Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
J Med Chem ; 61(10): 4635-4640, 2018 05 24.
Article in English | MEDLINE | ID: mdl-29718668

ABSTRACT

We disclose the discovery and X-ray cocrystal data of potent, selective quinazoline inhibitors of PDE1. Inhibitor ( S)-3 readily attains free plasma concentrations above PDE1 IC50 values and has restricted brain access. The racemic compound 3 inhibits >75% of PDE hydrolytic activity in soluble samples of human myocardium, consistent with heightened PDE1 activity in this tissue. These compounds represent promising new tools to probe the value of PDE1 inhibition in the treatment of cardiovascular disease.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 1/antagonists & inhibitors , Drug Discovery , Myocardium/enzymology , Phosphodiesterase Inhibitors/chemistry , Phosphodiesterase Inhibitors/pharmacology , Quinazolines/chemistry , Cyclic AMP/metabolism , Humans , Models, Molecular , Molecular Structure , Protein Conformation
2.
Circ Res ; 117(8): 707-19, 2015 Sep 25.
Article in English | MEDLINE | ID: mdl-26243800

ABSTRACT

RATIONALE: Chronic elevation of 3'-5'-cyclic adenosine monophosphate (cAMP) levels has been associated with cardiac remodeling and cardiac hypertrophy. However, enhancement of particular aspects of cAMP/protein kinase A signaling seems to be beneficial for the failing heart. cAMP is a pleiotropic second messenger with the ability to generate multiple functional outcomes in response to different extracellular stimuli with strict fidelity, a feature that relies on the spatial segregation of the cAMP pathway components in signaling microdomains. OBJECTIVE: How individual cAMP microdomains affect cardiac pathophysiology remains largely to be established. The cAMP-degrading enzymes phosphodiesterases (PDEs) play a key role in shaping local changes in cAMP. Here we investigated the effect of specific inhibition of selected PDEs on cardiac myocyte hypertrophic growth. METHODS AND RESULTS: Using pharmacological and genetic manipulation of PDE activity, we found that the rise in cAMP resulting from inhibition of PDE3 and PDE4 induces hypertrophy, whereas increasing cAMP levels via PDE2 inhibition is antihypertrophic. By real-time imaging of cAMP levels in intact myocytes and selective displacement of protein kinase A isoforms, we demonstrate that the antihypertrophic effect of PDE2 inhibition involves the generation of a local pool of cAMP and activation of a protein kinase A type II subset, leading to phosphorylation of the nuclear factor of activated T cells. CONCLUSIONS: Different cAMP pools have opposing effects on cardiac myocyte cell size. PDE2 emerges as a novel key regulator of cardiac hypertrophy in vitro and in vivo, and its inhibition may have therapeutic applications.


Subject(s)
Cardiomegaly/prevention & control , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Myocytes, Cardiac/enzymology , Second Messenger Systems , Adenoviridae/genetics , Animals , Animals, Newborn , Cardiomegaly/enzymology , Cardiomegaly/genetics , Cardiomegaly/pathology , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 2/antagonists & inhibitors , Cyclic Nucleotide Phosphodiesterases, Type 2/genetics , Disease Models, Animal , Genetic Vectors , Male , Membrane Microdomains/enzymology , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Phosphodiesterase Inhibitors/pharmacology , Phosphorylation , RNA Interference , Rats, Sprague-Dawley , Rats, Wistar , Second Messenger Systems/drug effects , Time Factors , Transduction, Genetic , Transfection
3.
Nat Genet ; 47(6): 647-53, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25961942

ABSTRACT

Cardiovascular disease is the most common cause of death worldwide, and hypertension is the major risk factor. Mendelian hypertension elucidates mechanisms of blood pressure regulation. Here we report six missense mutations in PDE3A (encoding phosphodiesterase 3A) in six unrelated families with mendelian hypertension and brachydactyly type E (HTNB). The syndrome features brachydactyly type E (BDE), severe salt-independent but age-dependent hypertension, an increased fibroblast growth rate, neurovascular contact at the rostral-ventrolateral medulla, altered baroreflex blood pressure regulation and death from stroke before age 50 years when untreated. In vitro analyses of mesenchymal stem cell-derived vascular smooth muscle cells (VSMCs) and chondrocytes provided insights into molecular pathogenesis. The mutations increased protein kinase A-mediated PDE3A phosphorylation and resulted in gain of function, with increased cAMP-hydrolytic activity and enhanced cell proliferation. Levels of phosphorylated VASP were diminished, and PTHrP levels were dysregulated. We suggest that the identified PDE3A mutations cause the syndrome. VSMC-expressed PDE3A deserves scrutiny as a therapeutic target for the treatment of hypertension.


Subject(s)
Brachydactyly/genetics , Cyclic Nucleotide Phosphodiesterases, Type 3/genetics , Hypertension/congenital , Adolescent , Adult , Amino Acid Sequence , Animals , Base Sequence , Case-Control Studies , Cell Differentiation , Child , Female , Genetic Association Studies , HeLa Cells , Humans , Hypertension/genetics , Kinetics , Male , Mesenchymal Stem Cells/physiology , Mice , Middle Aged , Molecular Sequence Data , Mutation, Missense , Myocytes, Smooth Muscle/physiology , Pedigree
4.
J Biol Chem ; 290(11): 6763-76, 2015 Mar 13.
Article in English | MEDLINE | ID: mdl-25593322

ABSTRACT

Cyclic nucleotide phosphodiesterase 3A (PDE3) regulates cAMP-mediated signaling in the heart, and PDE3 inhibitors augment contractility in patients with heart failure. Studies in mice showed that PDE3A, not PDE3B, is the subfamily responsible for these inotropic effects and that murine PDE3A1 associates with sarcoplasmic reticulum Ca(2+) ATPase 2 (SERCA2), phospholamban (PLB), and AKAP18 in a multiprotein signalosome in human sarcoplasmic reticulum (SR). Immunohistochemical staining demonstrated that PDE3A co-localizes in Z-bands of human cardiac myocytes with desmin, SERCA2, PLB, and AKAP18. In human SR fractions, cAMP increased PLB phosphorylation and SERCA2 activity; this was potentiated by PDE3 inhibition but not by PDE4 inhibition. During gel filtration chromatography of solubilized SR membranes, PDE3 activity was recovered in distinct high molecular weight (HMW) and low molecular weight (LMW) peaks. HMW peaks contained PDE3A1 and PDE3A2, whereas LMW peaks contained PDE3A1, PDE3A2, and PDE3A3. Western blotting showed that endogenous HMW PDE3A1 was the principal PKA-phosphorylated isoform. Phosphorylation of endogenous PDE3A by rPKAc increased cAMP-hydrolytic activity, correlated with shift of PDE3A from LMW to HMW peaks, and increased co-immunoprecipitation of SERCA2, cav3, PKA regulatory subunit (PKARII), PP2A, and AKAP18 with PDE3A. In experiments with recombinant proteins, phosphorylation of recombinant human PDE3A isoforms by recombinant PKA catalytic subunit increased co-immunoprecipitation with rSERCA2 and rat rAKAP18 (recombinant AKAP18). Deletion of the recombinant human PDE3A1/PDE3A2 N terminus blocked interactions with recombinant SERCA2. Serine-to-alanine substitutions identified Ser-292/Ser-293, a site unique to human PDE3A1, as the principal site regulating its interaction with SERCA2. These results indicate that phosphorylation of human PDE3A1 at a PKA site in its unique N-terminal extension promotes its incorporation into SERCA2/AKAP18 signalosomes, where it regulates a discrete cAMP pool that controls contractility by modulating phosphorylation-dependent protein-protein interactions, PLB phosphorylation, and SERCA2 activity.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Myocardium/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , A Kinase Anchor Proteins/analysis , A Kinase Anchor Proteins/metabolism , Calcium/metabolism , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/analysis , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/analysis , Humans , Myocardium/cytology , Myocardium/enzymology , Myocardium/ultrastructure , Phosphorylation , Protein Interaction Maps , Protein Isoforms/analysis , Protein Isoforms/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/analysis
5.
Proc Natl Acad Sci U S A ; 110(49): 19778-83, 2013 Dec 03.
Article in English | MEDLINE | ID: mdl-24248367

ABSTRACT

Inhibitors of cyclic nucleotide phosphodiesterase (PDE) PDE3A have inotropic actions in human myocardium, but their long-term use increases mortality in patients with heart failure. Two isoforms in cardiac myocytes, PDE3A1 and PDE3A2, have identical amino acid sequences except for a unique N-terminal extension in PDE3A1. We expressed FLAG-tagged PDE3A1 and PDE3A2 in HEK293 cells and examined their regulation by PKA- and PKC-mediated phosphorylation. PDE3A1, which is localized to intracellular membranes, and PDE3A2, which is cytosolic, were phosphorylated at different sites within their common sequence. Exposure to isoproterenol led to phosphorylation of PDE3A1 at the 14-3-3-binding site S312, whereas exposure to PMA led to phosphorylation of PDE3A2 at an alternative 14-3-3-binding site, S428. PDE3A2 activity was stimulated by phosphorylation at S428, whereas PDE3A1 activity was not affected by phosphorylation at either site. Phosphorylation of PDE3A1 by PKA and of PDE3A2 by PKC led to shifts in elution on gel-filtration chromatography consistent with increased interactions with other proteins, and 2D electrophoresis of coimmunoprecipitated proteins revealed that the two isoforms have distinct protein interactomes. A similar pattern of differential phosphorylation of endogenous PDE3A1 and PDE3A2 at S312 and S428 is observed in human myocardium. The selective phosphorylation of PDE3A1 and PDE3A2 at alternative sites through different signaling pathways, along with the different functional consequences of phosphorylation for each isoform, suggest they are likely to have distinct roles in cyclic nucleotide-mediated signaling in human myocardium, and raise the possibility that isoform-selective inhibition may allow inotropic responses without an increase in mortality.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Myocardial Contraction/drug effects , Myocytes, Cardiac/metabolism , Phosphodiesterase 3 Inhibitors/pharmacology , 14-3-3 Proteins/genetics , Binding Sites/genetics , Chromatography, Gel , Cyclic AMP-Dependent Protein Kinases/metabolism , Electrophoresis, Gel, Two-Dimensional , Enzyme Activation/physiology , HEK293 Cells , Humans , Immunoprecipitation , Isoenzymes/metabolism , Isoproterenol/pharmacology , Phosphodiesterase 3 Inhibitors/metabolism , Phosphorylation , Protein Kinase C/metabolism
6.
Curr Opin Pharmacol ; 11(6): 707-13, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21962613

ABSTRACT

In dilated cardiomyopathy, a condition characterized by chamber enlargement and reduced myocardial contractility, decreases in ß-adrenergic receptor density and increases in Gαi and ß-adrenergic receptor kinase activities attenuate the stimulation of adenylyl cyclase in response to catecholamines. PDE3 inhibitors have been used to 'overcome' the reduction in cAMP generation by blocking cAMP hydrolysis. These drugs increase contractility in the short-term, but long-term administration leads to an increase in mortality that correlates with an increase in sudden cardiac death. Whether separate mechanisms account for these beneficial and harmful effects, and, if so, whether PDE3 can be targeted so as to increase contractility without increasing mortality are questions that remain unanswered.


Subject(s)
Cardiomyopathy, Dilated/drug therapy , Molecular Targeted Therapy , Phosphodiesterase 3 Inhibitors/therapeutic use , Animals , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/physiopathology , Cyclic AMP/antagonists & inhibitors , Cyclic AMP/physiology , Cyclic Nucleotide Phosphodiesterases, Type 3/chemistry , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Heart/drug effects , Heart/physiopathology , Humans , Myocardial Contraction/drug effects , Myocardium/enzymology , Myocardium/metabolism , Phosphodiesterase 3 Inhibitors/adverse effects , Phosphodiesterase 3 Inhibitors/pharmacology , Second Messenger Systems/drug effects
7.
J Pharmacol Exp Ther ; 330(3): 884-91, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19546307

ABSTRACT

In mouse models of cardiac disease, the type 5 (PDE5)-selective cyclic nucleotide phosphodiesterase inhibitor sildenafil has antihypertrophic and cardioprotective effects attributable to the inhibition of cGMP hydrolysis. To investigate the relevance of these findings to humans, we quantified cGMP-hydrolytic activity and its inhibition by sildenafil in cytosolic and microsomal preparations from the left ventricular myocardium of normal and failing human hearts. The vast majority of cGMP-hydrolytic activity was attributable to PDE1 and PDE3. Sildenafil had no measurable effect on cGMP hydrolysis at 10 nM, at which it is selective for PDE5, but it had a marked effect on cGMP and cAMP hydrolysis at 1 microM, at which it inhibits PDE1. In contrast, in preparations from the left ventricles of normal mice and mice with heart failure resulting from coronary artery ligation, the effects of sildenafil on cGMP hydrolysis were attributable to inhibition of both PDE5 and PDE1; PDE5 comprised approximately 22 and approximately 43% of the cytosolic cGMP-hydrolytic activity in preparations from normal and failing mouse hearts, respectively. These differences in PDE5 activities in human and mouse hearts call into question the extent to which the effects of sildenafil in mouse models are likely to be applicable in humans and raise the possibility of PDE1 as an alternative therapeutic target.


Subject(s)
Cyclic AMP/metabolism , Heart Failure/metabolism , Myocardium/metabolism , Phosphodiesterase Inhibitors/pharmacology , Piperazines/pharmacology , Sulfones/pharmacology , Animals , Coronary Vessels/physiology , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Cytosol/drug effects , Cytosol/metabolism , Humans , Hydrolysis , Male , Mice , Mice, Inbred ICR , Microsomes/drug effects , Microsomes/metabolism , Phosphodiesterase 3 Inhibitors , Phosphodiesterase 5 Inhibitors , Phosphodiesterase I/antagonists & inhibitors , Phosphodiesterase I/metabolism , Purines/pharmacology , Sildenafil Citrate , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism
8.
Heart Fail Rev ; 14(4): 255-63, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19096931

ABSTRACT

Drugs that inhibit cyclic nucleotide phosphodiesterase activity act to increase intracellular cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) content. In total, 11 families of these enzymes-which differ with respect to affinity for cAMP and cGMP, cellular expression, intracellular localization, and mechanisms of regulation-have been identified. Inhibitors of enzymes in the PDE3 family of cyclic nucleotide phosphodiesterases raise intracellular cAMP content in cardiac and vascular smooth muscle, with inotropic and, to a lesser extent, vasodilatory actions. These drugs have been used for many years in the treatment of patients with heart failure, but their long-term use has generally been shown to increase mortality through mechanisms that remain unclear. More recently, inhibitors of PDE5 cyclic nucleotide phosphodiesterases have been used as cGMP-raising agents in vascular smooth muscle. With respect to cardiovascular disease, there is evidence that these drugs are more efficacious in the pulmonary than in the systemic vasculature, for which reason they are used principally in patients with pulmonary hypertension. Effects attributable to inhibition of myocardial PDE5 activity are less well characterized. New information indicating that enzymes from the PDE1 family of cyclic nucleotide phosphodiesterases constitute the majority of cAMP- and cGMP-hydrolytic activity in human myocardium raises questions as to their role in regulating these signaling pathways in heart failure.


Subject(s)
Heart Failure/drug therapy , Phosphodiesterase Inhibitors/therapeutic use , 3',5'-Cyclic-AMP Phosphodiesterases/drug effects , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 1/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Heart Failure/physiopathology , Hemodynamics/drug effects , Humans , Piperazines/pharmacology , Piperazines/therapeutic use , Purines/pharmacology , Purines/therapeutic use , Sildenafil Citrate , Sulfones/pharmacology , Sulfones/therapeutic use , Treatment Outcome
9.
Chembiochem ; 9(11): 1757-66, 2008 Jul 21.
Article in English | MEDLINE | ID: mdl-18574825

ABSTRACT

Novel benzene polyphosphates were synthesised as inositol polyphosphate mimics and evaluated against type-I inositol 1,4,5-trisphosphate 5-phosphatase, which only binds soluble inositol polyphosphates, and against the PH domain of protein kinase Balpha (PKBalpha), which can bind both soluble inositol polyphosphates and inositol phospholipids. The most potent trisphosphate 5-phosphatase inhibitor is benzene 1,2,4-trisphosphate (2, IC(50) of 14 microM), a potential mimic of D-myo-inositol 1,4,5-trisphosphate, whereas the most potent tetrakisphosphate Ins(1,4,5)P(3) 5-phosphatase inhibitor is benzene 1,2,4,5-tetrakisphosphate, with an IC(50) of 4 microM. Biphenyl 2,3',4,5',6-pentakisphosphate (4) was the most potent inhibitor evaluated against type I Ins(1,4,5)P(3) 5-phosphatase (IC(50) of 1 microM). All new benzene polyphosphates are resistant to dephosphorylation by type I Ins(1,4,5)P(3) 5-phosphatase. Unexpectedly, all benzene polyphosphates studied bind to the PH domain of PKBalpha with apparent higher affinity than to type I Ins(1,4,5)P(3) 5-phosphatase. The most potent ligand for the PKBalpha PH domain, measured by inhibition of biotinylated diC(8)-PtdIns(3,4)P(2) binding, is biphenyl 2,3',4,5',6-pentakisphosphate (4, K(i)=27 nm). The approximately 80-fold enhancement of binding relative to parent benzene trisphosphate is explained by the involvement of a cation-pi interaction. These new molecular tools will be of potential use in structural and cell signalling studies.


Subject(s)
Benzene/chemistry , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Polyphosphates/chemistry , Polyphosphates/pharmacology , Proto-Oncogene Proteins c-akt/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Binding Sites , Fluorescence Resonance Energy Transfer , Inositol Polyphosphate 5-Phosphatases , Ligands , Models, Molecular , Phosphoric Monoester Hydrolases/chemistry , Phosphoric Monoester Hydrolases/metabolism , Polyphosphates/chemical synthesis , Protein Structure, Tertiary , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Stereoisomerism , Structure-Activity Relationship
10.
J Biol Chem ; 282(45): 32749-57, 2007 Nov 09.
Article in English | MEDLINE | ID: mdl-17726023

ABSTRACT

Isoforms in the PDE1 family of cyclic nucleotide phosphodiesterases were recently found to comprise a significant portion of the cGMP-inhibited cAMP hydrolytic activity in human hearts. We examined the expression of PDE1 isoforms in human myocardium, characterized their catalytic activity, and quantified their contribution to cAMP hydrolytic and cGMP hydrolytic activity in subcellular fractions of this tissue. Western blotting with isoform-selective anti-PDE1 monoclonal antibodies showed PDE1C1 to be the principal isoform expressed in human myocardium. Immunohistochemical analysis showed that PDE1C1 is distributed along the Z-lines and M-lines of cardiac myocytes in a striated pattern that differs from that of the other major dual-specificity cyclic nucleotide phosphodiesterase in human myocardium, PDE3A. Most of the PDE1C1 activity was recovered in soluble fractions of human myocardium. It binds both cAMP and cGMP with K(m) values of approximately 1 microm and hydrolyzes both substrates with similar catalytic rates. PDE1C1 activity in subcellular fractions was quantified using a new PDE1-selective inhibitor, IC295. At substrate concentrations of 0.1 microm, PDE1C1 constitutes the great majority of cAMP hydrolytic and cGMP hydrolytic activity in soluble fractions and the majority of cGMP hydrolytic activity in microsomal fractions, whereas PDE3 constitutes the majority of cAMP hydrolytic activity in microsomal fractions. These results indicate that PDE1C1 is expressed at high levels in human cardiac myocytes with an intracellular distribution distinct from that of PDE3A and that it may have a role in the integration of cGMP-, cAMP- and Ca(2+)-mediated signaling in these cells.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Myocytes, Cardiac/enzymology , Animals , Cell Line , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 1/genetics , Gene Expression Regulation, Enzymologic , Humans , Hydrolysis , Isoenzymes/metabolism , Kinetics , Spodoptera , Subcellular Fractions/enzymology
11.
FASEB J ; 21(7): 1481-91, 2007 May.
Article in English | MEDLINE | ID: mdl-17264160

ABSTRACT

Benzene polyphosphates containing phosphate groups on one ring are Ins(1,4,5)P3 5-phosphatase inhibitors when evaluated against type-I Ins(1,4,5)P3 5-phosphatase. A novel biphenyl derivative, biphenyl 2,3',4,5',6-pentakisphosphate, with five phosphate groups on two rings was synthesized: It inhibited the activity of two inositol 5-phosphatases: type I and SHIP2 with Ins(1,3,4,5)P4 as substrate. The inhibition was competitive with respect to the substrate. IC50 value measured in rat hepatocytes, which contains the native Ins(1,4,5)P3 5-phosphatase, was in the micromolar range at 1.0 microM Ins(1,4,5)P3 as substrate. Biphenyl 2,3',4,5',6-pentakisphosphate did not affect the activity of Ins(1,4,5)P3 3-kinase A in the 5-100 microM range. Surprisingly, experimental evidence supports an effect of biphenyl 2,3',4,5',6-pentakisphosphate at the level of the Ins(1,4,5)P3 receptor. Finally, when injected into rat hepatocytes, the analog affected the frequency of Ca2+ oscillations in a positive or negative way depending on its concentration. At very high concentrations of the analog, Ca2+ oscillations were even suppressed. These data were interpreted as a dual effect of the biphenyl 2,3',4,5',6-pentakisphosphate on cytosolic [Ca2+] increases: an activation effect through an increase in Ins(1,4,5)P3 level via Ins(1,4,5)P3 5-phosphatase inhibition and an inhibitory effect, which was exerted directly on the Ins(1,4,5)P3 receptor. Thus, our data show for the first time that the frequency of Ca2+ oscillations in response to a Ca2+-mobilizing agonist can be controlled by inhibitors of type-I Ins(1,4,5)P3 5-phosphatase.


Subject(s)
Biphenyl Compounds/pharmacology , Calcium/metabolism , Hepatocytes/drug effects , Phosphates/pharmacology , Animals , Cell Line , Female , Hepatocytes/metabolism , Microinjections , Rats , Rats, Wistar , Spodoptera
12.
Chembiochem ; 7(11): 1696-706, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16964627

ABSTRACT

3-Hydroxybenzene 1,2,4-trisphosphate 4 is a new myo-inositol 1,4,5-trisphosphate analogue based on the core structure of benzene 1,2,4-trisphosphate 2 with an additional hydroxyl group at position-3, and is the first noninositol based compound to be a substrate for inositol 1,4,5-trisphosphate 5-phosphatase. In physicochemical studies on 2, when three equivalents of protons were added, the (31)P NMR spectrum displayed monophasic behaviour in which phosphate-1 and phosphate-2 behaved independently in most of the studied pH range. For compound 4, phosphate-2 and phosphate-4 interacted with the 3-OH group, which does not titrate at physiological pH, displaying complex biphasic behaviour which demonstrated co-operativity between these groups. Phosphate-1 and phosphate-2 strongly interacted with each other and phosphate-4 experienced repulsion because of the interaction of the 3-OH group. Benzene 1,2,4-trisphosphate 2 is resistant to inositol 1,4,5-trisphosphate type I 5-phosphatase catalysed dephosphorylation. However, surprisingly, 3-hydroxybenzene 1,2,4-trisphosphate 4 was dephosphorylated by this 5-phosphatase to give the symmetrical 2,3-dihydroxybenzene 1,4-bisphosphate 16. The extra hydroxyl group is shown to form a hydrogen bond with the vicinal phosphate groups at -15 degrees C, and (1)H NMR titration of the ring and hydroxyl protons in 4 shows the OH proton to be strongly stabilized as soon as the phosphate groups are deprotonated. The effect of the phenolic 3-OH group in compound 4 confirms a critical role for the 6-OH group of the natural messenger in the dephosphorylation mechanism that persists even in radically modified analogues.


Subject(s)
Molecular Mimicry , Organophosphates/chemical synthesis , Organophosphates/metabolism , Phosphoric Monoester Hydrolases/metabolism , Second Messenger Systems , Chemical Phenomena , Chemistry, Physical , Hydrogen-Ion Concentration , Inositol Polyphosphate 5-Phosphatases , Magnetic Resonance Spectroscopy , Molecular Structure , Organophosphates/chemistry , Phosphates/chemistry , Phosphorylation , Polyphosphates/chemistry , Protons , Rosaniline Dyes/chemistry , Rosaniline Dyes/metabolism , Substrate Specificity , Titrimetry
13.
Cell Signal ; 18(12): 2193-9, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16824732

ABSTRACT

The SH2 domain containing inositol 5-phosphatase 2 (SHIP2) catalyzes the dephosphorylation of phosphatidylinositol 3,4,5-trisphosphate (PtdIns(3,4,5)P(3)) to phosphatidylinositol 3,4-bisphosphate (PtdIns(3,4)P(2)) and participates in the insulin signalling pathway in vivo. In a comparative study of SHIP2 and the phosphatase and tensin homologue deleted on chromosome 10 (PTEN), we found that their lipid phosphatase activity was influenced by the presence of vesicles of phosphatidylserine (PtdSer). SHIP2 PtdIns(3,4,5)P(3) 5-phosphatase activity was greatly stimulated in the presence of vesicles of PtdSer. This effect appears to be specific for di-C8 and di-C16 fatty acids of PtdIns(3,4,5)P(3) as substrate. It was not observed with inositol 1,3,4,5-tetrakisphosphate (Ins(1,3,4,5)P(4)) another in vitro substrate of SHIP2, nor with Type I Ins(1,4,5)P(3)/Ins(1,3,4,5)P(4) 5-phosphatase activity, an enzyme which acts on soluble inositol phosphates. Vesicles of phosphatidylcholine (PtdCho) stimulated only twofold PtdIns(3,4,5)P(3) 5-phosphatase activity of SHIP2. Both a minimal catalytic construct and the full length SHIP2 were sensitive to the stimulation by PtdSer. In contrast, PtdIns(3,4,5)P(3) 5-phosphatase activity of the Skeletal muscle and Kidney enriched Inositol Phosphatase (SKIP), another member of the mammaliam Type II phosphoinositide 5-phosphatases, was not sensitive to PtdSer. Our enzymatic data establish a specificity in the control of SHIP2 lipid phosphatase activity with PtdIns(3,4,5)P(3) as substrate which is depending on the fatty acid composition of the substrate.


Subject(s)
Phosphatidylinositol Phosphates/metabolism , Phosphatidylserines/metabolism , Phosphoric Monoester Hydrolases/metabolism , Catalysis , Electrophoresis, Polyacrylamide Gel , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Humans , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/genetics , Phosphorylation , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Substrate Specificity , Transport Vesicles/metabolism
14.
Mol Cell Endocrinol ; 243(1-2): 58-65, 2005 Nov 24.
Article in English | MEDLINE | ID: mdl-16225984

ABSTRACT

In dog thyrocytes in primary culture, thyrotropin (TSH), through cAMP, positively controls proliferation and differentiation. As until now, the key events and the genes involved in the action of TSH remain largely uncharacterized, our goal was to identify new differentially expressed genes in TSH-induced thyroid proliferation. Using cDNA-AFLP, we visualized 105 different transcripts showing significant differential expression during the stimulation of dog thyrocytes with TSH for different times, in the presence of insulin. Northern blot and RT-PCR analyses confirmed the pattern expression of 5 clones encoding known proteins: thrombospondine 1, TNFr1, RhoE, RalB, and annexin A2. These regulations provide molecular counterparts of in vivo physiological effects of TSH: angiogenesis (decreased thrombospondin 1), decreased apoptosis (decreased TNFR1) and actin filament disruption, macropinocytosis and thyroid hormone secretion (decreased RhoE).


Subject(s)
Gene Expression Profiling , Thyroid Gland/metabolism , Thyrotropin/physiology , Animals , Annexin A2/metabolism , Cells, Cultured , DNA, Complementary/genetics , Dogs , GTPase-Activating Proteins/metabolism , Insulin/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Thrombospondins/metabolism , Thyroid Gland/cytology , Thyrotropin/pharmacology
15.
Endocrinology ; 144(4): 1341-9, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12639917

ABSTRACT

We have investigated the role of the different classes of MAPKs, i.e. ERKs, c-Jun N-terminal kinases (JNKs), and p38 MAPK in the proliferation of dog and human thyroid epithelial cells (thyrocytes) in primary cultures. In these cells, TSH, acting through cAMP, epidermal growth factor, hepatocyte growth factor (HGF), and phorbol 12-myristate 13-acetate induce DNA synthesis. With the exception of HGF, all of these factors require the presence of insulin for mitogenic effects to be expressed. We found that TSH and forskolin are without effect on the phosphorylation and activity of the different classes of MAPKs. In contrast, all the cAMP-independent growth factors, whereas without effect on the phosphorylation and activity of JNKs and p38 MAPK, stimulated the ERKs. This effect was strong and sustained in response to HGF, epidermal growth factor and 12-myristate 13-acetate but weak and transient in response to insulin. Moreover, whereas in stimulated cells DNA synthesis was inhibited by PD 098059, an inhibitor of MAPK kinase 1 and consequently of ERKs, it was not modified by SB 203580, an inhibitor of p38 MAPK. Taken together, these data 1) exclude a role of JNKs and p38 MAPK in the proliferation of dog and human thyrocytes; 2) suggest that the mitogenic action of the cAMP-independent agents requires a strong and sustained activation of both ERKs and phosphatidylinositol 3-kinase/protein kinase B as realized by HGF alone or by the other agents together with insulin; and 3) show that TSH and cAMP do not activate ERKs but that the weak activation of ERKs by insulin is nevertheless necessary for DNA synthesis to occur.


Subject(s)
Cyclic AMP/metabolism , Epidermal Growth Factor/pharmacology , Hepatocyte Growth Factor/pharmacology , MAP Kinase Signaling System/physiology , Thyroid Gland/cytology , Animals , Arsenites/pharmacology , Carcinogens/pharmacology , Cell Division/drug effects , Cell Division/physiology , Cells, Cultured , Colforsin/pharmacology , Dogs , Enzyme Inhibitors/pharmacology , Epithelial Cells/cytology , Epithelial Cells/enzymology , Flavonoids/pharmacology , Humans , Hypoglycemic Agents/pharmacology , Imidazoles/pharmacology , Insulin/pharmacology , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Pyridines/pharmacology , Sodium Compounds/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Thyrotropin/pharmacology , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...