Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Circulation ; 146(23): 1758-1778, 2022 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-36259389

RESUMEN

BACKGROUND: Phosphodiesterase 3A (PDE3A) gain-of-function mutations cause hypertension with brachydactyly (HTNB) and lead to stroke. Increased peripheral vascular resistance, rather than salt retention, is responsible. It is surprising that the few patients with HTNB examined so far did not develop cardiac hypertrophy or heart failure. We hypothesized that, in the heart, PDE3A mutations could be protective. METHODS: We studied new patients. CRISPR-Cas9-engineered rat HTNB models were phenotyped by telemetric blood pressure measurements, echocardiography, microcomputed tomography, RNA-sequencing, and single nuclei RNA-sequencing. Human induced pluripotent stem cells carrying PDE3A mutations were established, differentiated to cardiomyocytes, and analyzed by Ca2+ imaging. We used Förster resonance energy transfer and biochemical assays. RESULTS: We identified a new PDE3A mutation in a family with HTNB. It maps to exon 13 encoding the enzyme's catalytic domain. All hitherto identified HTNB PDE3A mutations cluster in exon 4 encoding a region N-terminally from the catalytic domain of the enzyme. The mutations were recapitulated in rat models. Both exon 4 and 13 mutations led to aberrant phosphorylation, hyperactivity, and increased PDE3A enzyme self-assembly. The left ventricles of our patients with HTNB and the rat models were normal despite preexisting hypertension. A catecholamine challenge elicited cardiac hypertrophy in HTNB rats only to the level of wild-type rats and improved the contractility of the mutant hearts, compared with wild-type rats. The ß-adrenergic system, phosphodiesterase activity, and cAMP levels in the mutant hearts resembled wild-type hearts, whereas phospholamban phosphorylation was decreased in the mutants. In our induced pluripotent stem cell cardiomyocyte models, the PDE3A mutations caused adaptive changes of Ca2+ cycling. RNA-sequencing and single nuclei RNA-sequencing identified differences in mRNA expression between wild-type and mutants, affecting, among others, metabolism and protein folding. CONCLUSIONS: Although in vascular smooth muscle, PDE3A mutations cause hypertension, they confer protection against hypertension-induced cardiac damage in hearts. Nonselective PDE3A inhibition is a final, short-term option in heart failure treatment to increase cardiac cAMP and improve contractility. Our data argue that mimicking the effect of PDE3A mutations in the heart rather than nonselective PDE3 inhibition is cardioprotective in the long term. Our findings could facilitate the search for new treatments to prevent hypertension-induced cardiac damage.


Asunto(s)
Insuficiencia Cardíaca , Hipertensión , Células Madre Pluripotentes Inducidas , Humanos , Ratas , Animales , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/metabolismo , Microtomografía por Rayos X , Células Madre Pluripotentes Inducidas/metabolismo , Hipertensión/complicaciones , Hipertensión/genética , Miocitos Cardíacos/metabolismo , Cardiomegalia , ARN
2.
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
3.
Proc Natl Acad Sci U S A ; 110(49): 19778-83, 2013 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-24248367

RESUMEN

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.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/metabolismo , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Inhibidores de Fosfodiesterasa 3/farmacología , Proteínas 14-3-3/genética , Sitios de Unión/genética , Cromatografía en Gel , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Electroforesis en Gel Bidimensional , Activación Enzimática/fisiología , Células HEK293 , Humanos , Inmunoprecipitación , Isoenzimas/metabolismo , Isoproterenol/farmacología , Inhibidores de Fosfodiesterasa 3/metabolismo , Fosforilación , Proteína Quinasa C/metabolismo
4.
Circ Res ; 109(9): 1024-1030, 2011 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-21903937

RESUMEN

RATIONALE: Baseline contractility of mouse hearts is modulated in a phosphatidylinositol 3-kinase-γ-dependent manner by type 4 phosphodiesterases (PDE4), which regulate cAMP levels within microdomains containing the sarcoplasmic reticulum (SR) calcium ATPase type 2a (SERCA2a). OBJECTIVE: The goal of this study was to determine whether PDE4D regulates basal cardiac contractility. METHODS AND RESULTS: At 10 to 12 weeks of age, baseline cardiac contractility in PDE4D-deficient (PDE4D(-/-)) mice was elevated mice in vivo and in Langendorff perfused hearts, whereas isolated PDE4D(-/-) cardiomyocytes showed increased whole-cell Ca2+ transient amplitudes and SR Ca2+content but unchanged L-type calcium current, compared with littermate controls (WT). The protein kinase A inhibitor R(p)-adenosine-3',5' cyclic monophosphorothioate (R(p)-cAMP) lowered whole-cell Ca2+ transient amplitudes and SR Ca2+ content in PDE4D(-/-) cardiomyocytes to WT levels. The PDE4 inhibitor rolipram had no effect on cardiac contractility, whole-cell Ca2+ transients, or SR Ca2+ content in PDE4D(-/-) preparations but increased these parameters in WT myocardium to levels indistinguishable from those in PDE4D(-/-). The functional changes in PDE4D(-/-) myocardium were associated with increased PLN phosphorylation but not cardiac ryanodine receptor phosphorylation. Rolipram increased PLN phosphorylation in WT cardiomyocytes to levels indistinguishable from those in PDE4D(-/-) cardiomyocytes. In murine and failing human hearts, PDE4D coimmunoprecipitated with SERCA2a but not with cardiac ryanodine receptor. CONCLUSIONS: PDE4D regulates basal cAMP levels in SR microdomains containing SERCA2a-PLN, but not L-type Ca2+ channels or ryanodine receptor. Because whole-cell Ca2+ transient amplitudes are reduced in failing human myocardium, these observations may have therapeutic implications for patients with heart failure.


Asunto(s)
Canales de Calcio Tipo L/fisiología , Calcio/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Contracción Miocárdica/fisiología , Miocitos Cardíacos/metabolismo , Retículo Sarcoplasmático/metabolismo , Animales , Proteínas de Unión al Calcio/metabolismo , Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/patología , AMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/genética , Femenino , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Humanos , Masculino , Ratones , Ratones Noqueados , Modelos Animales , Miocitos Cardíacos/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo
5.
Basic Res Cardiol ; 106(2): 249-62, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21161247

RESUMEN

PDE4 isoenzymes are critical in the control of cAMP signaling in rodent cardiac myocytes. Ablation of PDE4 affects multiple key players in excitation-contraction coupling and predisposes mice to the development of heart failure. As little is known about PDE4 in human heart, we explored to what extent cardiac expression and functions of PDE4 are conserved between rodents and humans. We find considerable similarities including comparable amounts of PDE4 activity expressed, expression of the same PDE4 subtypes and splicing variants, anchoring of PDE4 to the same subcellular compartments and macromolecular signaling complexes, and downregulation of PDE4 activity and protein in heart failure. The major difference between the species is a fivefold higher amount of non-PDE4 activity in human hearts compared to rodents. As a consequence, the effect of PDE4 inactivation is different in rodents and humans. PDE4 inhibition leads to increased phosphorylation of virtually all PKA substrates in mouse cardiomyocytes, but increased phosphorylation of only a restricted number of proteins in human cardiomyocytes. Our findings suggest that PDE4s have a similar role in the local regulation of cAMP signaling in rodent and human heart. However, inhibition of PDE4 has 'global' effects on cAMP signaling only in rodent hearts, as PDE4 comprises a large fraction of the total cardiac PDE activity in rodents but not in humans. These differences may explain the distinct pharmacological effects of PDE4 inhibition in rodent and human hearts.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Insuficiencia Cardíaca/enzimología , Miocardio/enzimología , Animales , Células Cultivadas , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Activación Enzimática , Humanos , Isoenzimas/metabolismo , Ratones , Miocitos Cardíacos/enzimología , Fosforilación , Ratas
6.
Handb Exp Pharmacol ; (204): 237-49, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21695643

RESUMEN

Compounds that inhibit the catalytic activity of cyclic nucleotide phosphodiesterases are used as therapeutic agents to increase intracellular cAMP and/or cGMP content in cells or tissues of interest. In patients with heart failure, inhibitors of enzymes in the PDE3 family of cyclic nucleotide phosphodiesterases are used to raise intracellular cAMP content in cardiac muscle, with inotropic actions. These drugs are effective in acute applications, but their long-term use has been complicated by an increase in cardiovascular mortality in clinical trials. Inhibitors of enzymes in the PDE5 family have been used to raise cGMP content in cardiac muscle in animal models of pressure overload, chronic ß-adrenergic receptor stimulation, ischemic injury, and doxorubicin toxicity, and have been shown to have antihypertrophic and cardioprotective actions. Recent experimental results raise some question as to the likely applicability of these findings to humans, in whose hearts PDE5 is present at much lower levels than those seen in animal models, and raise the possibility of PDE1, a dual-specificity phosphodiesterase present at high levels in human myocardium, as an alternative target for inotropic and cardioprotective actions.


Asunto(s)
Insuficiencia Cardíaca/tratamiento farmacológico , Inhibidores de Fosfodiesterasa/uso terapéutico , Animales , AMP Cíclico/análisis , GMP Cíclico/análisis , Insuficiencia Cardíaca/enzimología , Humanos , Inhibidores de Fosfodiesterasa 3/uso terapéutico , Inhibidores de Fosfodiesterasa 4/uso terapéutico , Inhibidores de Fosfodiesterasa 5/uso terapéutico
7.
J Pharmacol Exp Ther ; 330(3): 884-91, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19546307

RESUMEN

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.


Asunto(s)
AMP Cíclico/metabolismo , Insuficiencia Cardíaca/metabolismo , Miocardio/metabolismo , Inhibidores de Fosfodiesterasa/farmacología , Piperazinas/farmacología , Sulfonas/farmacología , Animales , Vasos Coronarios/fisiología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Citosol/efectos de los fármacos , Citosol/metabolismo , Humanos , Hidrólisis , Masculino , Ratones , Ratones Endogámicos ICR , Microsomas/efectos de los fármacos , Microsomas/metabolismo , Inhibidores de Fosfodiesterasa 3 , Inhibidores de Fosfodiesterasa 5 , Fosfodiesterasa I/antagonistas & inhibidores , Fosfodiesterasa I/metabolismo , Purinas/farmacología , Citrato de Sildenafil , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/metabolismo
8.
J Card Fail ; 15(1): 31-4, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19181291

RESUMEN

BACKGROUND: Phosphodiesterase-5 (PDE5) inhibitors, which induce relaxation of smooth muscle with some selectivity for the pulmonary vasculature, are used in the treatment of pulmonary hypertension. In some patients, the use of PDE5 inhibitors does not result in the desired magnitude of pulmonary vasodilation. The use of additional vasodilators to further reduce pulmonary vascular resistance is often accompanied by unacceptable reductions in systemic arterial pressure. METHODS AND RESULTS: In 3 patients with heart failure, pulmonary hypertension and low systemic arterial pressures treated with sildenafil, systemic nitrates were added to reduce pulmonary hypertension further. Hemodynamic measurements were made before and after addition of nitrates. Addition of systemic nitrates to sildenafil led to a reduction in mean pulmonary arterial pressure of 11 mm Hg, from 37 mm Hg to 26 mm Hg (P = .06), whereas mean systemic arterial pressure decreased by only 4 mm Hg, from 77 mm Hg to 73 mm Hg (P = .53). The ratio of pulmonary vascular resistance to systemic vascular resistance was reduced by 45% (P = .1). Treatment with sildenafil and nitrates was continued for two to eight months, with no episodes of marked systemic hypotension, syncope, or lightheadedness. CONCLUSIONS: These results suggest that addition of systemic nitrates to sildenafil results in a potentiation of vasodilation that is relatively selective for the pulmonary vasculature, and that this combination may be safe and effective in the treatment of pulmonary hypertension in patients with low systemic arterial pressures.


Asunto(s)
Insuficiencia Cardíaca/complicaciones , Hipertensión Pulmonar/tratamiento farmacológico , Nitrocompuestos/uso terapéutico , Inhibidores de Fosfodiesterasa 5 , Inhibidores de Fosfodiesterasa/uso terapéutico , Vasodilatadores/uso terapéutico , GMP Cíclico , Quimioterapia Combinada , Humanos , Hipertensión Pulmonar/complicaciones , Dinitrato de Isosorbide/uso terapéutico , Masculino , Persona de Mediana Edad , Nitroglicerina/uso terapéutico , Piperazinas/uso terapéutico , Arteria Pulmonar/efectos de los fármacos , Purinas/uso terapéutico , Citrato de Sildenafil , Sulfonas/uso terapéutico , Vasodilatación/efectos de los fármacos
9.
Circ Res ; 100(11): 1569-78, 2007 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-17556670

RESUMEN

Cyclic nucleotide phosphodiesterases regulate cAMP-mediated signaling by controlling intracellular cAMP content. The cAMP-hydrolyzing activity of several families of cyclic nucleotide phosphodiesterases found in human heart is regulated by cGMP. In the case of PDE2, this regulation primarily involves the allosteric stimulation of cAMP hydrolysis by cGMP. For PDE3, cGMP acts as a competitive inhibitor of cAMP hydrolysis. Several cGMP-mediated responses in cardiac cells, including a potentiation of Ca(2+) currents and a diminution of the responsiveness to beta-adrenergic receptor agonists, have been shown to result from the effects of cGMP on cAMP hydrolysis. These effects appear to be dependent on the specific spatial distribution of the cGMP-generating and cAMP-hydrolyzing proteins, as well as on the intracellular concentrations of the two cyclic nucleotides. Gaining a more precise understanding of how these cross-talk mechanisms are individually regulated and coordinated is an important direction for future research.


Asunto(s)
AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Miocardio/metabolismo , Hidrolasas Diéster Fosfóricas/fisiología , Receptor Cross-Talk/fisiología , Transducción de Señal/fisiología , Animales , Calcio/metabolismo , Humanos
10.
Endocrinology ; 159(5): 2142-2152, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29608743

RESUMEN

Luteinizing hormone (LH) acts on the granulosa cells that surround the oocyte in mammalian preovulatory follicles to cause meiotic resumption and ovulation. Both of these responses are mediated primarily by an increase in cyclic adenosine monophosphate (cAMP) in the granulosa cells, and the activity of cAMP phosphodiesterases (PDEs), including PDE4, contributes to preventing premature responses. However, two other cAMP-specific PDEs, PDE7 and PDE8, are also expressed at high levels in the granulosa cells, raising the question of whether these PDEs also contribute to preventing uncontrolled activation of meiotic resumption and ovulation. With the use of selective inhibitors, we show that inhibition of PDE7 or PDE8 alone has no effect on the cAMP content of follicles, and inhibition of PDE4 alone has only a small and variable effect. In contrast, a mixture of the three inhibitors elevates cAMP to a level comparable with that seen with LH. Correspondingly, inhibition of PDE7 or PDE8 alone has no effect on meiotic resumption or ovulation, and inhibition of PDE4 alone has only a partial and slow effect. However, the fraction of oocytes resuming meiosis and undergoing ovulation is increased when PDE4, PDE7, and PDE8 are simultaneously inhibited. PDE4, PDE7, and PDE8 also function together to suppress the premature synthesis of progesterone and progesterone receptors, which are required for ovulation. Our results indicate that three cAMP PDEs act in concert to suppress premature responses in preovulatory follicles.


Asunto(s)
3',5'-AMP Cíclico Fosfodiesterasas/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 7/metabolismo , Meiosis/fisiología , Oocitos/metabolismo , Ovulación/metabolismo , 3',5'-AMP Cíclico Fosfodiesterasas/antagonistas & inhibidores , Animales , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 7/antagonistas & inhibidores , Femenino , Meiosis/efectos de los fármacos , Ratones , Oocitos/efectos de los fármacos , Ovulación/efectos de los fármacos , Inhibidores de Fosfodiesterasa 4/farmacología , Inhibidores de Fosfodiesterasa/farmacología , Rolipram/farmacología
11.
Cardiovasc Res ; 62(3): 450-9, 2004 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15158137

RESUMEN

Alterations in the level and function of proteins involved in cAMP-mediated signalling are important in the pathophysiology and treatment of dilated cardiomyopathy. What is unclear is the extent to which these alterations, which attenuate receptor-stimulated cAMP generation, contribute to the pathogenesis of dilated cardiomyopathy and the extent to which they constitute a beneficial compensatory response. Studies in animals involving overexpression and ablation of proteins or peptides involved in cAMP-mediated signalling have yielded disparate results that are difficult to reconcile with a simple hypothesis. Our ability to understand these differences is limited by the lack of information on how these different genetic manipulations affect the phosphorylation of individual substrates of protein kinase A (PK-A) through which cAMP signals are transduced. This is important in view of evidence that the phosphorylation of individual PK-A substrates can be regulated selectively in different intracellular compartments, and that the phosphorylation of some PK-A substrates is increased in dilated cardiomyopathy while the phosphorylation of others is reduced. Approaches that quantify changes in the phosphorylation of individual PK-A substrates in models of dilated cardiomyopathy will provide information that may allow a better understanding of the pathogenesis of the syndrome and a more rational approach to its treatment.


Asunto(s)
Cardiomiopatía Dilatada/etiología , AMP Cíclico/metabolismo , Transducción de Señal/fisiología , Animales , Cardiomiopatía Dilatada/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Humanos , Ratones , Ratones Transgénicos , Modelos Animales , Fosforilación , Receptores Adrenérgicos beta/metabolismo
12.
Hypertension ; 66(4): 800-8, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26283042

RESUMEN

Autosomal-dominant hypertension with brachydactyly is a salt-independent Mendelian syndrome caused by activating mutations in the gene encoding phosphodiesterase 3A. These mutations increase the protein kinase A-mediated phosphorylation of phosphodiesterase 3A resulting in enhanced cAMP-hydrolytic affinity and accelerated cell proliferation. The phosphorylated vasodilator-stimulated phosphoprotein is diminished, and parathyroid hormone-related peptide is dysregulated, potentially accounting for all phenotypic features. Untreated patients die prematurely of stroke; however, hypertension-induced target-organ damage is otherwise hardly apparent. We conducted clinical studies of vascular function, cardiac functional imaging, platelet function in affected and nonaffected persons, and cell-based assays. Large-vessel and cardiac functions indeed seem to be preserved. The platelet studies showed normal platelet function. Cell-based studies demonstrated that available phosphodiesterase 3A inhibitors suppress the mutant isoforms. However, increasing cGMP to indirectly inhibit the enzyme seemed to have particular use. Our results shed more light on phosphodiesterase 3A activation and could be relevant to the treatment of severe hypertension in the general population.


Asunto(s)
Braquidactilia/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/genética , ADN/genética , Hipertensión/congénito , Mutación , Adolescente , Adulto , Presión Sanguínea/fisiología , Braquidactilia/diagnóstico , Braquidactilia/enzimología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/metabolismo , Análisis Mutacional de ADN , Ecocardiografía Doppler de Pulso , Femenino , Humanos , Hipertensión/diagnóstico , Hipertensión/enzimología , Hipertensión/genética , Immunoblotting , Imagen por Resonancia Cinemagnética , Masculino , Persona de Mediana Edad , Adulto Joven
13.
Nat Genet ; 47(6): 647-53, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25961942

RESUMEN

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.


Asunto(s)
Braquidactilia/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/genética , Hipertensión/congénito , Adolescente , Adulto , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Estudios de Casos y Controles , Diferenciación Celular , Niño , Femenino , Estudios de Asociación Genética , Células HeLa , Humanos , Hipertensión/genética , Cinética , Masculino , Células Madre Mesenquimatosas/fisiología , Ratones , Persona de Mediana Edad , Datos de Secuencia Molecular , Mutación Missense , Miocitos del Músculo Liso/fisiología , Linaje
14.
Expert Opin Investig Drugs ; 11(11): 1529-36, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12437500

RESUMEN

PDE3 cyclic nucleotide phosphodiesterases are important in cyclic AMP (cAMP) and possibly cyclic GMP-mediated signalling in cardiac and vascular smooth muscle myocytes. Drugs that inhibit these enzymes have inotropic and vasodilatory actions that have proven useful in the short-term treatment of contractile failure and pulmonary hypertension in dilated cardiomyopathy (both ischaemic and idiopathic). With long-term usage, however, these drugs appear to increase mortality in treated patients through an as yet undetermined mechanism that is in some way attributable to an increase in intracellular cAMP content in cardiac myocytes. Several recent clinical trials have raised the possibility that these drugs may be used to advantage in dilated cardiomyopathy when they are administered in combination with beta-adrenoceptor antagonists, which act to lower intracellular cAMP content. In this review, the relevant basic and clinical data are examined and the possible justification for the combination of two therapies with seemingly opposite effects on intracellular cAMP content is considered.


Asunto(s)
3',5'-AMP Cíclico Fosfodiesterasas/antagonistas & inhibidores , Cardiomiopatía Dilatada/tratamiento farmacológico , Inhibidores de Fosfodiesterasa/uso terapéutico , 3',5'-AMP Cíclico Fosfodiesterasas/metabolismo , Antagonistas Adrenérgicos beta/farmacología , Antagonistas Adrenérgicos beta/uso terapéutico , Animales , Cardiomiopatía Dilatada/enzimología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3 , Humanos , Inhibidores de Fosfodiesterasa/farmacología , Receptores Adrenérgicos beta/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
16.
Development ; 136(11): 1869-78, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19429786

RESUMEN

Mammalian oocytes are arrested in meiotic prophase by an inhibitory signal from the surrounding somatic cells in the ovarian follicle. In response to luteinizing hormone (LH), which binds to receptors on the somatic cells, the oocyte proceeds to second metaphase, where it can be fertilized. Here we investigate how the somatic cells regulate the prophase-to-metaphase transition in the oocyte, and show that the inhibitory signal from the somatic cells is cGMP. Using FRET-based cyclic nucleotide sensors in follicle-enclosed mouse oocytes, we find that cGMP passes through gap junctions into the oocyte, where it inhibits the hydrolysis of cAMP by the phosphodiesterase PDE3A. This inhibition maintains a high concentration of cAMP and thus blocks meiotic progression. LH reverses the inhibitory signal by lowering cGMP levels in the somatic cells (from approximately 2 microM to approximately 80 nM at 1 hour after LH stimulation) and by closing gap junctions between the somatic cells. The resulting decrease in oocyte cGMP (from approximately 1 microM to approximately 40 nM) relieves the inhibition of PDE3A, increasing its activity by approximately 5-fold. This causes a decrease in oocyte cAMP (from approximately 700 nM to approximately 140 nM), leading to the resumption of meiosis.


Asunto(s)
AMP Cíclico/metabolismo , GMP Cíclico/fisiología , Meiosis/fisiología , Oocitos/fisiología , Animales , Células Cultivadas , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3 , Femenino , Uniones Comunicantes/efectos de los fármacos , Uniones Comunicantes/fisiología , Humanos , Hormona Luteinizante/farmacología , Hormona Luteinizante/fisiología , Meiosis/efectos de los fármacos , Ratones , Oocitos/efectos de los fármacos , Folículo Ovárico/efectos de los fármacos , Folículo Ovárico/fisiología
17.
Am J Cardiol ; 103(5): 709-12, 2009 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-19231338

RESUMEN

An increasing number of patients are living with ventricular assist devices (VADs). Many of these patients will require noncardiac surgery for conditions not directly related to their VADs. The aim of this study was to assess the risks and outcomes of noncardiac surgery in these patients. Perioperative and follow-up data from patients with VADs who underwent noncardiac surgery from 1993 to 2006 were analyzed. In that period, 184 VADs were implanted in 155 patients. Thirty-seven patients (24%) subsequently underwent 59 noncardiac surgeries. The mean duration of VAD support before surgery was 229 days. Bleeding was the most common postsurgical complication (10%), necessitating reexploration in 20% of abdominal surgeries. Thirty-day mortality was 12%. No deaths were caused by direct complications of surgery. Successful transplantation occurred in 72% of bridge to transplantation patients who required noncardiac surgery, compared with 71% of these patients who did not require noncardiac surgery (relative risk 1.0, p = 0.9). The average duration of VAD support after noncardiac surgery for destination therapy patients was 324 days, most of which time was spent at home. In conclusion, outcomes after noncardiac surgery in patients with VADs are favorable, and most patients continue to benefit from the intended purpose of mechanical circulatory support after recovering from noncardiac surgery.


Asunto(s)
Corazón Auxiliar , Procedimientos Quirúrgicos Operativos , Femenino , Trasplante de Corazón , Corazón Auxiliar/efectos adversos , Humanos , Cuidados Intraoperatorios , Masculino , Persona de Mediana Edad , Complicaciones Posoperatorias , Cuidados Preoperatorios , Procedimientos Quirúrgicos Operativos/mortalidad , Resultado del Tratamiento
18.
J Heart Lung Transplant ; 28(11): 1129-34, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19782589

RESUMEN

BACKGROUND: Organ transplant candidates with serum antibodies directed against human leukocyte antigens (HLA) face longer waiting times and higher mortality while awaiting transplantation. This study examined the accuracy of virtual crossmatch, in which recipient HLA-specific antibodies, identified by solid-phase assays, are compared to the prospective donor HLA-type in heart transplantation. METHODS: We examined the accuracy of virtual crossmatch in predicting immune compatibility of donors and recipients in heart transplantation and clinical outcomes in immunologically sensitized heart transplant recipients in whom virtual crossmatch was used in allograft allocation. RESULTS: Based on analysis of 257 T-cell antihuman immunoglobulin complement-dependent cytotoxic (AHG-CDC) crossmatch tests, the positive predictive value of virtual crossmatch (the likelihood of an incompatible virtual crossmatch resulting in an incompatible T-cell CDC-AHG crossmatch) was 79%, and the negative predictive value of virtual crossmatch (the likelihood of a compatible virtual crossmatch resulting in a compatible T-cell CDC-AHG crossmatch) was 92%. When used in a cohort of 28 sensitized patients awaiting heart transplantation, 14 received allografts based on a compatible virtual crossmatch alone from donors in geographically distant locations. Compared with the other 14 sensitized patients who underwent transplant after a compatible prospective serologic crossmatch, the rejection rates and survival were similar. CONCLUSION: Our findings are evidence of the accuracy of virtual crossmatch and its utility in augmenting the opportunities for transplantation of sensitized patients.


Asunto(s)
Trasplante de Corazón/inmunología , Biomarcadores/sangre , Fibrosis Endomiocárdica/epidemiología , Fibrosis Endomiocárdica/mortalidad , Estudios de Seguimiento , Prueba de Histocompatibilidad/métodos , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/sangre , Complicaciones Posoperatorias/epidemiología , Complicaciones Posoperatorias/inmunología , Complicaciones Posoperatorias/mortalidad , Complicaciones Posoperatorias/prevención & control , Procolágeno-Prolina Dioxigenasa/sangre , Factores de Tiempo , Interfaz Usuario-Computador , Factor A de Crecimiento Endotelial Vascular/sangre
19.
J Biol Chem ; 282(45): 32749-57, 2007 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-17726023

RESUMEN

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.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 1/metabolismo , Miocitos Cardíacos/enzimología , Animales , Línea Celular , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 1/genética , Regulación Enzimológica de la Expresión Génica , Humanos , Hidrólisis , Isoenzimas/metabolismo , Cinética , Spodoptera , Fracciones Subcelulares/enzimología
20.
Expert Opin Investig Drugs ; 15(7): 733-42, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16787138

RESUMEN

Cyclic nucleotide phosphodiesterases (PDE) 3 and 5 regulate cAMP and cGMP signalling in cardiac and smooth muscle myocytes. Important advances in the understanding of the roles of these enzymes have recently been made. PDE3 inhibitors have inotropic and vasodilatory properties, and although they acutely improve haemodynamics in patients with heart failure, they do not improve long-term morbidity and mortality. Although combination therapy with beta-adrenergic receptor antagonists or selective inhibition of specific PDE3 isoforms might result in a more favourable long-term outcome, more clinical data are needed to test this proposition. The role of PDE5 inhibitors in the treatment of cardiac disease is evolving. PDE5 inhibitors cause pulmonary and systemic vasodilation. How these drugs will compare with other vasodilators in terms of long-term outcomes in patients with heart failure is unknown. Recent studies also suggest that PDE5 inhibitors may have antihypertropic effects, exerted through increased myocardial cGMP signalling, that could be of additional benefit in patients with heart failure.


Asunto(s)
3',5'-AMP Cíclico Fosfodiesterasas/antagonistas & inhibidores , 3',5'-GMP Cíclico Fosfodiesterasas/antagonistas & inhibidores , Cardiotónicos/uso terapéutico , Drogas en Investigación/uso terapéutico , Insuficiencia Cardíaca/tratamiento farmacológico , Inhibidores de Fosfodiesterasa/uso terapéutico , Vasodilatadores/uso terapéutico , 3',5'-AMP Cíclico Fosfodiesterasas/clasificación , 3',5'-AMP Cíclico Fosfodiesterasas/fisiología , 3',5'-GMP Cíclico Fosfodiesterasas/clasificación , 3',5'-GMP Cíclico Fosfodiesterasas/fisiología , Antagonistas Adrenérgicos beta/administración & dosificación , Antagonistas Adrenérgicos beta/uso terapéutico , Animales , Cardiomiopatía Hipertrófica/tratamiento farmacológico , Cardiomiopatía Hipertrófica/enzimología , Cardiomiopatía Hipertrófica/prevención & control , Cardiotónicos/farmacología , Circulación Coronaria/efectos de los fármacos , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3 , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5 , Evaluación Preclínica de Medicamentos , Quimioterapia Combinada , Drogas en Investigación/farmacología , Activación Enzimática/efectos de los fármacos , Predicción , Semivida , Insuficiencia Cardíaca/complicaciones , Insuficiencia Cardíaca/enzimología , Humanos , Hipertensión Pulmonar/tratamiento farmacológico , Hipertensión Pulmonar/enzimología , Hipertensión Pulmonar/etiología , Isoenzimas/antagonistas & inhibidores , Isoenzimas/fisiología , Estudios Multicéntricos como Asunto , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/enzimología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/enzimología , Inhibidores de Fosfodiesterasa/farmacología , Fosforilación/efectos de los fármacos , Estudios Prospectivos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Circulación Pulmonar/efectos de los fármacos , Ensayos Clínicos Controlados Aleatorios como Asunto , Ratas , Resultado del Tratamiento , Vasodilatadores/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA