAMP levels in COS-7 cells activates PDE4A11 concomitant with its phosphorylation at Ser119 by protein kinase A (PKA). PDE4A11 differs from PDE4A4 in sensitivity to cleavage by caspase-3, interaction with LYN SH3 domain, redistribution upon long-term rolipram challenge, and sensitivity to certain PDE4 inhibitors. PDE4A11, PDE4A10, and PDE4A4 all can interact with betaarrestin. PDE4A11 is a novel, widely expressed long isoform that is activated by PKA phosphorylation and shows a distinct intracellular localization, indicating that it may contribute to compartmentalized cAMP signaling in cells in which it is expressed.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/biosynthesis , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , Gene Expression Regulation, Enzymologic/physiology , 3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Chlorocebus aethiops , Cyclic AMP/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4 , Dose-Response Relationship, Drug , Gene Expression Regulation, Enzymologic/drug effects , Humans , Intracellular Fluid/enzymology , Isoenzymes/antagonists & inhibitors , Isoenzymes/biosynthesis , Isoenzymes/genetics , Mice , Molecular Sequence Data , Phosphodiesterase Inhibitors/pharmacology , Signal Transduction
15.
Circ Res ; 95(1): 67-75, 2004 Jul 09.
Article in English | MEDLINE | ID: mdl-15178638

ABSTRACT

Cardiac myocytes have provided a key paradigm for the concept of the compartmentalized cAMP generation sensed by AKAP-anchored PKA. Phosphodiesterases (PDEs) provide the sole route for degrading cAMP in cells and are thus poised to regulate intracellular cAMP gradients. PDE3 and PDE4 represent the major cAMP degrading activities in rat ventriculocytes. By performing real-time imaging of cAMP in situ, we establish the hierarchy of these PDEs in controlling cAMP levels in basal conditions and on stimulation with a beta-adrenergic receptor agonist. PDE4, rather than PDE3, appears to be responsible for modulating the amplitude and duration of the cAMP response to beta-agonists. PDE3 and PDE4 localize to distinct compartments and this may underpin their different functional roles. Our findings indicate the importance of distinctly localized PDE isoenzymes in determining compartmentalized cAMP signaling.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/physiology , Cyclic AMP/metabolism , Myocytes, Cardiac/enzymology , 3',5'-Cyclic-AMP Phosphodiesterases/analysis , 3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Adrenergic alpha-Agonists/pharmacology , Animals , Animals, Newborn , Cyclic Nucleotide Phosphodiesterases, Type 3 , Cyclic Nucleotide Phosphodiesterases, Type 4 , Fluorescence Resonance Energy Transfer , Norepinephrine/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Rats
16.
Biochem J ; 380(Pt 2): 371-84, 2004 Jun 01.
Article in English | MEDLINE | ID: mdl-15025561

ABSTRACT

PDE4A7 is an isoform encoded by the human PDE4A cAMP-specific phosphodiesterase gene that fails to hydrolyse cAMP and whose transcripts are widely expressed. Removal of either the N- or C-terminal unique portions of PDE4A7 did not reconstitute catalytic activity, showing that they did not exert a chronic inhibitory effect. A chimera (Hyb2), formed by swapping the unique N-terminal portion of PDE4A7 with that of the active PDE4A4C form, was not catalytically active. However, one formed (Hyb1) by swapping the unique C-terminal portion of PDE4A7 with that common to all active PDE4 isoforms was catalytically active. Compared with the active PDE4A4B isoform, Hyb1 exhibited a similar K(m) value for cAMP and IC50 value for rolipram inhibition, but was less sensitive to inhibition by Ro-20-1724 and denbufylline, and considerably more sensitive to thermal denaturation. The unique C-terminal region of PDE4A7 was unable to support an active catalytic unit, whereas its unique N-terminal region can. The N-terminal portion of the PDE4 catalytic unit is essential for catalytic activity and can be supplied by either highly conserved sequence found in active PDE4 isoforms from all four PDE4 subfamilies or the unique N-terminal portion of PDE4A7. A discrete portion of the conserved C-terminal region in active PDE4A isoforms underpins their aberrant migration on SDS/PAGE. Unlike active PDE4A isoforms, PDE4A7 is exclusively localized to the P1 particulate fraction in cells. A region located within the C-terminal portion of active PDE4 isoforms prevents such exclusive targeting. Three functional regions in PDE4A isoforms are identified, which influence catalytic activity, subcellular targeting and conformational status.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/biosynthesis , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Catalytic Domain/physiology , 3',5'-Cyclic-AMP Phosphodiesterases/chemistry , 3',5'-Cyclic-AMP Phosphodiesterases/physiology , Amino Acid Sequence , Animals , COS Cells/chemistry , COS Cells/metabolism , Cell Line , Chlorocebus aethiops , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4 , DNA/blood , Electrophoresis, Polyacrylamide Gel/methods , Humans , Isoenzymes/biosynthesis , Isoenzymes/chemistry , Isoenzymes/metabolism , Isoenzymes/physiology , Kidney/chemistry , Kidney/cytology , Kidney/embryology , Leukocytes, Mononuclear/chemistry , Molecular Sequence Data , Molecular Weight , Organ Specificity/physiology , Peptides/physiology , Protein Structure, Tertiary/physiology , RNA/blood , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/physiology
17.
J Biol Chem ; 278(49): 49230-8, 2003 Dec 05.
Article in English | MEDLINE | ID: mdl-14500724

ABSTRACT

Isoproterenol challenge of Hek-B2 cells causes a transient recruitment of the endogenous PDE4D isoforms found in these cells, namely PDE4D3 and PDE4D5, to the membrane fraction. PDE4D5 provides around 80% of the total PDE4D protein so recruited, although it only comprises about 40% of the total PDE4D protein in Hek-B2 cells. PDE4D5 provides about 80% of the total PDE4D protein found associated with beta-arrestins immunopurified from Hek-B2, COS1, and A549 cells as well as cardiac myocytes, whereas its overall level in these cells is between 15 and 50% of the total PDE4D protein. Truncation analyses indicate that two sites in PDE4D5 are involved in mediating its interaction with beta-arrestins, one associated with the common PDE4 catalytic region and the other located within its unique amino-terminal region. Truncation analyses indicate that two sites in beta-arrestin 2 are involved in mediating its interaction with PDE4D5, one associated with its extreme amino-terminal region and the other located within the carboxyl-terminal domain of the protein. We suggest that the unique amino-terminal region of PDE4D5 allows it to preferentially interact with beta-arrestins. This specificity appears likely to account for the preferential recruitment of PDE4D5, compared with PDE4D3, to membranes of Hek-B2 cells and cardiac myocytes upon challenge with isoproterenol.


Subject(s)
Arrestins/metabolism , Isoenzymes/metabolism , Phosphoric Diester Hydrolases/metabolism , Amino Acid Sequence , Cell Line , Cyclic Nucleotide Phosphodiesterases, Type 3 , Cyclic Nucleotide Phosphodiesterases, Type 4 , Humans , Isoenzymes/chemistry , Molecular Sequence Data , Phosphoric Diester Hydrolases/chemistry , Protein Binding , beta-Arrestin 2 , beta-Arrestins
18.
Cell Signal ; 15(10): 955-71, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12873709

ABSTRACT

In cells transfected to express wild-type PDE4A4 cAMP phosphodiesterase (PDE), the PDE4 selective inhibitor rolipram caused PDE4A4 to relocalise so as to form accretion foci. This process was followed in detail in living cells using a PDE4A4 chimera formed with Green Fluorescent Protein (GFP). The same pattern of behaviour was also seen in chimeras of PDE4A4 formed with various proteins and peptides, including LimK, RhoC, FRB and the V5-6His tag. Maximal PDE4A4 foci formation, occurred over a period of about 10 h, was dose-dependent on rolipram and was reversible upon washout of rolipram. Inhibition of protein synthesis, using cycloheximide, but not PKA activity with H89, inhibited foci generation. Foci formation was elicited by Ro20-1724 and RS25344 but not by either Ariflo or RP73401, showing that not all PDE4 selective inhibitors had this effect. Ariflo and RP73401 dose-dependently antagonised rolipram-induced foci formation and dispersed rolipram pre-formed foci as did the adenylyl cyclase activator, forskolin. Foci formation showed specificity for PDE4A4 and its rodent homologue, PDE4A5, as it was not triggered in living cells expressing the PDE4B2, PDE4C2, PDE4D3 and PDE4D5 isoforms as GFP chimeras. Altered foci formation was seen in the Deltab-LR2-PDE4A4 construct, which deleted a region within LRZ, showing that appropriate linkage between the N-terminal portion of PDE4A4 and the catalytic unit of PDE4A4 was needed for foci formation. Certain single point mutations within the PDE4A4 catalytic site (His505Asn, His506Asn and Val475Asp) were shown to ablate foci formation but still allow rolipram inhibition of PDE4A4 catalytic activity. We suggest that the binding of certain, but not all, PDE4 selective inhibitors to PDE4A4 induces a conformational change in this isoform by 'inside-out' signalling that causes it to redistribute in the cell. Displacing foci-forming inhibitors with either cAMP or inhibitors that do not form foci can antagonise this effect. Specificity of this effect for PDE4A4 and its homologue PDE4A5 suggests that interplay between the catalytic site and the unique N-terminal region of these isoforms is required. Thus, certain PDE4 selective inhibitors may exert effects on PDE4A4 that extend beyond simple catalytic inhibition. These require protein synthesis and may lead to redistribution of PDE4A4 and any associated proteins. Foci formation of PDE4A4 may be of use in probing for conformational changes in this isoform and for sub-categorising PDE4 selective inhibitors.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Cyclic AMP/metabolism , Phosphodiesterase Inhibitors/pharmacology , Rolipram/pharmacology , 3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , Animals , Binding Sites , Catalysis , Catalytic Domain , Cell Line , Cricetinae , Cyclic Nucleotide Phosphodiesterases, Type 4 , Female , Green Fluorescent Proteins , Humans , Luminescent Proteins/metabolism , Phosphodiesterase Inhibitors/metabolism , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Recombinant Fusion Proteins/metabolism , Rolipram/metabolism , Transfection
19.
J Biol Chem ; 278(35): 33351-63, 2003 Aug 29.
Article in English | MEDLINE | ID: mdl-12810716

ABSTRACT

The cyclic AMP-specific phosphodiesterase (PDE4) isoform PDE4A5 interacted with the immunophilin XAP2 in a yeast two-hybrid assay. The interaction was confirmed in biochemical pull-down analyses. The interaction was specific, in that PDE4A5 did not interact with the closely related immunophilins AIPL1, FKBP51, or FKBP52. XAP2 also did not interact with other PDE4A isoforms or typical isoforms from the three other PDE4 subfamilies. Functionally, XAP2 reversibly inhibited the enzymatic activity of PDE4A5, increased the sensitivity of PDE4A5 to inhibition by the prototypical PDE4 inhibitor 4-[3-(cyclopentyloxy)-4-methoxyphenyl]-2-pyrrolidinone (rolipram) and attenuated the ability of cAMP-dependent protein kinase to phosphorylate PDE4A5 in intact cells. XAP2 maximally inhibited PDE4A5 by approximately 60%, with an IC50 of 120 nm, and reduced the IC50 for rolipram from 390 nm to 70-90 nm. Co-expression of XAP2 and PDE4A5 in COS7 cells showed that they could be co-immunoprecipitated and also reduced both the enzymatic activity of PDE4A5 and its IC50 for rolipram. Native XAP2 and PDE4A5 could be co-immunoprecipitated from the brain. The isolated COOH-terminal half of XAP2 (amino acids 170-330), containing its tetratricopeptide repeat domain, but not the isolated NH2-terminal half (amino acids 1-169), containing the immunophilin homology region, similarly reduced PDE4A5 activity and its IC50 for rolipram. Mutation of Arg271 to alanine, in the XAP2 tetratricopeptide repeat region, attenuated its ability to both interact with PDE4A5 in two-hybrid assays and to inhibit PDE4A5 activity. Either the deletion of a specific portion of the unique amino-terminal region or specific mutations in the regulatory UCR2 domain of PDE4A5 attenuated its ability be inhibited by XAP2. We suggest that XAP2 functionally interacts with PDE4A5 in cells.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/chemistry , Cyclic AMP/metabolism , Proteins/chemistry , 1-Methyl-3-isobutylxanthine/pharmacology , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Alanine/chemistry , Amino Acid Sequence , Animals , COS Cells , Cloning, Molecular , Colforsin/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 4 , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Gene Deletion , Glutathione Transferase/metabolism , Humans , Immunoblotting , Inhibitory Concentration 50 , Intracellular Signaling Peptides and Proteins , Molecular Sequence Data , Mutation , Open Reading Frames , Phosphorylation , Precipitin Tests , Protein Binding , Protein Isoforms , Protein Structure, Tertiary , Proteins/metabolism , Rats , Recombinant Fusion Proteins/metabolism , Rolipram/pharmacology , Saccharomyces cerevisiae/metabolism , Sequence Homology, Amino Acid , Time Factors , Two-Hybrid System Techniques
20.
J Biol Chem ; 277(31): 28298-309, 2002 Aug 02.
Article in English | MEDLINE | ID: mdl-11994273

ABSTRACT

Here we identify an 11-residue helical module in the unique N-terminal region of the cyclic AMP-specific phosphodiesterase PDE4A1 that determines association with phospholipid bilayers and shows a profound selectivity for interaction with phosphatidic acid (PA). This module contains a core bilayer insertion unit that is formed by two tryptophan residues, Trp(19) and Trp(20), whose orientation is optimized for bilayer insertion by the Leu(16):Val(17) pairing. Ca(2+), at submicromolar levels, interacts with Asp(21) in this module and serves to gate bilayer insertion, which is completed within 10 ms. Selectivity for interaction with PA is suggested to be achieved primarily through the formation of a charge network of the form (Asp(21-):Ca(2+):PA(2-):Lys(24+)) with overall neutrality at the bilayer surface. This novel phospholipid-binding domain, which we call TAPAS-1 (tryptophan anchoring phosphatidic acid selective-binding domain 1), is here identified as being responsible for membrane association of the PDE4A1 cAMP-specific phosphodiesterase. TAPAS-1 may not only serve as a paradigm for other PA-binding domains but also aid in detecting related phospholipid-binding domains and in generating simple chimeras for conferring membrane association and intracellular targeting on defined proteins.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/chemistry , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Calcium Signaling/physiology , Cyclic AMP/metabolism , Peptide Fragments/metabolism , Phosphatidic Acids/metabolism , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , Amino Acid Sequence , Animals , Binding Sites , COS Cells , Cell Membrane/metabolism , Chloramphenicol O-Acetyltransferase/metabolism , Chlorocebus aethiops , Cloning, Molecular , Cyclic Nucleotide Phosphodiesterases, Type 4 , Kinetics , Lipid Bilayers , Models, Molecular , Mutagenesis, Site-Directed , Peptide Fragments/chemistry , Protein Conformation , Protein Structure, Secondary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Substrate Specificity , Transfection , Tryptophan
SELECTION OF CITATIONS
SEARCH DETAIL
...