Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Mol Pharmacol ; 99(5): 342-357, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33574048

RESUMEN

In recent years, highly sensitive mass spectrometry-based phosphoproteomic analysis is beginning to be applied to identification of protein kinase substrates altered downstream of increased cAMP. Such studies identify a very large number of phosphorylation sites regulated in response to increased cAMP. Therefore, we now are tasked with the challenge of determining how many of these altered phosphorylation sites are relevant to regulation of function in the cell. This minireview describes the use of phosphoproteomic analysis to monitor the effects of cyclic nucleotide phosphodiesterase (PDE) inhibitors on cAMP-dependent phosphorylation events. More specifically, it describes two examples of this approach carried out in the authors' laboratories using the selective PDE inhibitor approach. After a short discussion of several likely conclusions suggested by these analyses of cAMP function in steroid hormone-producing cells and also in T-cells, it expands into a discussion about some newer and more speculative interpretations of the data. These include the idea that multiple phosphorylation sites and not a single rate-limiting step likely regulate these and, by analogy, many other cAMP-dependent pathways. In addition, the idea that meaningful regulation requires a high stoichiometry of phosphorylation to be important is discussed and suggested to be untrue in many instances. These new interpretations have important implications for drug design, especially for targeting pathway agonists. SIGNIFICANCE STATEMENT: Phosphoproteomic analyses identify thousands of altered phosphorylation sites upon drug treatment, providing many possible regulatory targets but also highlighting questions about which phosphosites are functionally important. These data imply that multistep processes are regulated by phosphorylation at not one but rather many sites. Most previous studies assumed a single step or very few rate-limiting steps were changed by phosphorylation. This concept should be changed. Previous interpretations also assumed substoichiometric phosphorylation was not of regulatory importance. This assumption also should be changed.


Asunto(s)
AMP Cíclico/metabolismo , Fosforilación/fisiología , Proteoma/metabolismo , Animales , Humanos , Proteómica/métodos , Transducción de Señal/fisiología
2.
J Proteome Res ; 19(3): 1235-1247, 2020 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-32037842

RESUMEN

Kinase-catalyzed protein phosphorylation is fundamental to eukaryotic signal transduction, regulating most cellular processes. Kinases are frequently dysregulated in cancer, inflammation, and degenerative diseases, and because they can be inhibited with small molecules, they became important drug targets. Accordingly, analytical approaches that determine kinase activation states are critically important to understand kinase-dependent signal transduction and to identify novel drug targets and predictive biomarkers. Multiplexed inhibitor beads (MIBs or kinobeads) efficiently enrich kinases from cell lysates for liquid chromatography-mass spectrometry (LC-MS) analysis. When combined with phosphopeptide enrichment, kinobead/LC-MS can also quantify the phosphorylation state of kinases, which determines their activation state. However, an efficient kinobead/LC-MS kinase phospho-profiling protocol that allows routine analyses of cell lines and tissues has not yet been developed. Here, we present a facile workflow that quantifies the global phosphorylation state of kinases with unprecedented sensitivity. We also found that our kinobead/LC-MS protocol can measure changes in kinase complex composition and show how these changes can indicate kinase activity. We demonstrate the utility of our approach in specifying kinase signaling pathways that control the acute steroidogenic response in Leydig cells; this analysis establishes the first comprehensive framework for the post-translational control of steroid biosynthesis.


Asunto(s)
Transducción de Señal , Espectrometría de Masas en Tándem , Cromatografía Liquida , Humanos , Masculino , Fosforilación , Proteínas Quinasas/metabolismo
3.
Proc Natl Acad Sci U S A ; 114(30): E6240-E6249, 2017 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-28634298

RESUMEN

Specific functions for different cyclic nucleotide phosphodiesterases (PDEs) have not yet been identified in most cell types. Conventional approaches to study PDE function typically rely on measurements of global cAMP, general increases in cAMP-dependent protein kinase (PKA), or the activity of exchange protein activated by cAMP (EPAC). Although newer approaches using subcellularly targeted FRET reporter sensors have helped define more compartmentalized regulation of cAMP, PKA, and EPAC, they have limited ability to link this regulation to downstream effector molecules and biological functions. To address this problem, we have begun to use an unbiased mass spectrometry-based approach coupled with treatment using PDE isozyme-selective inhibitors to characterize the phosphoproteomes of the functional pools of cAMP/PKA/EPAC that are regulated by specific cAMP-PDEs (the PDE-regulated phosphoproteomes). In Jurkat cells we find multiple, distinct PDE-regulated phosphoproteomes that can be defined by their responses to different PDE inhibitors. We also find that little phosphorylation occurs unless at least two different PDEs are concurrently inhibited in these cells. Moreover, bioinformatics analyses of these phosphoproteomes provide insight into the unique functional roles, mechanisms of action, and synergistic relationships among the different PDEs that coordinate cAMP-signaling cascades in these cells. The data strongly suggest that the phosphorylation of many different substrates contributes to cAMP-dependent regulation of these cells. The findings further suggest that the approach of using selective, inhibitor-dependent phosphoproteome analysis can provide a generalized methodology for understanding the roles of different PDEs in the regulation of cyclic nucleotide signaling.


Asunto(s)
3',5'-AMP Cíclico Fosfodiesterasas/antagonistas & inhibidores , Fosfoproteínas/metabolismo , Linfocitos T/metabolismo , Algoritmos , Humanos , Células Jurkat , Redes y Vías Metabólicas
4.
Proc Natl Acad Sci U S A ; 113(38): E5685-93, 2016 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-27601673

RESUMEN

Luteinizing hormone (LH) stimulates steroidogenesis largely through a surge in cyclic AMP (cAMP). Steroidogenic rates are also critically dependent on the availability of cholesterol at mitochondrial sites of synthesis. This cholesterol is provided by cellular uptake of lipoproteins, mobilization of intracellular lipid, and de novo synthesis. Whether and how these pathways are coordinated by cAMP are poorly understood. Recent phosphoproteomic analyses of cAMP-dependent phosphorylation sites in MA10 Leydig cells suggested that cAMP regulates multiple steps in these processes, including activation of the SCAP/SREBP pathway. SCAP [sterol-regulatory element-binding protein (SREBP) cleavage-activating protein] acts as a cholesterol sensor responsible for regulating intracellular cholesterol balance. Its role in cAMP-mediated control of steroidogenesis has not been explored. We used two CRISPR (clustered regularly interspaced short palindromic repeat)-Cas9 (CRISPR associated protein 9) knockout approaches to test the role of SCAP in steroidogenesis. Our results demonstrate that SCAP is required for progesterone production induced by concurrent inhibition of the cAMP phosphodiesterases PDE4 and PDE8. These inhibitors increased SCAP phosphorylation, SREBP2 activation, and subsequent expression of cholesterol biosynthetic genes, whereas SCAP deficiency largely prevented these effects. Reexpression of SCAP in SCAP-deficient cells restored SREBP2 protein expression and partially restored steroidogenic responses, confirming the requirement of SCAP-SREBP2 in steroidogenesis. Inhibitors of 3-hydroxy-3-methylglutaryl-Coenzyme A reductase and isoprenylation attenuated, whereas exogenously provided cholesterol augmented, PDE inhibitor-induced steroidogenesis, suggesting that the cholesterol substrate needed for steroidogenesis is provided by both de novo synthesis and isoprenylation-dependent mechanisms. Overall, these results demonstrate a novel role for LH/cAMP in SCAP/SREBP activation and subsequent regulation of steroidogenesis.


Asunto(s)
AMP Cíclico/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Esteroides/biosíntesis , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo , Animales , Proteínas Portadoras , Colesterol/metabolismo , Regulación de la Expresión Génica , Hidroximetilglutaril-CoA Reductasas/efectos de los fármacos , Hidroximetilglutaril-CoA Reductasas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Células Intersticiales del Testículo/metabolismo , Lipoproteínas/metabolismo , Hormona Luteinizante/metabolismo , Masculino , Proteínas de la Membrana/genética , Mitocondrias/genética , Mitocondrias/metabolismo , Fosforilación , Esteroides/química , Proteína 2 de Unión a Elementos Reguladores de Esteroles/genética
5.
Biol Reprod ; 94(5): 110, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27009040

RESUMEN

The meiotic cell cycle of mammalian oocytes in preovulatory follicles is held in prophase arrest by diffusion of cGMP from the surrounding granulosa cells into the oocyte. Luteinizing hormone (LH) then releases meiotic arrest by lowering cGMP in the granulosa cells. The LH-induced reduction of cGMP is caused in part by a decrease in guanylyl cyclase activity, but the observation that the cGMP phosphodiesterase PDE5 is phosphorylated during LH signaling suggests that an increase in PDE5 activity could also contribute. To investigate this idea, we measured cGMP-hydrolytic activity in rat ovarian follicles. Basal activity was due primarily to PDE1A and PDE5, and LH increased PDE5 activity. The increase in PDE5 activity was accompanied by phosphorylation of PDE5 at serine 92, a protein kinase A/G consensus site. Both the phosphorylation and the increase in activity were promoted by elevating cAMP and opposed by inhibiting protein kinase A, supporting the hypothesis that LH activates PDE5 by stimulating its phosphorylation by protein kinase A. Inhibition of PDE5 activity partially suppressed LH-induced meiotic resumption as indicated by nuclear envelope breakdown, but inhibition of both PDE5 and PDE1 activities was needed to completely inhibit this response. These results show that activities of both PDE5 and PDE1 contribute to the LH-induced resumption of meiosis in rat oocytes, and that phosphorylation and activation of PDE5 is a regulatory mechanism.


Asunto(s)
GMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 1/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Hormona Luteinizante/farmacología , Meiosis/efectos de los fármacos , Folículo Ovárico/efectos de los fármacos , Animales , Células Cultivadas , Femenino , Ratones , Ratones Endogámicos C57BL , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Folículo Ovárico/metabolismo , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
6.
Cell Signal ; 28(7): 764-78, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26643407

RESUMEN

Many cellular processes are modulated by cyclic AMP and nucleotide phosphodiesterases (PDEs) regulate this second messenger by catalyzing its breakdown. The major unique function of testicular Leydig cells is to produce testosterone in response to luteinizing hormone (LH). Treatment of Leydig cells with PDE inhibitors increases cAMP levels and the activity of its downstream effector, cAMP-dependent protein kinase (PKA), leading to a series of kinase-dependent signaling and transcription events that ultimately increase testosterone release. We have recently shown that PDE4B and PDE4C as well as PDE8A and PDE8B are expressed in rodent Leydig cells and that combined inhibition of PDE4 and PDE8 leads to dramatically increased steroid biosynthesis. Here we investigated the effect of PDE4 and PDE8 inhibition on the molecular mechanisms of cAMP actions in a mouse MA10 Leydig cell line model with SILAC mass spectrometry-based phosphoproteomics. We treated MA10 cells either with PDE4 family specific inhibitor (Rolipram) and PDE8 family specific inhibitor (PF-04957325) alone or in combination and quantified the resulting phosphorylation changes at five different time points between 0 and 180min. We identified 28,336 phosphosites from 4837 proteins and observed significant regulation of 749 sites in response to PDE4 and PDE8 inhibitor treatment. Of these, 132 phosphosites were consensus PKA sites. Our data strongly suggest that PDE4 and PDE8 inhibitors synergistically regulate phosphorylation of proteins required for many different cellular processes, including cell cycle progression, lipid and glucose metabolism, transcription, endocytosis and vesicle transport. Our data suggests that cAMP, PDE4 and PDE8 coordinate steroidogenesis by acting on not one rate-limiting step but rather multiple pathways. Moreover, the pools of cAMP controlled by these PDEs also coordinate many other metabolic processes that may be regulated to assure timely and sufficient testosterone secretion in response to LH.


Asunto(s)
3',5'-AMP Cíclico Fosfodiesterasas/metabolismo , AMP Cíclico/metabolismo , Células Intersticiales del Testículo/enzimología , Fosfoproteínas/metabolismo , Proteómica/métodos , Transducción de Señal , 3',5'-AMP Cíclico Fosfodiesterasas/antagonistas & inhibidores , Animales , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Análisis por Conglomerados , Bases de Datos como Asunto , Endocitosis/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Ontología de Genes , Insulina/metabolismo , Marcaje Isotópico , Células Intersticiales del Testículo/efectos de los fármacos , Masculino , Ratones , Inhibidores de Fosfodiesterasa/farmacología , Fosforilación/efectos de los fármacos , Proteoma/metabolismo , Transducción de Señal/efectos de los fármacos , Vesículas Transportadoras/efectos de los fármacos , Vesículas Transportadoras/metabolismo
7.
Proc Natl Acad Sci U S A ; 111(35): 12925-9, 2014 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-25139994

RESUMEN

Conflicting results have been reported for the roles of cGMP and cGMP-dependent protein kinase I (cGKI) in various pathological conditions leading to cardiac hypertrophy and fibrosis. A cardioprotective effect of cGMP/cGKI has been reported in whole animals and isolated cardiomyocytes, but recent evidence from a mouse model expressing cGKIß only in smooth muscle (ßRM) but not in cardiomyocytes, endothelial cells, or fibroblasts has forced a reevaluation of the requirement for cGKI activity in the cardiomyocyte antihypertrophic effects of cGMP. In particular, ßRM mice developed the same hypertrophy as WT controls when subjected to thoracic aortic constriction or isoproterenol infusion. Here, we challenged ßRM and WT (Ctr) littermate control mice with angiotensin II (AII) infusion (7 d; 2 mg ⋅ kg(-1) ⋅ d(-1)) to induce hypertrophy. Both genotypes developed cardiac hypertrophy, which was more pronounced in Ctr animals. Cardiomyocyte size and interstitial fibrosis were increased equally in both genotypes. Addition of sildenafil, a phosphodiesterase 5 (PDE5) inhibitor, in the drinking water had a small effect in reducing myocyte hypertrophy in WT mice and no effect in ßRM mice. However, sildenafil substantially blocked the increase in collagen I, fibronectin 1, TGFß, and CTGF mRNA in Ctr but not in ßRM hearts. These data indicate that, for the initial phase of AII-induced cardiac hypertrophy, lack of cardiomyocyte cGKI activity does not worsen hypertrophic growth. However, expression of cGKI in one or more cell types other than smooth muscle is necessary to allow the antifibrotic effect of sildenafil.


Asunto(s)
Angiotensina II/farmacología , Cardiomegalia/metabolismo , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Hipertensión/metabolismo , Animales , Cardiomegalia/inducido químicamente , GMP Cíclico/metabolismo , Fibrosis/inducido químicamente , Fibrosis/metabolismo , Marcadores Genéticos , Hipertensión/inducido químicamente , Ratones , Músculo Liso/metabolismo , Contracción Miocárdica/efectos de los fármacos , Contracción Miocárdica/fisiología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Óxido Nítrico/metabolismo , Inhibidores de Fosfodiesterasa 5/farmacología , Piperazinas/farmacología , Purinas/farmacología , Citrato de Sildenafil , Sulfonas/farmacología , Vasoconstrictores/farmacología
8.
Trends Pharmacol Sci ; 35(8): 404-13, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24948380

RESUMEN

cGMP inhibits hypertrophy, decreases fibrosis, and protects against cardiac ischemia-reperfusion (I/R) injury. Gene-targeting studies have not defined a clear role for its major downstream effector, cGMP-dependent protein kinase I (cGKI), in cardiac hypertrophy, but do implicate cGMP-cGKI signaling in fibrosis and I/R injury. No direct cGKI activators have advanced to clinical trials, whereas cardiac trials of agents that modulate cGMP via particulate or soluble guanylyl cyclases (GCs) and phosphodiesterase 5 (PDE5) are ongoing. Here we review concerns arising from preclinical and clinical studies that question whether targeting the cGMP pathway remains an encouraging concept for management of heart dysfunction. So far, trial results for GC modulators are inconclusive, and sildenafil, a PDE5 inhibitor, although cardioprotective in mouse models, has not shown positive clinical results. Preclinical cardioprotection observed for sildenafil may result from inhibition of PDE5 in non-cardiomyocytes or off-target effects, possibly on PDE1C. On the basis of such mechanistic considerations, re-evaluation of the cellular localization of drug target(s) and intervention protocols for cGMP-elevating agents may be needed.


Asunto(s)
GMP Cíclico/metabolismo , Cardiopatías/tratamiento farmacológico , Cardiopatías/metabolismo , Animales , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Cardiopatías/enzimología , Humanos , Inhibidores de Fosfodiesterasa 5/farmacología
9.
PLoS One ; 8(8): e71279, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23967182

RESUMEN

A cell-based high-throughput screen (HTS) was developed to detect phosphodiesterase 8 (PDE8) and PDE4/8 combination inhibitors. By replacing the Schizosaccharomyces pombe PDE gene with the murine PDE8A1 gene in strains lacking adenylyl cyclase, we generated strains whose protein kinase A (PKA)-stimulated growth in 5-fluoro orotic acid (5FOA) medium reflects PDE8 activity. From our previously-identified PDE4 and PDE7 inhibitors, we identified a PDE4/8 inhibitor that allowed us to optimize screening conditions. Of 222,711 compounds screened, ∼0.2% displayed composite Z scores of >20. Additional yeast-based assays using the most effective 367 compounds identified 30 candidates for further characterization. Among these, compound BC8-15 displayed the lowest IC50 value for both PDE4 and PDE8 inhibition in in vitro enzyme assays. This compound also displays significant activity against PDE10A and PDE11A. BC8-15 elevates steroidogenesis in mouse Leydig cells as a single pharmacological agent. Assays using BC8-15 and two structural derivatives support a model in which PDE8 is a primary regulator of testosterone production by Leydig cells, with an additional role for PDE4 in this process. BC8-15, BC8-15A, and BC8-15C, which are commercially available compounds, display distinct patterns of activity against PDE4, PDE8, PDE10A, and PDE11A, representing a chemical toolkit that could be used to examine the biological roles of these enzymes in cell culture systems.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Células Intersticiales del Testículo/efectos de los fármacos , Células Intersticiales del Testículo/metabolismo , Inhibidores de Fosfodiesterasa 4/farmacología , Schizosaccharomyces/genética , Esteroides/biosíntesis , Animales , Línea Celular Tumoral , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/química , Ensayos Analíticos de Alto Rendimiento , Humanos , Masculino , Ratones , Simulación del Acoplamiento Molecular , Inhibidores de Fosfodiesterasa 4/química , Inhibidores de Fosfodiesterasa 4/metabolismo , Conformación Proteica , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología
10.
Methods Mol Biol ; 1020: 51-62, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23709025

RESUMEN

Cyclic nucleotides (cAMP and cGMP) as second messengers regulate a wide variety of biological processes such as cellular growth, secretary signaling, and neuroplasticity. These processes can be regulated by increasing the synthesis of cyclic nucleotides (cyclases), by regulation of cAMP and cGMP effector proteins such as cAMP- and cGMP-dependent protein kinases, or by regulation of cyclic nucleotide degradation via cyclic nucleotide phosphodiestases (PDEs). At present PDEs are classified into 11 gene families, each containing several different isoforms and splice variants. All PDEs share considerable homology in their catalytic domains but substantially differ in their N-terminal regions, that contain different types of regulatory. The different PDEs show complex substrate specificity. PDE5, PDE6, and PDE9 are considered to be cGMP specific, while PDE1, PDE2, PDE3, PDE10, and PDE11 can hydrolyze both cGMP and cAMP. PDE4, PDE7, and PDE8 use mainly cAMP as their substrates at physiological substrate levels. Here we describe two methods designed for measuring cGMP (cAMP) hydrolytic activities. The first one is a traditional method using radioactive substrates and the second one is a recently developed nonradioactive method based on Isothermal Titration Calorimetry.


Asunto(s)
GMP Cíclico/metabolismo , Pruebas de Enzimas/métodos , Hidrolasas Diéster Fosfóricas/metabolismo , Calorimetría/métodos , AMP Cíclico/metabolismo , Hidrólisis , Cinética , Especificidad por Sustrato , Temperatura
11.
Proc Natl Acad Sci U S A ; 110(16): E1533-42, 2013 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-23509299

RESUMEN

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.


Asunto(s)
3',5'-AMP Cíclico Fosfodiesterasas/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas Proto-Oncogénicas c-raf/metabolismo , 3',5'-AMP Cíclico Fosfodiesterasas/genética , Animales , Western Blotting , Cartilla de ADN/genética , Drosophila melanogaster , Eliminación de Gen , Células HEK293 , Células HeLa , Humanos , Inmunoprecipitación , Sistema de Señalización de MAP Quinasas/genética , Espectrometría de Masas , Ratones , Ratones Noqueados , Mutagénesis Sitio-Dirigida , Fosforilación , Resonancia por Plasmón de Superficie
12.
Mol Pharmacol ; 83(6): 1155-65, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23493317

RESUMEN

Brown adipose tissue (BAT) is a highly thermogenic organ that converts lipids and glucose into heat. Many of the metabolic and gene transcriptional hallmarks of BAT activation, namely increased lipolysis, uncoupling protein-1 (UCP1) mRNA, and glucose uptake, are regulated by the adrenergic second messenger, cAMP. Cyclic nucleotide phosphodiesterases (PDEs) catalyze the breakdown of cAMP, thereby regulating the magnitude and duration of this signaling molecule. In the absence of adrenergic stimulus, we found that it required a combination of a PDE3 and a PDE4 inhibitor to fully induce UCP1 mRNA and lipolysis in brown adipocytes, whereas neither PDE inhibitor alone had any substantial effect under basal conditions. Under submaximal ß-adrenoceptor stimulation of brown adipocytes, a PDE3 inhibitor alone could potentiate induction of UCP1 mRNA, whereas a PDE4 inhibitor alone could augment lipolysis, indicating differential roles for each of these two PDEs. Neither induction of UCP1 nor lipolysis was altered by inhibition of PDE1, PDE2, or PDE8A. Finally, when injected into mice, the combination of PDE3 and PDE4 inhibitors stimulated glucose uptake in BAT under thermoneutral and fasted conditions, a response that was further potentiated by the global ablation of PDE8A. Taken together, these data reveal that multiple PDEs work in concert to regulate three of the important pathways leading to BAT activation, a finding that may provide an improved conceptual basis for the development of therapies for obesity-related diseases.


Asunto(s)
Tejido Adiposo Pardo/efectos de los fármacos , Inhibidores de Fosfodiesterasa 3/farmacología , Inhibidores de Fosfodiesterasa 4/farmacología , 3',5'-AMP Cíclico Fosfodiesterasas/antagonistas & inhibidores , 3',5'-AMP Cíclico Fosfodiesterasas/genética , 3',5'-AMP Cíclico Fosfodiesterasas/metabolismo , Tejido Adiposo Pardo/citología , Tejido Adiposo Pardo/metabolismo , Agonistas Adrenérgicos beta/farmacología , Animales , Células Cultivadas , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Sinergismo Farmacológico , Glucosa/metabolismo , Canales Iónicos/genética , Canales Iónicos/metabolismo , Isoenzimas/metabolismo , Lipólisis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , ARN Mensajero/metabolismo , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Proteína Desacopladora 1
13.
J Pathol ; 228(1): 77-87, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22653783

RESUMEN

Duchenne muscular dystrophy (DMD) is the most common form of muscular dystrophy caused by mutations in the dystrophin gene. Loss of dystrophin initiates a progressive decline in skeletal muscle integrity and contractile capacity which weakens respiratory muscles including the diaphragm, culminating in respiratory failure, the leading cause of morbidity and mortality in DMD patients. At present, corticosteroid treatment is the primary pharmacological intervention in DMD, but has limited efficacy and adverse side effects. Thus, there is an urgent need for new safe, cost-effective, and rapidly implementable treatments that slow disease progression. One promising new approach is the amplification of nitric oxide-cyclic guanosine monophosphate (NO-cGMP) signalling pathways with phosphodiesterase 5 (PDE5) inhibitors. PDE5 inhibitors serve to amplify NO signalling that is attenuated in many neuromuscular diseases including DMD. We report here that a 14-week treatment of the mdx mouse model of DMD with the PDE5 inhibitor sildenafil (Viagra(®), Revatio(®)) significantly reduced mdx diaphragm muscle weakness without impacting fatigue resistance. In addition to enhancing respiratory muscle contractility, sildenafil also promoted normal extracellular matrix organization. PDE5 inhibition slowed the establishment of mdx diaphragm fibrosis and reduced matrix metalloproteinase-13 (MMP-13) expression. Sildenafil also normalized the expression of the pro-fibrotic (and pro-inflammatory) cytokine tumour necrosis factor α (TNFα). Sildenafil-treated mdx diaphragms accumulated significantly less Evans Blue tracer dye than untreated controls, which is also indicative of improved diaphragm muscle health. We conclude that sildenafil-mediated PDE5 inhibition significantly reduces diaphragm respiratory muscle dysfunction and pathology in the mdx mouse model of Duchenne muscular dystrophy. This study provides new insights into the therapeutic utility of targeting defects in NO-cGMP signalling with PDE5 inhibitors in dystrophin-deficient muscle.


Asunto(s)
Diafragma/efectos de los fármacos , Fibrosis/tratamiento farmacológico , Debilidad Muscular/tratamiento farmacológico , Distrofia Muscular de Duchenne/tratamiento farmacológico , Inhibidores de Fosfodiesterasa 5/farmacología , Piperazinas/farmacología , Sulfonas/farmacología , Animales , Creatina Quinasa/sangre , GMP Cíclico/metabolismo , Diafragma/metabolismo , Diafragma/patología , Modelos Animales de Enfermedad , Azul de Evans/metabolismo , Fibrosis/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Contracción Muscular/efectos de los fármacos , Fatiga Muscular/efectos de los fármacos , Fatiga Muscular/fisiología , Debilidad Muscular/etiología , Distrofia Muscular de Duchenne/complicaciones , Distrofia Muscular de Duchenne/patología , Óxido Nítrico/metabolismo , Purinas/farmacología , Citrato de Sildenafil
14.
Mol Pharmacol ; 81(4): 556-66, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22232524

RESUMEN

Phosphodiesterase (PDE) 8A and PDE8B are high-affinity, cAMP-specific phosphodiesterases that are highly expressed in Leydig cells. PDE8A is largely associated with mitochondria, whereas PDE8B is broadly distributed in the cytosol. We used a new, PDE8-selective inhibitor, PF-04957325, and genetically ablated PDE8A(-/-), PDE8B(-/-) and PDE8A(-/-)/B(-/-) mice to determine roles for these PDEs in the regulation of testosterone production. PF-04957325 treatment of WT Leydig cells or MA10 cells increased steroid production but had no effect in PDE8A (-/-)/B(-/-) double-knockout cells, confirming the selectivity of the drug. Moreover, under basal conditions, cotreatment with PF-04957325 plus rolipram, a PDE4-selective inhibitor, synergistically potentiated steroid production. These results suggest that the pool(s) of cAMP regulating androgen production are controlled by PDE8s working in conjunction with PDE4. Likewise, PDE8A (-/-)/B(-/-) cells had higher testosterone production than cells from either PDE8A(-/-) or PDE8B(-/-) mice, suggesting that both PDE8s work in concert to regulate steroid production. We further demonstrate that combined inhibition of PDE8s and PDE4 greatly increased PKA activity including phosphorylation of cholesterol-ester hydrolase (CEH)/hormone-sensitive lipase (HSL). CEH/HSL phosphorylation also was increased in PDE8A(-/-)/B(-/-) cells compared with WT cells. Finally, combined inhibition of PDE8s and PDE4 increased the expression of steroidogenic acute regulatory (StAR) protein. Together these findings suggest that both PDE8A and PDE8B play essential roles to maintain low cAMP levels, thereby suppressing resting steroidogenesis by keeping CEH/HSL inactive and StAR protein expression low. They also suggest that in order for PDE inhibitor therapy to be an effective stimulator of steroidogenesis, both PDE8 isozymes and PDE4 need to be simultaneously targeted.


Asunto(s)
3',5'-AMP Cíclico Fosfodiesterasas/metabolismo , Isoenzimas/metabolismo , Células Intersticiales del Testículo/metabolismo , Esteroides/biosíntesis , 3',5'-AMP Cíclico Fosfodiesterasas/genética , Animales , Inmunoprecipitación , Isoenzimas/genética , Células Intersticiales del Testículo/enzimología , Masculino , Ratones , Ratones Noqueados
15.
Curr Opin Pharmacol ; 11(6): 670-5, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21962440

RESUMEN

The second messenger, cAMP, is one of the most important regulatory signals for control of steroidogenesis. This review focuses on current knowledge about regulation of cyclic nucleotides by phosphodiesterases (PDEs) in steroidogenic tissues. The first PDE known to directly regulate steroidogenesis was PDE2, the cGMP-stimulated PDE. PDE2 mediates ANP/cGMP-induced decreases in aldosterone production. Recently, the PDE8 family has been shown to control steroidogenesis in two tissues. Specifically, PDE8A regulates testosterone production by itself and in concert with additional IBMX-sensitive PDEs. PDE8B modulates basal corticosterone synthesis via acute and chronic mechanisms. In addition to cAMP-dependent pathways, cGMP signaling also can promote steroidogenesis, and PDE5 modulates this process. Finally, PDE mutations may lead to several human diseases characterized by abnormal steroid levels.


Asunto(s)
3',5'-AMP Cíclico Fosfodiesterasas/metabolismo , 3',5'-GMP Cíclico Fosfodiesterasas/metabolismo , Aldosterona/metabolismo , Corticosterona/metabolismo , Testosterona/metabolismo , 3',5'-AMP Cíclico Fosfodiesterasas/genética , 3',5'-GMP Cíclico Fosfodiesterasas/genética , Corteza Suprarrenal/enzimología , Corteza Suprarrenal/metabolismo , Hiperplasia Suprarrenal Congénita/enzimología , Hiperplasia Suprarrenal Congénita/genética , Animales , Factor Natriurético Atrial/metabolismo , Síndrome de Cushing/enzimología , Síndrome de Cushing/genética , AMP Cíclico/fisiología , GMP Cíclico/fisiología , Femenino , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Células Intersticiales del Testículo/enzimología , Células Intersticiales del Testículo/metabolismo , Masculino , Mutación , Sistemas de Mensajero Secundario
16.
J Ethnopharmacol ; 137(3): 1437-41, 2011 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-21884777

RESUMEN

AIM OF THE STUDY: A number of medicinal plants are used in traditional medicine to treat erectile dysfunction. Since cyclic nucleotide PDEs inhibitors underlie several current treatments for this condition, we sought to show whether these plants might contain substantial amounts of PDE5 inhibitors. MATERIALS AND METHODS: Forty one plant extracts and eight 7-methoxyflavones from Kaempferia parviflora Wall. ex Baker were screened for PDE5 and PDE6 inhibitory activities using the two-step radioactive assay. The PDE5 and PDE6 were prepared from mice lung and chicken retinas, respectively. All plant extracts were tested at 50 µg/ml whereas the pure compounds were tested at 10 µM. RESULTS: From forty one plant extracts tested, four showed the PDE5 inhibitory effect. The chemical constituents isolated from rhizomes of Kaempferia parviflora were further investigated on inhibitory activity against PDE5 and PDE6. The results showed that 7-methoxyflavones from this plant showed inhibition toward both enzymes. The most potent PDE5 inhibitor was 5,7-dimethoxyflavone (IC(50) = 10.64 ± 2.09 µM, selectivity on PDE5 over PDE6 = 3.71). Structure activity relationship showed that the methoxyl group at C-5 position of 7-methoxyflavones was necessary for PDE5 inhibition. CONCLUSIONS: Kaempferia parviflora rhizome extract and its 7-methoxyflavone constituents had moderate inhibitory activity against PDE5. This finding provides an explanation for enhancing sexual performance in the traditional use of Kaempferia parviflora. Moreover, 5,7-dimethoxyflavones should make a useful lead compound to further develop clinically efficacious PDE5 inhibitors.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Erección Peniana/efectos de los fármacos , Inhibidores de Fosfodiesterasa 5/farmacología , Extractos Vegetales/farmacología , Zingiberaceae , Animales , Pollos , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/metabolismo , Humanos , Pulmón/enzimología , Masculino , Ratones , Estructura Molecular , Inhibidores de Fosfodiesterasa 5/química , Inhibidores de Fosfodiesterasa 5/aislamiento & purificación , Extractos Vegetales/química , Extractos Vegetales/aislamiento & purificación , Plantas Medicinales , Retina/enzimología , Rizoma , Relación Estructura-Actividad , Zingiberaceae/química
17.
Handb Exp Pharmacol ; (204): 193-210, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21695641

RESUMEN

The endothelium plays an important role in maintaining normal vascular function. Endothelial barrier dysfunction leading to increased permeability and vascular leakage is associated with several pathological conditions such as edema and sepsis. Thus, the development of drugs that improve endothelial barrier function is an active area of research. In this chapter, the current knowledge concerning the signaling pathways regulating endothelial barrier function is discussed with a focus on cyclic nucleotide second messengers (cAMP and cGMP) and cyclic nucleotide phosphodiesterases (PDEs). Both cAMP and cGMP have been shown to have differential effects on endothelial permeability in part due to the various effector molecules, crosstalk, and compartmentalization of cyclic nucleotide signaling. PDEs, by controlling the amplitude, duration, and localization of cyclic nucleotides, have been shown to play a critical role in regulating endothelial barrier function. Thus, PDEs are attractive drug targets for the treatment of disease states involving endothelial barrier dysfunction.


Asunto(s)
AMP Cíclico/fisiología , GMP Cíclico/fisiología , Endotelio Vascular/metabolismo , Hidrolasas Diéster Fosfóricas/fisiología , Animales , Humanos , Permeabilidad , Transducción de Señal
18.
Handb Exp Pharmacol ; (204): 323-44, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21695647

RESUMEN

Duchenne muscular dystrophy (DMD) is a devastating and ultimately fatal disease characterized by progressive muscle wasting and weakness. DMD is caused by the absence of a functional dystrophin protein, which in turn leads to reduced expression and mislocalization of dystrophin-associated proteins including neuronal nitric oxide (NO) synthase mu (nNOSµ). Disruption of nNOSµ signaling results in muscle fatigue and unopposed sympathetic vasoconstriction during exercise, thereby increasing contraction-induced damage in dystrophin-deficient muscles. The loss of normal nNOSµ signaling during exercise is central to the vascular dysfunction proposed over 40 years ago to be an important pathogenic mechanism in DMD. Recent preclinical studies focused on circumventing defective nNOSµ signaling in dystrophic skeletal and cardiac muscle by inhibiting phosphodiesterase 5A (PDE5A) have shown promising results. This review addresses nNOS signaling in normal and dystrophin-deficient muscles and the potential of PDE5A inhibition as a therapeutic approach for the treatment of cardiovascular deficits in DMD.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/fisiología , Distrofia Muscular de Duchenne/tratamiento farmacológico , Inhibidores de Fosfodiesterasa 5/uso terapéutico , Animales , GMP Cíclico/fisiología , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/enzimología , Distrofia Muscular de Duchenne/etiología , Miocitos Cardíacos/enzimología , Óxido Nítrico/fisiología , Óxido Nítrico Sintasa de Tipo I/fisiología , Transducción de Señal
19.
Handb Exp Pharmacol ; (204): 365-90, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21695649

RESUMEN

Monocytes are immune cells that can differentiate into a number of cell types including macrophages, dendritic cells, and osteoclasts upon exposure to various cytokines. The phenotypes of these differentiated cells are highly heterogeneous and their differentiation can be affected by the cyclic nucleotides, 3'-5'-cyclic adenosine monophosphate (cAMP) and 3'-5'-cyclic guanosine monophosphate (cGMP). The intracellular levels of cAMP and cGMP are controlled through regulation of production by adenylyl and guanylyl cyclases and through degradation by cyclic nucleotide phosphodiesterases (PDEs). PDE inhibition and subsequent changes in cyclic nucleotide levels can alter the final phenotype of a differentiating monocyte with regards to surface marker expression, gene expression, or changes in secreted chemokine and cytokine levels. The differentiation process itself can also be either inhibited or augmented by changes in cyclic nucleotide levels, depending on the system being studied and the timing of cyclic nucleotide elevation. This chapter explores the effects of PDE inhibition and increases in cGMP and cAMP on monocytic differentiation into osteoclasts, dendritic cells, and macrophages.


Asunto(s)
Monocitos/citología , Nucleótidos Cíclicos/fisiología , Hidrolasas Diéster Fosfóricas/fisiología , Animales , Diferenciación Celular/efectos de los fármacos , Células Dendríticas/citología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Humanos , Factor Estimulante de Colonias de Macrófagos/farmacología , Osteoclastos/citología , Inhibidores de Fosfodiesterasa/farmacología
20.
Mol Pharmacol ; 79(4): 639-48, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21187369

RESUMEN

The functions of the phosphodiesterase 8B (PDE8) family of phosphodiesterases have been largely unexplored because of the unavailability of selective pharmacological inhibitors. Here, we report a novel function of PDE8B as a major regulator of adrenal steroidogenesis using a genetically ablated PDE8B mouse model as well as cell lines treated with either a new PDE8-selective inhibitor or a short hairpin RNA (shRNA) construct against PDE8B. We demonstrate that PDE8B is highly enriched in mouse adrenal fasciculata cells, and show that PDE8B knockout mice have elevated urinary corticosterone as a result of adrenal hypersensitivity toward adrenocorticotropin. Likewise, ablation of PDE8B mRNA transcripts by an shRNA construct potentiates steroidogenesis in the commonly used Y-1 adrenal cell line. We also observed that the PDE8-selective inhibitor (PF-04957325) potentiates adrenocorticotropin stimulation of steroidogenesis by increasing cAMP-dependent protein kinase activity in both primary isolated adrenocortical cells and Y-1 cells. It is noteworthy that PDE8s have their greatest control under low adrenocorticotropin-stimulated conditions, whereas other higher K(m) PDE(s) modulate steroidogenesis more effectively when cells are fully stimulated. Finally, both genetic ablation of PDE8B and long-term pharmacological inhibition of PDE8s cause increased expression of steroidogenic enzymes. We conclude that PDE8B is a major regulator of one or more pools of cAMP that promote steroidogenesis via both short- and long-term mechanisms. These findings further suggest PDE8B as a potential therapeutic target for the treatment of several different adrenal diseases.


Asunto(s)
3',5'-AMP Cíclico Fosfodiesterasas/fisiología , Corteza Suprarrenal/enzimología , Esteroides/biosíntesis , 3',5'-AMP Cíclico Fosfodiesterasas/deficiencia , 3',5'-AMP Cíclico Fosfodiesterasas/metabolismo , Corteza Suprarrenal/metabolismo , Animales , Células Cultivadas , AMP Cíclico/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA