Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Commun Biol ; 3(1): 596, 2020 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-33087821

RESUMEN

Programmed degradation of mitochondria by mitophagy, an essential process to maintain mitochondrial homeostasis, is not completely understood. Here we uncover a regulatory process that controls mitophagy and involves the cAMP-degrading enzyme phosphodiesterase 2A2 (PDE2A2). We find that PDE2A2 is part of a mitochondrial signalosome at the mitochondrial inner membrane where it interacts with the mitochondrial contact site and organizing system (MICOS). As part of this compartmentalised signalling system PDE2A2 regulates PKA-mediated phosphorylation of the MICOS component MIC60, resulting in modulation of Parkin recruitment to the mitochondria and mitophagy. Inhibition of PDE2A2 is sufficient to regulate mitophagy in the absence of other triggers, highlighting the physiological relevance of PDE2A2 in this process. Pharmacological inhibition of PDE2 promotes a 'fat-burning' phenotype to retain thermogenic beige adipocytes, indicating that PDE2A2 may serve as a novel target with potential for developing therapies for metabolic disorders.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/metabolismo , Mitocondrias/metabolismo , Mitofagia , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Línea Celular , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/química , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/genética , Técnica del Anticuerpo Fluorescente , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Ratones , Mitocondrias/genética , Proteínas Mitocondriales/metabolismo , Mitofagia/genética , Fosforilación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estabilidad Proteica , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/genética
2.
Molecules ; 24(15)2019 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-31370274

RESUMEN

Phosphodiesterase 2A (PDE2A) is highly expressed in distinct areas of the brain, which are known to be related to neuropsychiatric diseases. The development of suitable PDE2A tracers for Positron Emission Tomography (PET) would permit the in vivo imaging of the PDE2A and evaluation of disease-mediated alterations of its expression. A series of novel fluorinated PDE2A inhibitors on the basis of a Benzoimidazotriazine (BIT) scaffold was prepared leading to a prospective inhibitor for further development of a PDE2A PET imaging agent. BIT derivatives (BIT1-9) were obtained by a seven-step synthesis route, and their inhibitory potency towards PDE2A and selectivity over other PDEs were evaluated. BIT1 demonstrated much higher inhibition than other BIT derivatives (82.9% inhibition of PDE2A at 10 nM). BIT1 displayed an IC50 for PDE2A of 3.33 nM with 16-fold selectivity over PDE10A. This finding revealed that a derivative bearing both a 2-fluoro-pyridin-4-yl and 2-chloro-5-methoxy-phenyl unit at the 8- and 1-position, respectively, appeared to be the most potent inhibitor. In vitro studies of BIT1 using mouse liver microsomes (MLM) disclosed BIT1 as a suitable ligand for 18F-labeling. Nevertheless, future in vivo metabolism studies are required.


Asunto(s)
Encéfalo/enzimología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/antagonistas & inhibidores , Radiofármacos/química , Triazinas/síntesis química , Animales , Encéfalo/diagnóstico por imagen , Encéfalo/efectos de los fármacos , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/química , Humanos , Ligandos , Ratones , Tomografía de Emisión de Positrones , Radiofármacos/síntesis química , Radiofármacos/farmacología , Triazinas/química , Triazinas/farmacología
3.
Biochemistry ; 57(42): 6070-6077, 2018 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-30231198

RESUMEN

The cAMP signaling system plays important roles in the physiological processes of pathogen yeast Candida albicans, but its functional mechanism has not been well illustrated. Here, we report the enzymatic characterization and crystal structures of C. albicans phosphodiesterase 2 (caPDE2) in the unliganded and 3-isobutyl-1-methylxanthine-complexed forms. caPDE2 is a monomer in liquid and crystal states and specifically hydrolyzes cAMP with a KM of 35 nM. It does not effectively hydrolyze cGMP as shown by the 1.32 × 105-fold specificity of cAMP/cGMP. The crystal structure of caPDE2 shows significant differences from those of human PDEs. First, the N-terminal fragment of caPDE2 (residues 1-201) tightly associates with the catalytic domain to form a rigid molecular entity, implying its stable molecular conformation for C. albicans to resist environmental stresses. Second, the M-loop, a critical fragment for binding of the substrate and inhibitors to human PDEs, is not a part of the caPDE2 active site. This feature of caPDE2 may provide a structural basis for the design of selective inhibitors for the treatment of yeast infection.


Asunto(s)
Candida albicans/enzimología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/química , Proteínas Fúngicas/química , Cristalografía por Rayos X , Dominios Proteicos , Estructura Secundaria de Proteína , Relación Estructura-Actividad
4.
J Med Chem ; 61(17): 7754-7766, 2018 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-30070482

RESUMEN

In medicinal chemistry, accurate prediction of additivity-based structure-activity relationship (SAR) analysis rests on three assumptions: (1) a consistent binding pose of the central scaffold, (2) no interaction between the R group substituents, and (3) a relatively rigid binding pocket in which the R group substituents act independently. Previously, examples of nonadditive SAR have been documented in systems that deviate from the first two assumptions. Local protein structural change upon ligand binding, through induced fit or conformational selection, although a well-known phenomenon that invalidates the third assumption, has not been linked to nonadditive SAR conclusively. Here, for the first time, we present clear structural evidence that the formation of a hydrophobic pocket upon ligand binding in PDE2 catalytic site reduces the size of another distinct subpocket and contributes to strong nonadditive SAR between two otherwise distant R groups.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/química , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/metabolismo , Inhibidores Enzimáticos/farmacología , Modelos Teóricos , Conformación Proteica , Quinazolinas/química , Triazoles/química , Sitios de Unión , Cristalografía por Rayos X , Inhibidores Enzimáticos/química , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Ligandos , Unión Proteica , Relación Estructura-Actividad
5.
Sci Rep ; 8(1): 4883, 2018 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-29559702

RESUMEN

A congeneric series of 21 phosphodiesterase 2 (PDE2) inhibitors are reported. Crystal structures show how the molecules can occupy a 'top-pocket' of the active site. Molecules with small substituents do not enter the pocket, a critical leucine (Leu770) is closed and water molecules are present. Large substituents enter the pocket, opening the Leu770 conformation and displacing the waters. We also report an X-ray structure revealing a new conformation of the PDE2 active site domain. The relative binding affinities of these compounds were studied with free energy perturbation (FEP) methods and it represents an attractive real-world test case. In general, the calculations could predict the energy of small-to-small, or large-to-large molecule perturbations. However, accurately capturing the transition from small-to-large proved challenging. Only when using alternative protein conformations did results improve. The new X-ray structure, along with a modelled dimer, conferred stability to the catalytic domain during the FEP molecular dynamics (MD) simulations, increasing the convergence and thereby improving the prediction of ΔΔG of binding for some small-to-large transitions. In summary, we found the most significant improvement in results when using different protein structures, and this data set is useful for future free energy validation studies.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/antagonistas & inhibidores , Inhibidores de Fosfodiesterasa/química , Inhibidores de Fosfodiesterasa/metabolismo , Sitios de Unión , Fenómenos Biofísicos , Cristalografía por Rayos X/métodos , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/química , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/metabolismo , Entropía , Simulación de Dinámica Molecular , Unión Proteica , Conformación Proteica , Proteínas , Relación Estructura-Actividad , Termodinámica
6.
J Med Chem ; 61(3): 1001-1018, 2018 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-29293004

RESUMEN

Computational modeling was used to direct the synthesis of analogs of previously reported phosphodiesterase 2A (PDE2A) inhibitor 1 with an imidazotriazine core to yield compounds of significantly enhanced potency. The analog PF-05180999 (30) was subsequently identified as a preclinical candidate targeting cognitive impairment associated with schizophrenia. Compound 30 demonstrated potent binding to PDE2A in brain tissue, dose responsive mouse brain cGMP increases, and reversal of N-methyl-d-aspartate (NMDA) antagonist-induced (MK-801, ketamine) effects in electrophysiology and working memory models in rats. Preclinical pharmacokinetics revealed unbound brain/unbound plasma levels approaching unity and good oral bioavailability resulting in an average concentration at steady state (Cav,ss) predicted human dose of 30 mg once daily (q.d.). Modeling of a modified release formulation suggested that 25 mg twice daily (b.i.d.) could maintain plasma levels of 30 at or above targeted efficacious plasma levels for 24 h, which became part of the human clinical plan.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/antagonistas & inhibidores , Descubrimiento de Drogas , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Animales , Disponibilidad Biológica , Encéfalo/fisiología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/química , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacocinética , Humanos , Imidazoles/química , Imidazoles/metabolismo , Imidazoles/farmacocinética , Imidazoles/farmacología , Concentración 50 Inhibidora , Memoria a Corto Plazo/efectos de los fármacos , Simulación del Acoplamiento Molecular , Conformación Proteica
7.
Structure ; 25(12): 1887-1897.e4, 2017 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-29107484

RESUMEN

The concentration of messenger molecules in bacterial cells needs to be tightly regulated. This can be achieved by either controlling the synthesis rate, degradation, or export by specific transporters, respectively. The regulation of the essential second messenger c-di-AMP is achieved by modulation of the diadenylate cyclase activity as well as by specific phosphodiesterases that hydrolyze c-di-AMP in the cell. We provide here structural and biochemical data on the DHH-type phosphodiesterase TmPDE (TM1595) from Thermotoga maritima. Our analysis shows that TmPDE is preferentially degrading linear dinucleotides, such as 5'-pApA, 5'-pGpG, and 5'-pApG, compared with cyclic dinucleotide substrates. The high-resolution structural data provided here describe all steps of the PDE reaction: the ligand-free enzyme, two substrate-bound states, and three post-reaction states. We can furthermore show that Pde2 from Streptococcus pneumoniae shares both structural features and substrate specificity based on small-angle X-ray scattering data and biochemical assays.


Asunto(s)
Proteínas Bacterianas/química , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/química , Thermotoga maritima/enzimología , Proteínas Bacterianas/metabolismo , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/metabolismo , Especificidad por Sustrato
8.
J Med Chem ; 60(18): 7677-7702, 2017 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-28796496

RESUMEN

Phosphodiesterase (PDE) 2A inhibitors have emerged as a novel mechanism with potential therapeutic option to ameliorate cognitive dysfunction in schizophrenia or Alzheimer's disease through upregulation of cyclic nucleotides in the brain and thereby achieve potentiation of cyclic nucleotide signaling pathways. This article details the expedited optimization of our recently disclosed pyrazolo[1,5-a]pyrimidine lead compound 4b, leading to the discovery of clinical candidate 36 (TAK-915), which demonstrates an appropriate combination of potency, PDE selectivity, and favorable pharmacokinetic (PK) properties, including brain penetration. Successful identification of 36 was realized through application of structure-based drug design (SBDD) to further improve potency and PDE selectivity, coupled with prospective design focused on physicochemical properties to deliver brain penetration. Oral administration of 36 demonstrated significant elevation of 3',5'-cyclic guanosine monophosphate (cGMP) levels in mouse brains and improved cognitive performance in a novel object recognition task in rats. Consequently, compound 36 was advanced into human clinical trials.


Asunto(s)
Encéfalo/efectos de los fármacos , Cognición/efectos de los fármacos , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/antagonistas & inhibidores , Inhibidores de Fosfodiesterasa/farmacología , Inhibidores de Fosfodiesterasa/farmacocinética , Pirazinas/farmacología , Pirazinas/farmacocinética , Animales , Encéfalo/metabolismo , Trastornos del Conocimiento/tratamiento farmacológico , Trastornos del Conocimiento/metabolismo , Cristalografía por Rayos X , GMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/química , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/metabolismo , Diseño de Fármacos , Halogenación , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Inhibidores de Fosfodiesterasa/química , Pirazinas/química , Pirazoles/química , Pirazoles/farmacocinética , Pirazoles/farmacología , Pirimidinas/química , Pirimidinas/farmacocinética , Pirimidinas/farmacología , Ratas , Ratas Sprague-Dawley
9.
J Med Chem ; 60(13): 5673-5698, 2017 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-28574706

RESUMEN

Phosphodiesterase 2A (PDE2A) inhibitors have been reported to demonstrate in vivo activity in preclinical models of cognition. To more fully explore the biology of PDE2A inhibition, we sought to identify potent PDE2A inhibitors with improved brain penetration as compared to current literature compounds. Applying estimated human dose calculations while simultaneously leveraging synthetically enabled chemistry and structure-based drug design has resulted in a highly potent, selective, brain penetrant compound 71 (PF-05085727) that effects in vivo biochemical changes commensurate with PDE2A inhibition along with behavioral and electrophysiological reversal of the effects of NMDA antagonists in rodents. This data supports the ability of PDE2A inhibitors to potentiate NMDA signaling and their further development for clinical cognition indications.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/antagonistas & inhibidores , Diseño de Fármacos , Inhibidores de Fosfodiesterasa/química , Inhibidores de Fosfodiesterasa/farmacología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Cristalografía por Rayos X , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/química , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/metabolismo , Perros , Haplorrinos , Humanos , Ratones , Simulación del Acoplamiento Molecular , Inhibidores de Fosfodiesterasa/administración & dosificación , Inhibidores de Fosfodiesterasa/farmacocinética , Ratas
10.
J Mol Model ; 23(1): 7, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27966018

RESUMEN

The phosphodiesterase (PDE) family of proteins are important regulators of signal transduction, which they achieve by controlling the secondary messengers cyclic AMP (cAMP) and cyclic GMP (cGMP). cAMP and cGMP are involved in many critical intracellular processes such as gene transcription, kinase activation, signal transduction in learning and memory, and channel function as secondary messengers. The involvement of PDEs in neuronal communication has made them important therapeutic targets. Considering the recent discovery that PDE2A inhibition can improve cognitive functioning, a combined molecular dynamics simulation and scoring and docking study was carried out to identify selective inhibitors of PDE2A that specifically interact with the recently discovered hydrophobic groove in PDE2A. Using the X-ray crystal structure of PDE2A (from PDB ID: 4HTX), we investigated the binding modes of a range of promising inhibitors based on the known PDE2A inhibitor BAY60-7550 to PDE2A. Graphical abstract The lead molecule showing highest MMPBSA binding energy with 2D and 3D binding pose in hydrophobic groove.


Asunto(s)
AMP Cíclico/química , GMP Cíclico/química , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/antagonistas & inhibidores , Imidazoles/química , Nootrópicos/química , Inhibidores de Fosfodiesterasa/química , Triazinas/química , Secuencias de Aminoácidos , Dominio Catalítico , Cristalografía por Rayos X , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/química , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Imidazoles/síntesis química , Cinética , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Nootrópicos/síntesis química , Inhibidores de Fosfodiesterasa/síntesis química , Fosforilación , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Electricidad Estática , Relación Estructura-Actividad , Termodinámica , Triazinas/síntesis química
11.
Anal Biochem ; 511: 1-9, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27480498

RESUMEN

To establish a strategy to identify dually fatty acylated proteins from cDNA resources, seven N-myristoylated proteins with cysteine (Cys) residues within the 10 N-terminal residues were selected as potential candidates among 27 N-myristoylated proteins identified from a model human cDNA resource. Seven proteins C-terminally tagged with FLAG tag or EGFP were generated and their susceptibility to protein N-myristoylation and S-palmitoylation were evaluated by metabolic labeling with [(3)H]myristic acid or [(3)H]palmitic acid either in an insect cell-free protein synthesis system or in transfected mammalian cells. As a result, EEPD1, one of five proteins (RFTN1, EEPD1, GNAI1, PDE2A, RNF11) found to be dually acylated, was shown to be a novel dually fatty acylated protein. Metabolic labeling experiments using G2A and C7S mutants of EEPD1-EGFP revealed that the palmitoylation site of EEPD1 is Cys at position 7. Analysis of the intracellular localization of EEPD1 C-terminally tagged with FLAG tag or EGFP and its G2A and C7S mutants revealed that the dual acylation directs EEPD1 to localize to the plasma membrane. Thus, dually fatty acylated proteins can be identified from cDNA resources by cell-free and cellular metabolic labeling of N-myristoylated proteins with Cys residue(s) close to the N-myristoylated N-terminus.


Asunto(s)
Proteínas Portadoras/biosíntesis , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/biosíntesis , ADN Complementario/metabolismo , Endodesoxirribonucleasas/biosíntesis , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/biosíntesis , Lipoilación , Ácido Palmítico/metabolismo , Acilación , Animales , Células COS , Proteínas Portadoras/química , Sistema Libre de Células , Chlorocebus aethiops , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/química , ADN Complementario/química , Proteínas de Unión al ADN , Endodesoxirribonucleasas/química , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/química , Humanos
12.
J Am Chem Soc ; 135(32): 11708-11, 2013 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-23899287

RESUMEN

To better understand the structural origins of inhibitor selectivity of human phosphodieasterase families (PDEs 1-11), here we report the X-ray crystal structure of PDE2 in complex with a highly selective, nanomolar inhibitor (BAY60-7550) at 1.9 Å resolution, and the structure of apo PDE2 at 2.0 Å resolution. The crystal structures reveal that the inhibitor binds to the PDE2 active site by using not only the conserved glutamine-switch mechanism for substrate binding, but also a binding-induced, hydrophobic pocket that was not reported previously. In silico affinity profiling by molecular docking indicates that the inhibitor binding to this pocket contributes significantly to the binding affinity and thereby improves the inhibitor selectivity for PDE2. Our results highlight a structure-based design strategy that exploits the potential binding-induced pockets to achieve higher selectivity in the PDE inhibitor development.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/química , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/metabolismo , Imidazoles/farmacología , Inhibidores de Fosfodiesterasa/farmacología , Triazinas/farmacología , Sitios de Unión , Cristalografía por Rayos X , Humanos , Imidazoles/química , Simulación del Acoplamiento Molecular , Inhibidores de Fosfodiesterasa/química , Unión Proteica , Triazinas/química
13.
J Phys Chem B ; 114(48): 16020-8, 2010 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-21077589

RESUMEN

Phosphodiesterase-2 (PDE2) is a key enzyme catalyzing hydrolysis of both cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) that serve as intracellular second messengers. PDE2 has been recognized as an attractive drug target, and selective inhibitors of PDE2 are expected to be promising candidates for the memory enhancer, antidepressant, and anxiolytic agent. In the present study, we examined the detailed binding structures and free energies for PDE2 interacting with a promising series of inhibitors, i.e., benzo[1,4]diazepin-2-one derivatives, by carrying out molecular docking, molecular dynamics (MD) simulations, binding free energy calculations, and binding energy decompositions. The computational results provide valuable insights into the detailed enzyme-inhibitor binding modes including important intermolecular interactions, e.g., the π-π stacking interactions with the common benzo[1,4]diazepin-2-one scaffold of the inhibitors, hydrogen bonding and hydrophobic interactions with the substituents on the benzo[1,4]diazepin-2-one scaffold. Future rational design of new, more potent inhibitors of PDE2 should carefully account for all of these favorable intermolecular interactions. By use of the MD-simulated binding structures, the calculated binding free energies are in good agreement with the experimental activity data for all of the examined benzo[1,4]diazepin-2-one derivatives. The enzyme-inhibitor binding modes determined and the agreement between the calculated and experimental results are expected to be valuable for future rational design of more potent inhibitors of PDE2.


Asunto(s)
Benzodiazepinonas/química , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/química , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/metabolismo , Simulación de Dinámica Molecular , Termodinámica , Benzodiazepinonas/síntesis química , Benzodiazepinonas/metabolismo , Benzodiazepinonas/farmacología , Sitios de Unión , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/antagonistas & inhibidores , Estructura Molecular , Estereoisomerismo , Relación Estructura-Actividad
14.
Br J Pharmacol ; 161(7): 1645-60, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20698857

RESUMEN

BACKGROUND AND PURPOSE: By controlling intracellular cyclic nucleotide levels, phosphodiesterases (PDE) serve important functions within various signalling pathways. The PDE2 and PDE5 families are allosterically activated by their substrate cGMP via regulatory so-called GAF domains. Here, we set out to identify synthetic ligands for the GAF domains of PDE2 and PDE5. EXPERIMENTAL APPROACH: Using fluorophore-tagged, isolated GAF domains of PDE2 and PDE5, promising cGMP analogues were selected. Subsequently, the effects of these analogues on the enzymatic activity of PDE2 and PDE5 were analysed. KEY RESULTS: The PDE2 ligands identified, 5,6-DM-cBIMP and 5,6-DCl-cBIMP, caused pronounced, up to 40-fold increases of the cAMP- and cGMP-hydrolysing activities of PDE2. The ligand for the GAF domains of PDE5, 8-Br-cGMP, elicited a 20-fold GAF-dependent activation and moreover revealed a time-dependent increase in PDE5 activity that occurred independently of a GAF ligand. Although GAF-dependent PDE5 activation was fast at high ligand concentrations, it was slow at physiologically relevant cGMP concentrations; PDE5 reached its final catalytic rates at 1µM cGMP after approximately 10min. CONCLUSIONS AND IMPLICATIONS: We conclude that the delayed activation of PDE5 is required to shape biphasic, spike-like cGMP signals. Phosphorylation of PDE5 further enhances activity and conserves PDE5 activation, thereby enabling PDE5 to act as a molecular memory balancing cGMP responses to nitric oxide or natriuretic peptide signals.


Asunto(s)
GMP Cíclico/análogos & derivados , GMP Cíclico/farmacología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Animales , GMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/química , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/química , Transferencia Resonante de Energía de Fluorescencia , Células HEK293 , Humanos , Ligandos , Ratones , Péptidos Natriuréticos/metabolismo , Óxido Nítrico/metabolismo , Fosforilación , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
15.
Proc Natl Acad Sci U S A ; 106(43): 18225-30, 2009 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-19828435

RESUMEN

We report the X-ray crystal structure of a phosphodiesterase (PDE) that includes both catalytic and regulatory domains. PDE2A (215-900) crystallized as a dimer in which each subunit had an extended organization of regulatory GAF-A and GAF-B and catalytic domains connected by long alpha-helices. The subunits cross at the GAF-B/catalytic domain linker, and each side of the dimer contains in series the GAF-A and GAF-B of one subunit and the catalytic domain of the other subunit. A dimer interface extends over the entire length of the molecule. The substrate binding pocket of each catalytic domain is occluded by the H-loop. We deduced from comparisons with structures of isolated, ligand-bound catalytic subunits that the H-loop swings out to allow substrate access. However, in dimeric PDE2A (215-900), the H-loops of the two catalytic subunits pack against each other at the dimer interface, necessitating movement of the catalytic subunits to allow for H-loop movement. Comparison of the unliganded GAF-B of PDE2A (215-900) with previous structures of isolated, cGMP-bound GAF domains indicates that cGMP binding induces a significant shift in the GAF-B/catalytic domain linker. We propose that cGMP binding to GAF-B causes movement, through this linker region, of the catalytic domains, such that the H-loops no longer pack at the dimer interface and are, instead, free to swing out to allow substrate access. This increase in substrate access is proposed as the basis for PDE2A activation by cGMP and may be a general mechanism for regulation of all PDEs.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/química , Regulación Alostérica , Animales , Biocatálisis , Línea Celular , Cristalografía por Rayos X , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/metabolismo , Activación Enzimática , Humanos , Modelos Moleculares , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Homología Estructural de Proteína
16.
J Phys Chem B ; 113(9): 2896-908, 2009 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-19708117

RESUMEN

Although an X-ray crystal structure of the catalytic domain of human cyclic nucleotide phosphodiesterase-2 (PDE2A) was reported in the literature, the shape of the binding pocket is not suitable for binding with its known potent inhibitors. Extensive molecular modeling, docking, and dynamics simulations in the present study have demonstrated for the first time that the structure of PDE2A catalytic domain may exist in two different forms corresponding to the inhibitor-bound and unbound states of the enzyme. The structural change from the unbound state to the bound state leads to a substantial variation in the size of the pocket but does not affect the general structural feature of the catalytic site. The flexible binding pocket and conserved structural feature of the catalytic site lead us to better understand why this enzyme can catalyze hydrolysis of two different intercellular second messengers. It has been demonstrated that the available X-ray crystal structure of PDE2A was in the unbound state, explaining why it is not suitable for molecular docking studies on the enzyme-inhibitor binding. We have developed a reasonable 3D model of the PDE2A structure in the bound state and determined the detailed binding modes and binding free energies for PDE2A binding with its known potent inhibitors. The calculated binding free energies are in good agreement with available experimental data. The general structural insights, PDE2A model in the bound state, and detailed PDE2A-inhibitor binding structures obtained in this study will be valuable for future rational design of novel, potent inhibitors of PDE2A as therapeutic agents.


Asunto(s)
Biofisica/métodos , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/química , Secuencia de Aminoácidos , Sitios de Unión , Dominio Catalítico , Simulación por Computador , Cristalografía por Rayos X/métodos , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/metabolismo , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Humanos , Hidrólisis , Imagenología Tridimensional , Modelos Químicos , Datos de Secuencia Molecular , Unión Proteica , Homología de Secuencia de Aminoácido
17.
J Biol Chem ; 283(28): 19657-64, 2008 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-18477562

RESUMEN

Cyclic nucleotide phosphodiesterases (PDEs) catalyze the degradation of the cyclic nucleotides cAMP and cGMP, which are important second messengers. Five of the 11 mammalian PDE families have tandem GAF domains at their N termini. PDE10A may be the only mammalian PDE for which cAMP is the GAF domain ligand, and it may be allosterically stimulated by cAMP. PDE10A is highly expressed in striatal medium spiny neurons. Here we report the crystal structure of the C-terminal GAF domain (GAF-B) of human PDE10A complexed with cAMP at 2.1-angstroms resolution. The conformation of the PDE10A GAF-B domain monomer closely resembles those of the GAF domains of PDE2A and the cyanobacterium Anabaena cyaB2 adenylyl cyclase, except for the helical bundle consisting of alpha1, alpha2, and alpha5. The PDE10A GAF-B domain forms a dimer in the crystal and in solution. The dimerization is mainly mediated by hydrophobic interactions between the helical bundles in a parallel arrangement, with a large buried surface area. In the PDE10A GAF-B domain, cAMP tightly binds to a cNMP-binding pocket. The residues in the alpha3 and alpha4 helices, the beta6 strand, the loop between 3(10) and alpha4, and the loop between alpha4 and beta5 are involved in the recognition of the phosphate and ribose moieties. This recognition mode is similar to those of the GAF domains of PDE2A and cyaB2. In contrast, the adenine base is specifically recognized by the PDE10A GAF-B domain in a unique manner, through residues in the beta1 and beta2 strands.


Asunto(s)
Hidrolasas Diéster Fosfóricas/química , Anabaena/enzimología , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Sitios de Unión/fisiología , Cristalografía por Rayos X , AMP Cíclico/química , AMP Cíclico/genética , AMP Cíclico/metabolismo , GMP Cíclico/química , GMP Cíclico/genética , GMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/química , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/metabolismo , Dimerización , Humanos , Neuronas/enzimología , Hidrolasas Diéster Fosfóricas/metabolismo , Estructura Secundaria de Proteína/fisiología , Estructura Terciaria de Proteína/fisiología , Homología Estructural de Proteína , Corteza Visual/enzimología
18.
Mol Interv ; 2(5): 317-23, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-14993386

RESUMEN

GAF domains represent one of the largest families of small-molecule binding units present in nature. The first mammalian GAF domains discovered were the cGMP-binding regulatory domains of several cyclic nucleotide phosphodiesterases (PDEs). The crystal structure of the PDE2A GAF domains has provided our first look at the architecture of the binding site for the second messenger cGMP. The topology of this site differs greatly from all other previously determined cyclic nucleotide binding sites. In PDE2A, cGMP binds to a well-defined pocket in one of the two GAF domains that is analogous to the ligand-binding pocket of the distantly related PAS domains of photoactive yellow protein and FixL. The consensus cGMP-binding motif suggests strongly that only certain GAF domains will bind cGMP. Although the detailed mechanism for how cGMP binding to the GAF domain regulates catalysis remains to be determined, recent data from a GAF domain-containing cAMP-stimulated adenylyl cyclase from Anabaena suggest a mechanism conserved across two billion years of evolution. Because of their unique ligand-binding topologies, the GAF domains of PDEs are likely to offer good new targets for rational drug design.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2 , Evolución Molecular , Nucleótidos Cíclicos , Estructura Terciaria de Proteína , Animales , Sitios de Unión , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/química , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/metabolismo , Modelos Moleculares , Nucleótidos Cíclicos/química , Nucleótidos Cíclicos/metabolismo , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...