Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Int J Urol ; 24(6): 412-424, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28332240

RESUMO

It is well known that there is an association of lower urinary tract symptoms/benign prostatic hypertrophy with cardiovascular disease, suggesting that lower urinary tract symptoms/benign prostatic hypertrophy is a risk factor for cardiovascular events. Vascular function, including endothelial function and vascular smooth muscle function, is involved in the pathogenesis, maintenance and development of atherosclerosis, leading to cardiovascular events. Vascular dysfunction per se should also contribute to lower urinary tract symptoms/benign prostatic hypertrophy. Both lower urinary tract symptoms/benign prostatic hypertrophy and vascular dysfunction have cardiovascular risk factors, such as hypertension, dyslipidemia, diabetes mellitus, aging, obesity and smoking. Inactivation of the phosphodiesterase type 5-cyclic guanosine 3',5'-monophosphate-nitric oxide pathway causes lower urinary tract symptoms/benign prostatic hypertrophy through an enhancement of sympathetic nervous activity, endothelial dysfunction, increase in Rho-associated kinase activity and vasoconstriction, and decrease in blood flow of pelvic viscera. Both endogenous nitric oxide and exogenous nitric oxide act as vasodilators on vascular smooth muscle cells through an increase in the content of cyclic guanosine 3',5'-monophosphate, which is inactivated by phosphodiesterase type 5. In a clinical setting, phosphodiesterase type 5 inhibitors are widely used in patients with lower urinary tract symptoms/benign prostatic hypertrophy. Phosphodiesterase type 5 inhibitors might have beneficial effects on vascular function through not only inhibition of cyclic guanosine 3',5'-monophosphate degradation, but also increases in testosterone levels and nitric oxide bioavailability, increase in the number and improvement of the function of endothelial progenitor cells, and decrease in insulin resistance. In the present review, the relationships between lower urinary tract symptoms/benign prostatic hypertrophy, the phosphodiesterase type 5-nitric oxide-cyclic guanosine 3',5'-monophosphate pathway, vascular function and cardiovascular outcomes are examined.


Assuntos
Doenças Cardiovasculares/fisiopatologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/fisiologia , Sintomas do Trato Urinário Inferior/fisiopatologia , Óxido Nítrico/fisiologia , Hiperplasia Prostática/fisiopatologia , 3',5'-GMP Cíclico Fosfodiesterases/fisiologia , Animais , Aterosclerose/fisiopatologia , Sistema Nervoso Autônomo/fisiopatologia , Endotélio Vascular/fisiopatologia , Feminino , Humanos , Masculino , Músculo Liso Vascular/fisiopatologia , Ratos , Fatores de Risco , Transdução de Sinais , Doenças Vasculares/fisiopatologia , Quinases Associadas a rho/fisiologia , Proteína rhoA de Ligação ao GTP/fisiologia
2.
Br J Cancer ; 115(7): 895-900, 2016 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-27529513

RESUMO

BACKGROUND: Phosphodiesterase 5A inhibitors (PDEIs), a common treatment for erectile dysfunction, were recently linked to an increased risk of melanoma. METHODS: We conducted two parallel case-control studies, using the Danish Nationwide Health Registries (DNHR) and the Kaiser Permanente Northern California (KPNC) electronic health records. Identifying men with histologically verified melanoma (cases) matched on birth year to 10 cancer-free controls, we estimated odds ratios (OR) for melanoma associated with high use of PDEIs (⩾100 tablets filled), adjusting for available confounders. RESULTS: We identified 7045 DNHR and 2972 KPNC cases with invasive melanoma. The adjusted OR for invasive melanoma associated with high PDEI use was 1.22 (95% confidence interval (CI), 0.99-1.49) in DNHR and 0.95 (95% CI, 0.78-1.14) in KPNC. Odds ratios were highest for localised invasive melanoma in DNHR (OR, 1.21) and melanoma in situ in KPNC (OR, 1.15), and lowest for non-localised disease in both populations (ORs 0.75 and 0.61, respectively). The increased ORs were slightly attenuated upon adjustment for markers of health-care utilisation. CONCLUSIONS: We found little evidence for a causal association between PDEI use and risk of melanoma. The marginally increased risk of early stage disease likely resulted from more frequent health-care contacts among PDEI users.


Assuntos
Melanoma/epidemiologia , Inibidores da Fosfodiesterase 5/efeitos adversos , Citrato de Sildenafila/efeitos adversos , Idoso , California/epidemiologia , Estudos de Casos e Controles , Causalidade , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/fisiologia , Dinamarca/epidemiologia , Relação Dose-Resposta a Droga , Humanos , Masculino , Melanoma/enzimologia , Melanoma/patologia , Pessoa de Meia-Idade , Invasividade Neoplásica , Proteínas de Neoplasias/fisiologia , Estadiamento de Neoplasias , Razão de Chances , Aceitação pelo Paciente de Cuidados de Saúde , Inibidores da Fosfodiesterase 5/uso terapêutico , Proteínas Proto-Oncogênicas B-raf/fisiologia , Citrato de Sildenafila/uso terapêutico
3.
Circ Res ; 115(1): 176-88, 2014 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-24951766

RESUMO

The right ventricle (RV) is the major determinant of functional state and prognosis in pulmonary arterial hypertension. RV hypertrophy (RVH) triggered by pressure overload is initially compensatory but often leads to RV failure. Despite similar RV afterload and mass some patients develop adaptive RVH (concentric with retained RV function), while others develop maladaptive RVH, characterized by dilatation, fibrosis, and RV failure. The differentiation of adaptive versus maladaptive RVH is imprecise, but adaptive RVH is associated with better functional capacity and survival. At the molecular level, maladaptive RVH displays greater impairment of angiogenesis, adrenergic signaling, and metabolism than adaptive RVH, and these derangements often involve the left ventricle. Clinically, maladaptive RVH is characterized by increased N-terminal pro-brain natriuretic peptide levels, troponin release, elevated catecholamine levels, RV dilatation, and late gadolinium enhancement on MRI, increased (18)fluorodeoxyglucose uptake on positron emission tomography, and QTc prolongation on the ECG. In maladaptive RVH there is reduced inotrope responsiveness because of G-protein receptor kinase-mediated downregulation, desensitization, and uncoupling of ß-adrenoreceptors. RV ischemia may result from capillary rarefaction or decreased right coronary artery perfusion pressure. Maladaptive RVH shares metabolic abnormalities with cancer including aerobic glycolysis (resulting from a forkhead box protein O1-mediated transcriptional upregulation of pyruvate dehydrogenase kinase), and glutaminolysis (reflecting ischemia-induced cMyc activation). Augmentation of glucose oxidation is beneficial in experimental RVH and can be achieved by inhibition of pyruvate dehydrogenase kinase, fatty acid oxidation, or glutaminolysis. Therapeutic targets in RV failure include chamber-specific abnormalities of metabolism, angiogenesis, adrenergic signaling, and phosphodiesterase-5 expression. The ability to restore RV function in experimental models challenges the dogma that RV failure is irreversible without regression of pulmonary vascular disease.


Assuntos
Insuficiência Cardíaca/fisiopatologia , Hipertensão Pulmonar/fisiopatologia , Hipertrofia Ventricular Direita/fisiopatologia , Neovascularização Patológica/etiologia , Receptores Adrenérgicos beta/fisiologia , Transdução de Sinais/fisiologia , Animais , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/fisiologia , Hipertensão Pulmonar Primária Familiar , Fibrose , Glicólise , Ventrículos do Coração/anatomia & histologia , Ventrículos do Coração/metabolismo , Ventrículos do Coração/patologia , Humanos , Hipertrofia Ventricular Direita/complicações , Função Ventricular Direita
4.
J Urol ; 190(4): 1430-5, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23545097

RESUMO

PURPOSE: Phosphodiesterase type 5 inhibitors were recently introduced as a new treatment option for men with lower urinary tract symptoms. Safety and clinical effectiveness are well documented but the mode of action is still unclear. We determined and compared the expression of phosphodiesterase type 5 in the spinal cord of normal (sham operated) rats and rats with partial urethral obstruction induced bladder overactivity. We also assessed the urodynamic effects of intravenously and intrathecally administered sildenafil in the rats to determine whether phosphodiesterase type 5 inhibitors exert effects on the sacral spinal cord. MATERIALS AND METHODS: A total of 65 male Sprague-Dawley rats were used for molecular/morphological and functional experiments. Bladder overactivity was induced via surgical partial urethral obstruction in 39 of 65 rats. Spinal phosphodiesterase type 5 expression was assessed by histology and polymerase chain reaction. The effects of sildenafil administered intravenously or intrathecally were studied urodynamically. RESULTS: Phosphodiesterase type 5 was expressed in various regions of the lumbosacral spinal cord, including the sacral regions of micturition control. Expression was similar in normal rats and rats with partial urethral obstruction/bladder overactivity. In normal rats intravenous and intrathecal sildenafil had no urodynamic effect. When administered intravenously and intrathecally to rats with partial urethral obstruction/bladder overactivity, sildenafil decreased micturition frequency and bladder pressure. Doses tested intrathecally had no effect when given intravenously. CONCLUSIONS: Phosphodiesterase type 5 is expressed in the rat spinal cord. Intravenous sildenafil may exert part of its urodynamic effect in rats with partial urethral obstruction/bladder overactivity via an effect on the sacral spinal cord.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/fisiologia , Medula Espinal/metabolismo , Obstrução do Colo da Bexiga Urinária/complicações , Bexiga Urinária Hiperativa/etiologia , Animais , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/análise , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/biossíntese , Masculino , Inibidores da Fosfodiesterase 5/farmacologia , Piperazinas/farmacologia , Purinas/farmacologia , Ratos , Ratos Sprague-Dawley , Citrato de Sildenafila , Medula Espinal/química , Medula Espinal/efeitos dos fármacos , Sulfonas/farmacologia
5.
Neurobiol Learn Mem ; 97(4): 370-9, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22426465

RESUMO

A promising target for memory improvement is phosphodiesterase type 5 (PDE5), which selectively hydrolyzes cyclic guanosine monophosphate (cGMP). In rodents, PDE5 inhibitors (PDE5-Is) have been shown to improve memory performance in many behavioral paradigms. However, it is questioned whether the positive effects in animal studies result from PDE5 inhibition in the central nervous system or the periphery. Therefore, we studied the effects of PDE5 inhibition on memory and determined whether compound penetration of the blood-brain barrier (BBB) is required for this activity. Two selective PDE5-Is, vardenafil and UK-343,664, were tested in the object recognition task (ORT) in both a MK-801- and scopolamine-induced memory deficit model, and a time-delay model without pharmacological intervention. Compounds were dosed 30 min before the learning trial of the task. To determine if the PDE5-Is crossed the BBB, their concentrations were determined in plasma and brain tissue collected 30 min after oral administration. Vardenafil improved object recognition memory in all three variants of the ORT. UK-343,664 was ineffective at either preventing MK-801-induced memory disruption or time-dependent memory decay. However, UK-343,664 attenuated the memory impairment of scopolamine. Vardenafil crossed the BBB whereas UK-343,664 did not. Further, co-administration of UK-343,664 and scopolamine did not alter the brain partitioning of either molecule. This suggests that the positive effect of UK-343,664 on scopolamine-induced memory decay might arise from peripheral PDE5 inhibition. The results herein suggest that there may be multiple mechanisms that mediate the efficacy of PDE5 inhibition to improve memory performance in tasks such as the ORT and that these involve PDE5 located both within and outside of the brain. To further elucidate the underlying mechanisms, the cellular and subcellular localization of PDE5 needs to be determined.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/fisiologia , Imidazóis/farmacologia , Inibidores da Fosfodiesterase 5/farmacologia , Piperazinas/farmacologia , Pirimidinonas/farmacologia , Reconhecimento Psicológico/fisiologia , Animais , Barreira Hematoencefálica , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/efeitos dos fármacos , Maleato de Dizocilpina/farmacocinética , Maleato de Dizocilpina/farmacologia , Imidazóis/farmacocinética , Masculino , Inibidores da Fosfodiesterase 5/farmacocinética , Piperazinas/farmacocinética , Pirimidinonas/farmacocinética , Ratos , Ratos Wistar , Reconhecimento Psicológico/efeitos dos fármacos , Escopolamina/farmacocinética , Escopolamina/farmacologia , Sulfonas/farmacocinética , Sulfonas/farmacologia , Triazinas/farmacocinética , Triazinas/farmacologia , Dicloridrato de Vardenafila
6.
J Sex Med ; 9(1): 79-87, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21554553

RESUMO

INTRODUCTION: Priapism is defined as an erectile disorder, in which erection persists uncontrollably without sexual purpose. The precise mechanisms involved in the development of sickle cell disease-associated priapism are ill defined. AIM: To summarize the recent developments that increase our understanding of the molecular mechanisms of priapism. METHODS: This article reviews the literature (Medline search 2000-2010) that relates the key molecular signaling pathways that contribute to the development of priapism associated with sickle-cell disease. It focuses on basic science investigations using multiple animal models. MAIN OUTCOME MEASURES: The reader will be informed of the most current research regarding the role of endothelial nitric oxide synthase, phosphodiesterase type 5 (PDE5), adenosine, RhoA/Rho-kinase (ROCK), and opiorphins in the pathophysiology of priapism. RESULTS: New concepts in the field of priapism research suggest that priapism often results from altered vascular homeostatic actions in the penis and is associated with deficient erection control mechanisms on a molecular level. A leading proposal in this regard is the notion of aberrant signaling of the endothelium-derived nitric oxide and PDE5 signal transduction pathway in the penis. Additionally, dysfunctional regulatory control of signal transduction systems which interact with this pathway such as adenosine and RhoA/Rho-kinase may contribute to the development of priapism. Recent investigations of opiorphins also demonstrate a role in regulating corporal smooth muscle tone and thereby dysregulation of erection physiology in priapism. These advances have paved the way for understanding this disorder as having a molecular pathogenesis. CONCLUSIONS: As the science underlying priapism further emerges, increasingly effective therapeutics for sickle cell disease-associated priapism is certain to follow.


Assuntos
Anemia Falciforme/complicações , Priapismo/etiologia , Adenosina/fisiologia , Anemia Falciforme/fisiopatologia , Animais , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/fisiologia , Humanos , Masculino , Camundongos , Óxido Nítrico Sintase Tipo III/fisiologia , Oligopeptídeos/fisiologia , Ereção Peniana/fisiologia , Priapismo/fisiopatologia , Proteínas e Peptídeos Salivares/fisiologia , Transdução de Sinais/fisiologia , Quinases Associadas a rho/fisiologia , Proteína rhoA de Ligação ao GTP/fisiologia
7.
Handb Exp Pharmacol ; (204): 323-44, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21695647

RESUMO

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.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/fisiologia , Distrofia Muscular de Duchenne/tratamento farmacológico , Inibidores da Fosfodiesterase 5/uso terapêutico , Animais , GMP Cíclico/fisiologia , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos mdx , Músculo Esquelético/enzimologia , Distrofia Muscular de Duchenne/etiologia , Miócitos Cardíacos/enzimologia , Óxido Nítrico/fisiologia , Óxido Nítrico Sintase Tipo I/fisiologia , Transdução de Sinais
8.
Ann N Y Acad Sci ; 1487(1): 21-30, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32860248

RESUMO

The nitric oxide (NO)-cyclic guanosine monophosphate (cGMP)-protein kinase G (PKG) pathway plays a critical role in skeletal homeostasis. Preclinical data using NO and its donors and genetically modified mice demonstrated that NO was required in bone remodeling and partly mediated the anabolic effects of mechanical stimuli and estrogen. However, the off-target effects and tachyphylaxis of NO limit its long-term use, and previous clinical trials using organic nitrates for osteoporosis have been disappointing. Among the other components in the downstream pathway, targeting cGMP-specific phosphodiesterase to promote the NO-cGMP-PKG signal is a viable option. There are growing in vitro and in vivo data that, among many other PDE families, PDE5A is highly expressed in skeletal tissue, and inhibiting PDE5A using currently available PDE5A inhibitors might increase the osteoanabolic signal and protect the skeleton. These preclinical data open the possibility of repurposing PDE5A inhibitors for treating osteoporosis. Further research is needed to address the primary target bone cell of PDE5A inhibition, the contribution of direct and indirect effects of PDE5A inhibition, and the pathophysiological changes in skeletal PDE5A expression in aging and hypogonadal animal models.


Assuntos
Remodelação Óssea/fisiologia , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Óxido Nítrico/metabolismo , Animais , Osso e Ossos/fisiologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/fisiologia , Humanos , Camundongos , Transdução de Sinais/fisiologia
9.
Circulation ; 119(3): 408-16, 2009 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-19139381

RESUMO

BACKGROUND: Ventricular expression of phosphodiesterase-5 (PDE5), an enzyme responsible for cGMP catabolism, is increased in human right ventricular hypertrophy, but its role in left ventricular (LV) failure remains incompletely understood. We therefore measured LV PDE5 expression in patients with advanced systolic heart failure and characterized LV remodeling after myocardial infarction in transgenic mice with cardiomyocyte-specific overexpression of PDE5 (PDE5-TG). METHODS AND RESULTS: Immunoblot and immunohistochemistry techniques revealed that PDE5 expression was greater in explanted LVs from patients with dilated and ischemic cardiomyopathy than in control hearts. To evaluate the impact of increased ventricular PDE5 levels on cardiac function, PDE5-TG mice were generated. Confocal and immunoelectron microscopy revealed increased PDE5 expression in cardiomyocytes, predominantly localized to Z-bands. At baseline, myocardial cGMP levels, cell shortening, and calcium handling in isolated cardiomyocytes and LV hemodynamic measurements were similar in PDE5-TG and wild-type littermates. Ten days after myocardial infarction, LV cGMP levels had increased to a greater extent in wild-type mice than in PDE5-TG mice (P<0.05). Ten weeks after myocardial infarction, LV end-systolic and end-diastolic volumes were larger in PDE5-TG than in wild-type mice (57+/-5 versus 39+/-4 and 65+/-6 versus 48+/-4 muL, respectively; P<0.01 for both). LV systolic dysfunction and diastolic dysfunction were more marked in PDE5-TG than in wild-type mice, associated with enhanced hypertrophy and reduced contractile function in isolated cardiomyocytes from remote myocardium. CONCLUSIONS: Increased PDE5 expression predisposes mice to adverse LV remodeling after myocardial infarction. Increased myocardial PDE5 expression in patients with advanced cardiomyopathy may contribute to the development of heart failure and represents an important therapeutic target.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/biossíntese , Regulação Enzimológica da Expressão Gênica/fisiologia , Insuficiência Cardíaca/enzimologia , Infarto do Miocárdio/enzimologia , Remodelação Ventricular/genética , Animais , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/genética , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/fisiologia , Insuficiência Cardíaca/fisiopatologia , Ventrículos do Coração/enzimologia , Ventrículos do Coração/fisiopatologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Infarto do Miocárdio/fisiopatologia , Miocárdio/enzimologia , Miocárdio/patologia
10.
Kidney Int ; 78(7): 635-7, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20842148

RESUMO

The serine/threonine protease corin, which proteolytically activates atrial natriuretic peptide (ANP), is reduced in the kidneys of animals with nephrotic syndrome and glomerular nephritis. Polzin et al. provide evidence for a linkage between the decreased corin and ß-epithelial sodium channel, phosphodiesterase 5, and cGMP-dependent protein kinase II in the nephrotic kidney. They propose that decreases in cGMP resulting from the reduced corin may be responsible for the Na(+) retention and volume expansion that are hallmarks of these kidney diseases.


Assuntos
Síndrome Nefrótica/metabolismo , Serina Endopeptidases/fisiologia , Sódio/metabolismo , Absorção , Animais , Fator Natriurético Atrial/metabolismo , GMP Cíclico/fisiologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/fisiologia , Humanos
11.
Am J Physiol Regul Integr Comp Physiol ; 298(2): R433-8, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19955496

RESUMO

It has been suggested that the phosphodiesterase-5 (PDE5) inhibitor sildenafil may be useful in the treatment of hypertension during pregnancy. However, we have reported a selective increase in renal inner medullary PDE5 that participates in the sodium retention of pregnancy. Therefore, the purpose of this study was to determine whether oral sildenafil treatment impairs maternal plasma volume expansion and/or fetal growth during rat pregnancy. Rats received sildenafil (10 mg x kg(-1) x day(-1), 50 mg x kg(-1) x day(-1), or 90 mg x kg(-1) x day(-1)) or vehicle on days 4-20 of pregnancy. On days 14-19, rats were housed in metabolic cages for collection of urine and measurement of food and water intake. Terminal hemodynamic and fetal measurements were taken on day 20. None of the sildenafil doses lowered blood pressure, and although all doses increased plasma cGMP concentrations, only the highest dose increased aortic and inner medullary cGMP content. Sildenafil had no effect on maternal weight gain; however, the highest dose decreased both plasma volume and renal sodium retention. The pup number and size were similar among the groups. Therefore, these studies suggest that low doses of systemic sildenafil may be safe during pregnancy in the rat, but higher doses may interfere with the physiological sodium retention and volume expansion of pregnancy. The effects of systemic sildenafil on blood pressure and sodium retention during hypertension in human pregnancy remain to be examined.


Assuntos
Desenvolvimento Fetal/efeitos dos fármacos , Hemodinâmica/efeitos dos fármacos , Inibidores de Fosfodiesterase/farmacologia , Piperazinas/farmacologia , Prenhez/fisiologia , Sulfonas/farmacologia , Vasodilatadores/farmacologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Creatinina/sangue , Creatinina/urina , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/fisiologia , Feminino , Feto/irrigação sanguínea , Hematócrito , Inibidores da Fosfodiesterase 5 , Volume Plasmático/efeitos dos fármacos , Gravidez , Purinas/farmacologia , Ratos , Ratos Sprague-Dawley , Fluxo Sanguíneo Regional/fisiologia , Citrato de Sildenafila , Sódio/metabolismo
12.
J Sex Med ; 7(6): 2104-2111, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20487244

RESUMO

INTRODUCTION: The cyclic adenosine monophosphate-dependent protein kinase (cAK) is considered a key protein in the control of smooth muscle tone in the cardiovascular system. There is evidence that erectile dysfunction might be linked to systemic vascular disorders and arterial insufficiency, subsequently resulting in structural changes in the penile tissue. The expression and significance of cAK in human cavernous arteries (HCA) have not been evaluated. AIMS: To evaluate the expression of cAK isoforms in HCA and examine the role of cAK in the cyclic adenosine monophosphate (cAMP)- and cyclic guanosine monophosphate (cGMP)-mediated control of penile vascular smooth muscle. METHODS: The expression and distribution of phosphodiesterase type 4 (PDE4) and cAK isoforms in sections of HCA were investigated by means of immunohistochemistry and Western blot analysis. The effects of the cAK inhibitor Rp-8-CPT-cAMPS on the relaxation of isolated preparations of HCA (diameter > 100 µm) induced by rolipram, sildenafil, tadalafil, and vardenafil were studied using the organ bath technique. MAIN OUTCOME MEASURES: Investigate the expression of cAK in relation to α-actin and PDE4 in HCA and evaluate the effects of an inhibition of cAK on the relaxation induced by inhibitors of PDE4 and PDE5 of isolated penile arteries. RESULTS: Immunosignals specific for cAKIα, IIα, and IIß were observed within the wall of HCA. Double stainings revealed colocalization of cAK with α-actin and PDE4. The expression of cAK isoforms was confirmed by Western blot analysis. The reversion of tension induced by inhibitors of PDE4 and PDE5 of isolated penile vascular tissue were attenuated significantly by Rp-8-CPT-cAMPS. CONCLUSIONS: Our results demonstrate the expression of cAK isoforms in the smooth musculature of HCA and its colocalization with PDE4. A significant role for cAK in the regulation mediated by cAMP and cGMP of vascular smooth muscle tone in HCA can also be assumed.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/fisiologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/fisiologia , Músculo Liso Vascular/fisiologia , Pênis/irrigação sanguínea , Vasodilatação/fisiologia , Actinas/fisiologia , Adulto , Artérias/fisiologia , Quinases Ciclina-Dependentes/fisiologia , Humanos , Técnicas In Vitro , Masculino , Pessoa de Meia-Idade , Isoformas de Proteínas/fisiologia , Transdução de Sinais/fisiologia , Quinase Ativadora de Quinase Dependente de Ciclina
13.
Int J Neurosci ; 120(12): 746-51, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20942592

RESUMO

Although Alzheimer's disease (AD) poses a major health problem in both developing and developed countries, no definite treatment is available for its cure; hence efforts are being focused on introducing disease-modifying agents for slowing down its course. Recent studies on the effects of sildenafil on different organs have shown that PDE-5 inhibitors may offer new horizons in therapeutic treatment of pulmonary hypertension, multiple sclerosis, neuropathic pain, and age-related memory impairment. In this paper we introduce PDE-5 inhibitors as novel disease-modifying agents against AD and review the different impacts of PDE-5 inhibition on various pathogenic mechanisms leading to AD, including neuronal apoptosis, neurovascular dysfunction, neurotransmitter modulation, and impairment of neurogenesis.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/enzimologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/metabolismo , Inibidores da Fosfodiesterase 5/administração & dosagem , Doença de Alzheimer/patologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/fisiologia , Progressão da Doença , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/enzimologia , Endotélio Vascular/patologia , Humanos , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Neurônios/patologia , Inibidores da Fosfodiesterase 5/uso terapêutico
14.
Yakugaku Zasshi ; 130(11): 1427-33, 2010 Nov.
Artigo em Japonês | MEDLINE | ID: mdl-21048399

RESUMO

Calcium ions (Ca²(+)) play an essential role in homeostasis and the activity of cardiovascular cells. Ca²(+) influx across the plasma membrane induced by neurohumoral factors or mechanical stress elicits physiologically relevant timing and spatial patterns of Ca²(+) signaling, which leads to the activation of various cardiovascular functions, such as muscle contraction, gene expression, and hypertrophic growth of myocytes. A canonical transient receptor potential protein subfamily member, TRPC6, which is activated by diacylglycerol and mechanical stretch, works as an upstream regulator of the Ca²(+) signaling pathway required for pathological hypertrophy. We have recently found that the inhibition of cGMP-selective phosphodiesterase 5 (PDE5) suppresses agonist- and mechanical stretch-induced hypertrophy through inhibition of Ca²(+) influx in rat cardiomyocytes. The inhibition of PDE5 suppressed the increase in frequency of Ca²(+) spikes induced by receptor stimulation or mechanical stretch. Activation of protein kinase G by PDE5 inhibition phosphorylated TRPC6 proteins at Thr69 and prevented TRPC6-mediated Ca²(+) influx. Substitution of Ala for Thr69 in TRPC6 abolished the antihypertrophic effects of PDE5 inhibition. These results suggest that phosphorylation and functional suppression of TRPC6 underlies the prevention of cardiac hypertrophy by PDE5 inhibition. As TRPC6 proteins are also expressed in vascular smooth muscle cells and reportedly participate in vascular remodeling, TRPC6 blockade may be an effective therapeutic strategy for preventing pathologic cardiovascular remodeling.


Assuntos
Fenômenos Fisiológicos Cardiovasculares , Canais de Cátion TRPC/fisiologia , Substituição de Aminoácidos , Animais , Sinalização do Cálcio/fisiologia , Cardiomegalia/etiologia , Cardiomegalia/prevenção & controle , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/fisiologia , Desenho de Fármacos , Camundongos , Músculo Liso Vascular , Inibidores da Fosfodiesterase 5/farmacologia , Fosforilação , Ratos , Estresse Mecânico , Canais de Cátion TRPC/antagonistas & inibidores , Canais de Cátion TRPC/química , Canal de Cátion TRPC6
15.
Circ Res ; 101(11): 1084-95, 2007 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-18040025

RESUMO

Phosphodiesterase type 5A (PDE5A) selectively hydrolyzes cyclic GMP. Inhibitors of PDE5A such as sildenafil are widely used to treat erectile dysfunction, but growing evidence supports important roles for the enzyme in both the vasculature and heart. In disorders such as cardiac failure, PDE5A upregulation may contribute to a decline in cGMP and protein kinase G signaling, exacerbating dysfunction. PDE5A plays an important role in the pulmonary vasculature where its inhibition benefits patients with pulmonary hypertension. In the heart, PDE5A signaling appears compartmentalized, and its inhibition is cardioprotective against ischemia-reperfusion and antracycline toxicity, blunts acute adrenergic contractile stimulation, and can suppress chronic hypertrophy and dysfunction attributable to pressure-overload. In this review, we discuss the molecular biology, pharmacology, and physiology of PDE5A, mechanisms of vascular and cardiac regulation, and recent evidence supporting the utility of selective PDE5A inhibition for the treatment of cardiovascular disorders.


Assuntos
Sistema Cardiovascular/enzimologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/fisiologia , Animais , Doenças Cardiovasculares/tratamento farmacológico , Doenças Cardiovasculares/enzimologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/química , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/genética , Humanos , Inibidores de Fosfodiesterase/farmacologia , Inibidores de Fosfodiesterase/uso terapêutico
16.
Sex Med Rev ; 7(4): 661-668, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30987932

RESUMO

INTRODUCTION: Testosterone deficiency is known to induce endothelial dysfunction, which can lead to erectile dysfunction and/or vascular dysfunction. In some basic and clinical reports, testosterone has been shown to regulate the nitric oxide (NO)/cyclic guanosine monophosphate (cGMP) pathway and thereby influence endothelial function and endothelial progenitor cells (EPCs), which are key for the endothelial repair system. AIM: To review the association between testosterone and endothelial dysfunction focusing on NO and EPCs. METHODS: A review of relevant literature up to September 2018 was performed via PubMed. MAIN OUTCOME MEASURES: We reviewed the association between testosterone and endothelial dysfunction focusing on NO derived from endothelial NO synthase, phosphodiesterase type 5 (PDE-5), asymmetric dimethylarginine (ADMA), inflammation, and EPCs. RESULTS: Numerous articles describing the association between testosterone deficiency and endothelial dysfunction have been published. Some reports have shown that testosterone deficiency decreases NO production by altering the expression and activity of NO synthase and by regulating ADMA expression. Testosterone also regulates the expression of phosphodiesterase type 5. In addition, some basic and clinical studies have shown that testosterone affects the function and number of EPCs. However, some inconsistencies among these reports have been noted. CONCLUSION: Testosterone deficiency might cause endothelial dysfunction by decreasing NO levels through regulating the expression and activity of NO synthase and increasing ADMA expression. In addition, testosterone might affect the endothelial repair system by regulating the proliferation and migration of EPCs. Testosterone replacement therapy might be useful for treating endothelial dysfunction, considering that some reports have shown that this therapy improved NO bioavailability and EPC function. Hotta Y, Kataoka T, Kimura K. Testosterone Deficiency and Endothelial Dysfunction: Nitric Oxide, Asymmetric Dimethylarginine, and Endothelial Progenitor Cells. Sex Med Rev 2019;7:661-668.


Assuntos
Arginina/análogos & derivados , Células Progenitoras Endoteliais/fisiologia , Endotélio Vascular/fisiologia , Óxido Nítrico/fisiologia , Testosterona/deficiência , Arginina/fisiologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/fisiologia , Humanos , Masculino , Óxido Nítrico Sintase Tipo III/fisiologia , Testosterona/uso terapêutico , Vasculite/fisiopatologia
17.
J Am Heart Assoc ; 8(15): e012273, 2019 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-31311394

RESUMO

Background In murine heart failure models and in humans with diabetic-related heart hypertrophy, inhibition of phosphodiesterase 5 (PDE5) by sildenafil improves cardiac outcomes. However, the mechanism by which sildenafil improves cardiac function is unclear. We have observed a relationship between PDE5 and ß2 adrenergic receptor (ß2AR), which is characterized here as a novel mechanistic axis by which sildenafil improves symptoms of diabetic cardiomyopathy. Methods and Results Wild-type and ß2AR knockout mice fed a high fat diet (HFD) were treated with sildenafil, and echocardiogram analysis was performed. Cardiomyocytes were isolated for excitation-contraction (E-C) coupling, fluorescence resonant energy transfer, and proximity ligation assays; while heart tissues were implemented for biochemical and histological analyses. PDE5 selectively associates with ß2AR, but not ß1 adrenergic receptor, and inhibition of PDE5 with sildenafil restores the impaired response to adrenergic stimulation in HFD mice and isolated ventriculomyocytes. Sildenafil enhances ß adrenergic receptor (ßAR)-stimulated cGMP and cAMP signals in HFD myocytes. Consequently, inhibition of PDE5 leads to protein kinase G-, and to a lesser extent, calcium/calmodulin-dependent kinase II-dependent improvements in adrenergically stimulated E-C coupling. Deletion of ß2AR abolishes sildenafil's effect. Although the PDE5-ß2AR association is not altered in HFD, phosphodiesterase 3 displays an increased association with the ß2AR-PDE5 complex in HFD myocytes. Conclusions This study elucidates mechanisms by which the ß2AR-PDE5 axis can be targeted for treating diabetic cardiomyopathy. Inhibition of PDE5 enhances ß2AR stimulation of cGMP and cAMP signals, as well as protein kinase G-dependent E-C coupling in HFD myocytes.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/fisiologia , Diabetes Mellitus Tipo 2/fisiopatologia , Cardiomiopatias Diabéticas/tratamento farmacológico , Cardiomiopatias Diabéticas/fisiopatologia , Coração/fisiopatologia , Inibidores da Fosfodiesterase 5/farmacologia , Inibidores da Fosfodiesterase 5/uso terapêutico , Receptores Adrenérgicos beta 2/efeitos dos fármacos , Receptores Adrenérgicos beta 2/fisiologia , Citrato de Sildenafila/farmacologia , Citrato de Sildenafila/uso terapêutico , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
18.
Eur Respir J ; 32(1): 198-209, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18591337

RESUMO

The pulmonary vascular bed is both a source of and target for a number of vasoactive factors. Among the most important for pulmonary vascular homeostasis are factors that utilise cyclic guanosine monophosphate (cGMP) as an intracellular second messenger. These include nitric oxide and the natriuretic peptide family (atrial, brain and C-type natriuretic peptides). In the search for therapeutic strategies that engage the cGMP signalling pathway for the treatment of pulmonary arterial hypertension (PAH), inhibition of cGMP metabolism by phosphodiesterase type 5 (PDE5)-targeted compounds has proven most successful to date. One PDE5 inhibitor, sildenafil, has been shown to improve pulmonary haemodynamics and exercise capacity in patients with PAH and is now an approved treatment. Others are under investigation. An interesting, although still tentative, observation is the potential of sildenafil to reduce pulmonary vascular resistance without adversely affecting ventilation-perfusion matching. Another is the expression of phosphodiesterase type 5 in the hypertrophied right ventricle. These data suggest that phosphodiesterase type 5 inhibitors may have effects that distinguish them from other treatments for pulmonary hypertension and merit further study.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/efeitos dos fármacos , Hipertensão Pulmonar/tratamento farmacológico , Inibidores de Fosfodiesterase/farmacologia , GMP Cíclico/fisiologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/fisiologia , Humanos , Hipertensão Pulmonar/enzimologia , Inibidores de Fosfodiesterase/farmacocinética , Ensaios Clínicos Controlados Aleatórios como Assunto , Transdução de Sinais/efeitos dos fármacos , Resistência Vascular
20.
Eur J Pharmacol ; 582(1-3): 102-9, 2008 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-18234184

RESUMO

Cyclic nucleotides (cAMP and cGMP) are the main second messengers linked to vasodilatation. They are synthesized by cyclases and degraded by different types of phosphodiesterases (PDE). The effect of PDE inhibition and cyclases stimulation on 5-hydroxytryptamine (5-HT; 1 microM) and histamine (10 microM) contracted arteries was analysed. Stimulation of guanylate cyclase or adenylate cyclase relaxed the histamine- and 5-HT-induced contractions indicating that intracellular increase of cyclic nucleotides leads to vasodilatation of the human umbilical artery. We investigated the role of different PDE families in the regulation of this effect. The presence of the different PDE types in human umbilical artery smooth muscle was analysed by RT-PCR and the expression of PDE1B, PDE3A, PDE3B, PDE4C, PDE4D and PDE5A was detected. The unspecific PDE inhibitor 3-isobutyl-1-methylxanthine (IBMX; 50 microM) relaxed histamine-contracted human umbilical artery on 47.4+/-7.2%. This effect seems to be due to PDE4 and PDE5 inhibition because among the selective PDE inhibitors used only the PDE4 inhibitor (rolipram; 1 microM) and the PDE5 inhibitors (dipyridamole and T0156; 3 microM and 1 microM respectively) induced significant relaxation (39.0+/-8.7, 30.4+/-6.0 and 36.3+/-2.8 respectively). IBMX, dipyridamole and T0156 produced similar relaxation on 5-HT-induced contraction. After forskolin, the addition of IBMX or rolipram increased the effect of the adenylate cyclase stimulator and almost completely relaxed the human umbilical artery contracted by histamine (92.5+/-4.9 and 90.9+/-4.7 respectively), suggesting a main role of PDE4. The data obtained with 5-HT contracted arteries confirmed this, because only rolipram and IBMX significantly increased the forskolin vasodilator effect. The administration of dipyridamole and T0156 after sodium nitroprusside (SNP) induced a significant increase of the SNP relaxant effect on histamine-contracted arteries, but PDE1 and PDE3 inhibition did not increase the effect of the guanylate cyclase stimulator. Similar effects were obtained in 5-HT contracted arteries, the SNP induced relaxation was increased by the PDE5 inhibition, but not by PDE1 or PDE3 inhibition. In summary, our results demonstrate that: 1) the increase of cAMP and/or cGMP levels induces relaxation of the human umbilical vascular smooth muscle; 2) four families of PDE are expressed in this smooth muscle: PDE1, PDE3, PDE4 and PDE5; 3) between these families, PDE4 and PDE5 are the key enzymes involved in the regulation of the relaxation associated to cAMP and cGMP, respectively.


Assuntos
AMP Cíclico/fisiologia , GMP Cíclico/fisiologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/fisiologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/fisiologia , Músculo Liso Vascular/fisiologia , Artérias Umbilicais/fisiologia , 1-Metil-3-Isobutilxantina/farmacologia , Adenilil Ciclases/metabolismo , Colforsina/farmacologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/biossíntese , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/biossíntese , Dipiridamol/farmacologia , Feminino , Guanilato Ciclase/metabolismo , Histamina/farmacologia , Humanos , Técnicas In Vitro , Isoenzimas/antagonistas & inibidores , Isoenzimas/biossíntese , Isoenzimas/fisiologia , Contração Muscular , Relaxamento Muscular , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/enzimologia , Naftiridinas/farmacologia , Nitroprussiato/farmacologia , Inibidores da Fosfodiesterase 4 , Inibidores da Fosfodiesterase 5 , Pirimidinas/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Rolipram/farmacologia , Serotonina/farmacologia , Artérias Umbilicais/efeitos dos fármacos , Artérias Umbilicais/enzimologia , Vasodilatadores/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA