Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 220
Filtrar
1.
Int J Mol Sci ; 25(2)2024 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-38255829

RESUMEN

Fracture healing in the aged is associated with a reduced healing capacity, which often results in delayed healing or non-union formation. Many factors may contribute to this deterioration of bone regeneration, including a reduced 'angiogenic trauma response'. The phosphodiesterase-3 (PDE-3) inhibitor cilostazol has been shown to exert pro-angiogenic and pro-osteogenic effects in preclinical studies. Therefore, we herein analyzed in a stable closed femoral fracture model whether this compound also promotes fracture healing in aged mice. Forty-two aged CD-1 mice (age: 16-18 months) were daily treated with 30 mg/kg body weight cilostazol (n = 21) or vehicle (control, n = 21) by oral gavage. At 2 and 5 weeks after fracture, the femora were analyzed by X-ray, biomechanics, micro-computed tomography (µCT), histology, immunohistochemistry, and Western blotting. These analyses revealed a significantly increased bending stiffness at 2 weeks (2.2 ± 0.4 vs. 4.3 ± 0.7 N/mm) and an enhanced bone formation at 5 weeks (4.4 ± 0.7 vs. 9.1 ± 0.7 mm3) in cilostazol-treated mice when compared to controls. This was associated with a higher number of newly formed CD31-positive microvessels (3.3 ± 0.9 vs. 5.5 ± 0.7 microvessels/HPF) as well as an elevated expression of phosphoinositide-3-kinase (PI3K) (3.6 ± 0.8 vs. 17.4 ± 5.5-pixel intensity × 104) and runt-related transcription factor (RUNX)2 (6.4 ± 1.2 vs. 18.2 ± 2.7-pixel intensity × 104) within the callus tissue. These findings indicate that cilostazol accelerates fracture healing in aged mice by stimulating angiogenesis and the expression of PI3K and RUNX2. Hence, cilostazol may represent a promising compound to promote bone regeneration in geriatric patients.


Asunto(s)
Fracturas del Fémur , Fosfatidilinositol 3-Quinasa , Animales , Femenino , Masculino , Ratones , Angiogénesis , Cilostazol/farmacología , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Curación de Fractura , Fosfatidilinositol 3-Quinasas , Inhibidores de Fosfodiesterasa 3/farmacología , Inhibidores de Fosfodiesterasa 3/uso terapéutico , Microtomografía por Rayos X
3.
Toxicon ; 220: 106922, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36167141

RESUMEN

The mechanisms of pathogenesis of acute kidney injury (AKI) in snakebites is multifactorial and involves hemodynamic disturbances, with release of free radical causing cytotoxic effects. The phosphodiesterase-3 (PDE3) inhibitor, Cilostazol, has been reported to provide protection against renal oxidative stress. OBJECTIVE: We evaluated the protective effects of cilostazol against Bothrops alternatus snake venom (BaV)-induced nephrotoxicity. METHODS: Wistar rat kidneys (n = 6, 260-300 g) were isolated and perfused with Krebs-Henseleit solution containing 6 g/100 mL of bovine serum albumin. After 30 min, the kidneys were perfused with BaV to a final concentration of 1 and 3 µg/mL, and subsequently evaluated for perfusion pressure (PP), renal vascular resistance (RVR), urinary flow (UF), glomerular filtration rate (GFR), and percentage of electrolyte tubular sodium and chloride transport (%TNa+, %TCl-). Oxidative stress and renal histological analyses were performed. RESULTS: BaV caused a reduction in all the evaluated renal parameters (PP, RVR, GFR, UF, %TNa+, and %TCl-). Although only the effects on PP and UF were reversed with cilostazol treatment, the decrease in the malondialdehyde levels, without changes in glutathione levels, further reduced the venom-induced renal tissue changes. CONCLUSION: Our data suggest that PDE3 is involved in BaV-induced nephrotoxicity, as cilostazol administration significantly ameliorated these effects.


Asunto(s)
Lesión Renal Aguda , Bothrops , Venenos de Crotálidos , Animales , Ratas , Venenos de Crotálidos/farmacología , Cilostazol/farmacología , Inhibidores de Fosfodiesterasa 3/farmacología , Ratas Wistar , Riñón , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/tratamiento farmacológico , Lesión Renal Aguda/patología , Venenos de Serpiente/farmacología , Oxidación-Reducción , Hidrolasas Diéster Fosfóricas/farmacología
4.
Naunyn Schmiedebergs Arch Pharmacol ; 395(7): 789-801, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35384464

RESUMEN

Atherosclerosis is a multifactorial chronic disease associated with pro-inflammatory and pro-oxidative cardiovascular states. Cilostazol, a selective phosphodiesterase 3 inhibitor (PDE3), is clinically used in the treatment of intermittent claudication and secondary prevention of cerebral infarction. The aim of this study was to evaluate the cardioprotective effects of cilostazol and the molecular mechanisms involved in hypercholesterolemic rats. Male Wistar rats were divided into four groups: control group (C) and control + cilostazol group (C+CILO), that were fed a standard chow diet, and hypercholesterolemic diet group (HCD) and HCD + cilostazol (HCD+CILO) that were fed a hypercholesterolemic diet. Cilostazol treatment started after 30 days for C+CILO and HCD+CILO groups. Animals were administered cilostazol once a day for 15 days. Subsequently, serum and left ventricles were extracted for evaluation of lipid profile, inflammatory, and oxidative biomarkers. The HCD group displayed increased serum lipid levels, inflammatory cytokines production, and cardiac NF-kB protein expression and decreased cardiac Nrf2-mediated antioxidant activity. Conversely, the cilostazol treatment improved all these cardiac deleterious effects, inhibiting NF-kB activation and subsequently decreasing inflammatory mediators, reestablishing the antioxidant properties through Nrf2-mediated pathway, including increased SOD, GPx, and catalase expression. Taken together, our results indicated that cilostazol protects hypercholesterolemia-induced cardiac damage by molecular mechanisms targeting the crosstalk between Nrf2 induction and NF-kB inhibition in the heart.


Asunto(s)
Factor 2 Relacionado con NF-E2 , FN-kappa B , Animales , Antioxidantes/metabolismo , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Cilostazol/farmacología , Inflamación/tratamiento farmacológico , Lípidos , Masculino , Factor 2 Relacionado con NF-E2/metabolismo , FN-kappa B/metabolismo , Estrés Oxidativo , Inhibidores de Fosfodiesterasa 3/farmacología , Inhibidores de Fosfodiesterasa 3/uso terapéutico , Ratas , Ratas Wistar
5.
J Clin Pharmacol ; 62(3): 320-358, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34671983

RESUMEN

Cilostazol, a phosphodiesterase III inhibitor, has vasodilating and antiplatelet properties with a low rate of bleeding complications. It has been used over the past 25 years for improving intermittent claudication in patients with peripheral artery disease (PAD). Cilostazol also has demonstrated efficacy in patients undergoing percutaneous revascularization procedures for both PAD and coronary artery disease. In addition to its antithrombotic and vasodilating actions, cilostazol also inhibits vascular smooth muscle cell proliferation via phosphodiesterase III inhibition, thus mitigating restenosis. Accumulated evidence has shown that cilostazol, due to its "pleiotropic" effects, is a useful, albeit underutilized, agent for both coronary artery disease and PAD. It is also potentially useful after ischemic stroke and is an alternative in those who are allergic or intolerant to classical antithrombotic agents (eg, aspirin or clopidogrel). These issues are herein reviewed together with the pharmacology and pharmacodynamics of cilostazol. Large studies and meta-analyses are presented and evaluated. Current guidelines are also discussed, and the spectrum of cilostazol's actions and therapeutic applications are illustrated.


Asunto(s)
Cilostazol/farmacología , Cilostazol/uso terapéutico , Inhibidores de Fosfodiesterasa 3/farmacología , Inhibidores de Fosfodiesterasa 3/uso terapéutico , Animales , Cilostazol/efectos adversos , Cilostazol/farmacocinética , Enfermedad de la Arteria Coronaria/tratamiento farmacológico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Quimioterapia Combinada , Terapia Antiplaquetaria Doble/métodos , Hemorragia/inducido químicamente , Humanos , Claudicación Intermitente/tratamiento farmacológico , Lípidos/sangre , Metaanálisis como Asunto , Músculo Liso Vascular/efectos de los fármacos , Intervención Coronaria Percutánea/métodos , Enfermedad Arterial Periférica/tratamiento farmacológico , Inhibidores de Fosfodiesterasa 3/efectos adversos , Inhibidores de Fosfodiesterasa 3/farmacocinética , Ensayos Clínicos Controlados Aleatorios como Asunto , Insuficiencia Renal/tratamiento farmacológico , Stents , Accidente Cerebrovascular/prevención & control
6.
CNS Neurosci Ther ; 27(12): 1540-1548, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34545997

RESUMEN

AIMS: Cilostazol (CLS) has shown antidepressant effect in cardiovascular patients, post-stroke depression, and animal models through its neurotrophic and antiinflammatory activities. Consequently, we aimed to investigate its safety and efficacy in patients with MDD by conducting double-blind, randomized, placebo-controlled pilot study. METHODS: 80 participants with MDD (DSM-IV criteria) and Hamilton Depression Rating Scale (HDRS) score >20 were treated with CLS 50 mg or placebo twice daily plus escitalopram (ESC) 20 mg once daily for six weeks. Patients were evaluated by HDRS scores (weeks 0, 2, 4, and 6). Serum levels of CREB1, BDNF, 5-HT, TNF-α, NF- κB, and FAM19A5 were assessed pre- and post-treatment. RESULTS: Co-administration of CLS had markedly decreased HDRS score at all-time points compared to the placebo group (p < 0.001). Early improvement, response, and remission rates after 6 weeks were significantly higher in the CLS group (90%, 90%, 80%, respectively) than in the placebo group (25%, 65%, 50% respectively) (p < 0.001). Moreover, the CLS group was superior to the placebo group in modulation of the measured neurotrophic and inflammatory biomarkers. CONCLUSION: CLS is safe and effective short-term adjunctive therapy in patients with MDD with no other comorbid conditions. Trial registration ID:NCT04069819.


Asunto(s)
Cilostazol/farmacología , Trastorno Depresivo Mayor/tratamiento farmacológico , Escitalopram/farmacología , Inhibidores de Fosfodiesterasa 3/farmacología , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Adulto , Cilostazol/administración & dosificación , Cilostazol/efectos adversos , Trastorno Depresivo Mayor/sangre , Método Doble Ciego , Quimioterapia Combinada , Escitalopram/administración & dosificación , Femenino , Humanos , Inflamación/sangre , Inflamación/tratamiento farmacológico , Masculino , Persona de Mediana Edad , Evaluación de Resultado en la Atención de Salud , Inhibidores de Fosfodiesterasa 3/administración & dosificación , Inhibidores de Fosfodiesterasa 3/efectos adversos , Proyectos Piloto , Inhibidores Selectivos de la Recaptación de Serotonina/administración & dosificación
7.
Drug Des Devel Ther ; 15: 3391-3409, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34376973

RESUMEN

Levosimendan is a distinctive inodilator combing calcium sensitization, phosphodiesterase inhibition and vasodilating properties through the opening of adenosine triphosphate-dependent potassium channels. It was first approved in Sweden in 2000 for the short-term treatment of acutely decompensated severe chronic heart failure when conventional therapy is not sufficient, and in cases where inotropic support is considered appropriate. After more than 20 years, clinical applications have considerably expanded across critical care and emergency medicine, and levosimendan is now under investigation in different cardiac settings (eg, septic shock, pulmonary hypertension) and for non-cardiac applications (eg, amyotrophic lateral sclerosis). This narrative review outlines key milestones in levosimendan history, by addressing regulatory issues, pharmacological peculiarities and clinical aspects (efficacy and safety) of a drug that did not receive great attention in the heart failure guidelines. A brief outlook to the ongoing clinical trials is also offered.


Asunto(s)
Cardiotónicos/farmacología , Insuficiencia Cardíaca/tratamiento farmacológico , Simendán/farmacología , Adenosina Trifosfato/metabolismo , Cardiotónicos/efectos adversos , Insuficiencia Cardíaca/fisiopatología , Humanos , Inhibidores de Fosfodiesterasa 3/efectos adversos , Inhibidores de Fosfodiesterasa 3/farmacología , Vasodilatadores/efectos adversos , Vasodilatadores/farmacología
8.
Cells ; 10(8)2021 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-34440764

RESUMEN

OBJECTIVE: platelets possess not only haemostatic but also inflammatory properties, which combined are thought to play a detrimental role in thromboinflammatory diseases such as acute coronary syndromes and stroke. Phosphodiesterase (PDE) 3 and -5 inhibitors have demonstrated efficacy in secondary prevention of arterial thrombosis, partially mediated by their antiplatelet action. Yet it is unclear whether such inhibitors also affect platelets' inflammatory functions. Here, we aimed to examine the effect of the PDE3A inhibitor cilostazol and the PDE5 inhibitor tadalafil on platelet function in various aspects of thromboinflammation. Approach and results: cilostazol, but not tadalafil, delayed ex vivo platelet-dependent fibrin formation under whole blood flow over type I collagen at 1000 s-1. Similar results were obtained with blood from Pde3a deficient mice, indicating that cilostazol effects are mediated via PDE3A. Interestingly, cilostazol specifically reduced the release of phosphatidylserine-positive extracellular vesicles (EVs) from human platelets while not affecting total EV release. Both cilostazol and tadalafil reduced the interaction of human platelets with inflamed endothelium under arterial flow and the release of the chemokines CCL5 and CXCL4 from platelets. Moreover, cilostazol, but not tadalafil, reduced monocyte recruitment and platelet-monocyte interaction in vitro. CONCLUSIONS: this study demonstrated yet unrecognised roles for platelet PDE3A and platelet PDE5 in platelet procoagulant and proinflammatory responses.


Asunto(s)
Antiinflamatorios/farmacología , Plaquetas/efectos de los fármacos , Cilostazol/farmacología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/metabolismo , Fibrinolíticos/farmacología , Inhibidores de Fosfodiesterasa 3/farmacología , Activación Plaquetaria/efectos de los fármacos , Animales , Coagulación Sanguínea/efectos de los fármacos , Plaquetas/enzimología , Plaquetas/inmunología , Células Cultivadas , Quimiocinas/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/genética , Fibrina/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Inhibidores de Fosfodiesterasa 5/farmacología , Adhesividad Plaquetaria/efectos de los fármacos , Transducción de Señal , Tadalafilo/farmacología
9.
Arch Physiol Biochem ; 127(3): 250-257, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31240952

RESUMEN

OBJECTIVE: Elevated intracellular cAMP concentrations potentiate insulin secretion from pancreatic ß cells. Phosphodiesterase 3B (PDE3B) is highly expressed in these cells and plays a role in the regulation of insulin secretion. MATERIALS AND METHODS: In this study, effects of amrinone, an inhibitor of PDE3B on insulin release from isolated pancreatic islets, were determined. RESULTS: Exposure of islets to amrinone for 15, 30 and 90 min markedly increased secretion induced by 6.7 mM glucose. Amrinone enhanced also secretion stimulated by 6.7 mM glucose and DB-cAMP, an activator of PKA. It was also demonstrated that amrinone potentiated insulin secretion induced by 6.7 mM glucose in the combination with PMA (activator of PKC) or acetylcholine. However, the insulin-secretory response to glucose and glibenclamide was unchanged by amrinone. CONCLUSIONS: These results indicate that amrinone is capable of increasing insulin secretion; however, its action is restricted.


Asunto(s)
Amrinona/farmacología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/metabolismo , Secreción de Insulina/efectos de los fármacos , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Inhibidores de Fosfodiesterasa 3/farmacología , Animales , Relación Dosis-Respuesta a Droga , Glucosa/farmacología , Transducción de Señal/efectos de los fármacos
10.
J Biomol Struct Dyn ; 39(7): 2302-2317, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32299297

RESUMEN

The need of circumventing life-threatening cardiovascular disorders (CVDs) and pulmonary hypertension (PHT) worldwide prompts researchers to develop effective therapeutic agents. Crucial role of cyclic nucleotide phosphodiesterase-5 (PDE5A) and cyclic nucleotide phosphodiesterase-3 (PDE3A) in cardiovascular signaling makes them potential drug targets for the treatment of CVDs and PHT. In this study, one-drug-multiple-target strategy has been employed to screen inhibitors exhibiting dual specificity through Phase-generated and statistically validated e-pharmacophore models of PDE5A and PDE3A. An extensive CoCoCo database of 7 million compounds with ∼150,000,000 conformations was virtually screened by sequential e-pharmacophore mapping followed by Lipinski Rule of Five (RO5) evaluation and hierarchical docking simulations. Finally, docked hits were subjected to rigorous MMGBSA analysis to estimate the relative spatial affinity of the drug-like compounds. The hits (354 and 366 ligands against PDE5A and PDE3A, respectively) were further optimized through 2D clustering followed by a comprehensive 2D and 3D interaction analysis. Five structurally diverse hits mapped equally well with the e-pharmacophore models and showed promising inhibitory interactions with conserved four catalytic features of PDE5A and PDE3A, thus exhibiting dual specificity. Proposed lead compounds exhibited the lowest MMGBSA binding energies and were found to be in agreement with Lipinski Rule of Five (RO5) and ADME/Tox criteria as compared to sildenafil. The proposed dual inhibitors could thus provide promising outcomes for the discovery of dual as well as multipotent drug like compounds after lead optimization and primary therapeutic interventions.


Asunto(s)
Enfermedades Cardiovasculares , Inhibidores de Fosfodiesterasa 3 , Enfermedades Cardiovasculares/tratamiento farmacológico , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3 , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5 , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Inhibidores de Fosfodiesterasa 3/farmacología
11.
J Cardiovasc Pharmacol ; 76(6): 684-691, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33002964

RESUMEN

Ischemic preconditioning and postconditioning are strong measures preserving the heart against ischemia-reperfusion injury in experimental setting but are too invasive and impractical for clinical routine. The cardioprotective effects of ischemic preconditioning and postconditioning can be imitated pharmacologically, for example, with the phosphodiesterase inhibitors sildenafil and milrinone. We hypothesize that sildenafil-induced preconditioning is concentration dependent and further that a combined treatment of "nonprotective" versus "protective" concentrations of sildenafil and milrinone leads to a significant infarct size reduction. Experiments were performed on isolated hearts of male Wistar rats, randomized into 12 groups, mounted onto a Langendorff system, and perfused with Krebs-Henseleit buffer. All hearts underwent 33 minutes ischemia and 60 minutes of reperfusion. For determination of a concentration-dependent effect of sildenafil, hearts were perfused with increasing concentrations of sildenafil (0.1-1 µM) over 10 minutes before ischemia. In a second series of experiments, hearts were treated with 0.3 µM sildenafil or 1 µM milrinone as the "protective" concentrations. A higher concentration of respective drugs did not further reduce infarct size. In addition, a combination of "protective" and "nonprotective" concentrations of sildenafil and milrinone was applied. Sildenafil and milrinone in lower concentrations led to significant infarct size reduction, whereas combining both substances in cardioprotective concentrations did not enhance this effect. Sildenafil in a concentration of 0.3 µM induces myocardial protection. Furthermore, treatment with sildenafil and milrinone in lower concentrations had an equally strong cardioprotective effect regarding infarct size reduction compared with the administration of "protective" concentrations.


Asunto(s)
Milrinona/farmacología , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/patología , Inhibidores de Fosfodiesterasa 3/farmacología , Inhibidores de Fosfodiesterasa 5/farmacología , Citrato de Sildenafil/farmacología , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada , Hemodinámica/efectos de los fármacos , Preparación de Corazón Aislado , Masculino , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Ratas Wistar , Función Ventricular Izquierda/efectos de los fármacos
12.
Int J Mol Sci ; 21(18)2020 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-32967310

RESUMEN

Phosphodiesterases (PDEs) are essential enzymes for the regulation of pathways mediated by cyclic adenosine monophosphate (cAMP). Secondary plant compounds like anthocyanins (ACs) can inhibit PDE activity and, consequently, may be beneficial for lipid metabolism. This study investigated 18 AC-rich juice extracts and pure reference compounds from red fruits for potential inhibitory effects on PDE 3B activity. Extracts were obtained through adsorption on Amberlite® XAD 7 resin. Based on this screening, the chokeberry, blueberry, pomegranate, and cranberry extracts were active, with half maximal inhibitory concentrations (IC50) ranging from 163 ± 3 µg/mL to 180 ± 3 µg/mL. The ACs in these extracts, peonidin-3-glucoside and cyanidin-3-arabinoside, were the most active single compounds (IC50 = 56 ± 20 µg/mL, 108 ± 6 µg/mL). All extracts comprised high amounts of phenolic compounds, as determined by the Folin-Ciocalteu assay, ranging from 39.8 ± 1.5 to 73.5 ± 4.8 g gallic acid equivalents (GAE)/100 g extract. Pomegranate and chokeberry extracts exhibited the largest amounts of polyphenols (72.3 ± 0.7 g GAE/100 g, 70.6 ± 4.1 g GAE/100 g, respectively). Overall, our results showed that fruit juice extracts and their ACs can inhibit PDE activity. Any potential health benefits in vivo will be investigated in the future.


Asunto(s)
Antocianinas , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/metabolismo , Jugos de Frutas y Vegetales , Frutas/química , Inhibidores de Fosfodiesterasa 3 , Extractos Vegetales/química , Antocianinas/química , Antocianinas/farmacología , Células HT29 , Humanos , Inhibidores de Fosfodiesterasa 3/química , Inhibidores de Fosfodiesterasa 3/farmacología
13.
Sci Rep ; 10(1): 9175, 2020 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-32514052

RESUMEN

Cilostazol, a phosphodiesterase 3 inhibitor, reduces the amyloid-beta (Aß) burden in mouse models of Alzheimer disease by as yet unidentified mechanisms. In the present study, we examined the possibility that cilostazol ameliorates lysosomal dysfunction. Astrocytes treated with bafilomycin A1 (BafA1) exhibited markedly reduced DND-189 and acridine orange (AO) fluorescence, indicating reduced lysosomal acidity. In both cases, BafA1-induced alkalization was reversed by addition of cilostazol, dibutyryl cAMP or forskolin. All three agents significantly increased free zinc levels in lysosomes, and addition of the zinc chelator TPEN abrogated lysosomal reacidification. These treatments did not raise free zinc levels or reverse BafA1-mediated lysosomal alkalization in metallothionein 3 (Mt3)-null astrocytes, indicating that the increases in zinc in astrocytes were derived mainly from Mt3. Lastly, in FITC-Aß-treated astrocytes, cilostazol reversed lysosomal alkalization, increased cathepsin D activity, and reduced Aß accumulation in astrocytes. Cilostazol also reduced mHtt aggregate formation in GFP-mHttQ74-expressing astrocytes. Collectively, our results present the novel finding that cAMP/PKA can overcome the v-ATPase blocking effect of BafA1 in a zinc- and Mt3-dependent manner.


Asunto(s)
Astrocitos/citología , Cilostazol/farmacología , AMP Cíclico/metabolismo , Lisosomas/metabolismo , Macrólidos/farmacología , Proteínas del Tejido Nervioso/metabolismo , Inhibidores de Fosfodiesterasa 3/farmacología , Zinc/metabolismo , Adenosina Trifosfatasas/antagonistas & inhibidores , Péptidos beta-Amiloides/metabolismo , Animales , Autofagia , Catepsina D/metabolismo , Células Cultivadas , Inhibidores Enzimáticos , Concentración de Iones de Hidrógeno/efectos de los fármacos , Metalotioneína 3 , Ratones
14.
Nat Commun ; 11(1): 3236, 2020 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-32591543

RESUMEN

The promotion of apoptosis in tumor cells is a popular strategy for developing anti-cancer drugs. Here, we demonstrate that the plant indole alkaloid natural product nauclefine induces apoptosis of diverse cancer cells via a PDE3A-SLFN12 dependent death pathway. Nauclefine binds PDE3A but does not inhibit the PDE3A's phosphodiesterase activity, thus representing a previously unknown type of PDE3A modulator that can initiate apoptosis without affecting PDE3A's canonical function. We demonstrate that PDE3A's H840, Q975, Q1001, and F1004 residues-as well as I105 in SLFN12-are essential for nauclefine-induced PDE3A-SLFN12 interaction and cell death. Extending these molecular insights, we show in vivo that nauclefine inhibits tumor xenograft growth, doing so in a PDE3A- and SLFN12-dependent manner. Thus, beyond demonstrating potent cytotoxic effects of an alkaloid natural product, our study illustrates a potentially side-effect-reducing strategy for targeting PDE3A for anti-cancer therapeutics without affecting its phosphodiesterase activity.


Asunto(s)
Alcaloides/farmacología , Apoptosis/efectos de los fármacos , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/metabolismo , Indoles/farmacología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Naftiridinas/farmacología , Alcaloides/química , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cilostazol/farmacología , Femenino , Humanos , Indoles/química , Ratones Desnudos , Naftiridinas/química , Inhibidores de Fosfodiesterasa 3/farmacología , Estabilidad Proteica/efectos de los fármacos , Tetrahidroisoquinolinas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Ann Vasc Surg ; 69: 391-399, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32599107

RESUMEN

BACKGROUND: Phosphodiesterase enzymes play a pivotal role in the pathogenesis of ischemia/reperfusion (IR). We examined the role of milrinone (MIL), a phosphodiesterase 3 inhibitor, on remote injury of the heart and lung after abdominal aortic cross-clamping. DESIGN: Experimental study. METHODS: Twenty-one Wistar rats were divided into 3 groups: (1) control (C, n = 7), underwent laparotomy and exploration of abdominal aorta only; (2) IR (n = 7), normal saline was applied intraperitoneally (i.p) before IR induced by clamping of the abdominal aorta for 1 hr and then allowing reperfusion for 1 hr; and (3) MIL + IR (n = 7), MIL was given (0.5 mg/kg, i.p) before IR. After sacrification, the lungs and hearts were taken out for analyses and the tissue malondialdehyde (MDA) and glutathione (GSH) were studied. All tissues were examined under light microscopy and transmission electron microscopy (TEM). Expressions of caveolin (Cav)-1 in the lung and Cav-1 and Cav-3 in the heart were examined immunohistochemically. RESULTS: The MIL + IR group had significantly a lower magnitude of oxidative stress than the IR group both in the lung and heart (lung: P = 0.03 for MDA and 0.001 for GSH and heart: P = 0.002 for MDA and 0.000 for GSH). In light microscopy, the MIL + IR group had statistically a lower total injury score than the IR group for both the lung and heart tissue (P = 0.03 and P = 0.04, respectively). In TEM, regression of mitochondrial degeneration and lamellar bodies in type II pneumocytes in the lungs and obvious improvements in disruption at the intercalated discs and mitochondrial degeneration in the hearts in the MIL + IR group were detected compared with the IR group. The expression of both Cav-1 and Cav-3 in the MIL + IR group was improved compared with the IR group (P = 0.03 for both). CONCLUSIONS: MIL attenuates remote injury of heart and lung in lower body IR by inhibiting oxidative stress. Moreover, Cav-1 and Cav-3 might have a potential role in MIL-induced cardioprotection.


Asunto(s)
Aorta Abdominal/cirugía , Corazón/efectos de los fármacos , Lesión Pulmonar/prevención & control , Pulmón/efectos de los fármacos , Milrinona/farmacología , Daño por Reperfusión Miocárdica/prevención & control , Inhibidores de Fosfodiesterasa 3/farmacología , Animales , Antioxidantes/farmacología , Caveolina 1/metabolismo , Caveolina 3/metabolismo , Constricción , Modelos Animales de Enfermedad , Pulmón/metabolismo , Pulmón/ultraestructura , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Daño por Reperfusión Miocárdica/etiología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocardio/metabolismo , Miocardio/ultraestructura , Estrés Oxidativo/efectos de los fármacos , Ratas Wistar , Transducción de Señal
16.
Naunyn Schmiedebergs Arch Pharmacol ; 393(10): 1859-1870, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32424476

RESUMEN

Acute pancreatitis (AP) is an inflammatory disorder with a high mortality rate. Cilostazol is a selective phosphodiesterase-3 inhibitor drug that is commonly used as an antiplatelet, antithrombotic, and vasodilator drug. It exhibits antioxidant, anti-inflammatory, and anti-apoptotic activities, but its effect on AP has not been fully elucidated yet. The present study aimed to investigate the effects of cilostazol on L-arginine-induced AP and the possible protective mechanisms. A rat model of AP was established by a single i.p. injection of 3-g/kg L-arginine on day 13 of the experiment. The treated groups received a single daily oral dose of either 100 or 300 mg/kg/day for 14 consecutive days. Rats with AP showed histopathological changes of pancreatic tissue injury together with increased serum amylase enzyme activity and decreased serum insulin, pancreatic adiponectin, and cGMP levels. Moreover, AP rats showed increased pancreatic inflammatory biomarker (TNF-α, VCAM-1, and MPO) levels with decreased anti-inflammatory IL-10 levels. In addition, oxidative stress biomarkers (MDA and NO) were increased in AP with decreased antioxidant SOD activity and GSH level. Moreover, HO-1 immunostaining was increased in the AP group. Cilostazol pretreatment reversed the histopathological change; decreased the amylase activity and the levels of TNF-α, VCAM-1, and MPO; and increased the levels of insulin, adiponectin, cGMP, cAMP, and IL-10. Moreover, cilostazol decreased MDA and NO but increased SOD and GSH. Lastly, cilostazol increased the HO-1 immunostaining more than in the AP group. These data suggest that cilostazol protects against L-arginine-induced AP, which may be related to an increase in cGMP, cAMP, and upregulation of HO-1 with subsequent anti-inflammatory and antioxidant properties.


Asunto(s)
Arginina/toxicidad , Cilostazol/uso terapéutico , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Hemo Oxigenasa (Desciclizante)/metabolismo , Pancreatitis/metabolismo , Animales , Cilostazol/farmacología , Masculino , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Pancreatitis/inducido químicamente , Pancreatitis/prevención & control , Inhibidores de Fosfodiesterasa 3/farmacología , Inhibidores de Fosfodiesterasa 3/uso terapéutico , Ratas , Ratas Wistar
17.
Int J Mol Sci ; 21(9)2020 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-32403267

RESUMEN

This study aimed to investigate whether a selective phosphodiesterase-3 (PDE3) inhibitor olprinone can positively influence the inflammation, apoptosis, and respiratory parameters in animals with acute respiratory distress syndrome (ARDS) model induced by repetitive saline lung lavage. Adult rabbits were divided into 3 groups: ARDS without therapy (ARDS), ARDS treated with olprinone i.v. (1 mg/kg; ARDS/PDE3), and healthy ventilated controls (Control), and were oxygen-ventilated for the following 4 h. Dynamic lung-thorax compliance (Cdyn), mean airway pressure (MAP), arterial oxygen saturation (SaO2), alveolar-arterial gradient (AAG), ratio between partial pressure of oxygen in arterial blood to a fraction of inspired oxygen (PaO2/FiO2), oxygenation index (OI), and ventilation efficiency index (VEI) were evaluated every hour. Post mortem, inflammatory and oxidative markers (interleukin (IL)-6, IL-1ß, a receptor for advanced glycation end products (RAGE), IL-10, total antioxidant capacity (TAC), 3-nitrotyrosine (3NT), and malondialdehyde (MDA) and apoptosis (apoptotic index and caspase-3) were assessed in the lung tissue. Treatment with olprinone reduced the release of inflammatory mediators and markers of oxidative damage decreased apoptosis of epithelial cells and improved respiratory parameters. The results indicate a future potential of PDE3 inhibitors also in the therapy of ARDS.


Asunto(s)
Apoptosis/efectos de los fármacos , Modelos Animales de Enfermedad , Imidazoles/farmacología , Inflamación/prevención & control , Inhibidores de Fosfodiesterasa 3/farmacología , Piridonas/farmacología , Síndrome de Dificultad Respiratoria/prevención & control , Animales , Biomarcadores/metabolismo , Citocinas/metabolismo , Inflamación/metabolismo , Inflamación/fisiopatología , Mediadores de Inflamación/metabolismo , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/fisiopatología , Conejos , Síndrome de Dificultad Respiratoria/metabolismo , Síndrome de Dificultad Respiratoria/fisiopatología
18.
Molecules ; 25(8)2020 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-32326556

RESUMEN

A simple, quick, easy and cheap tandem mass spectrometry (MS/MS) method for the determination of adenosine monophosphate (AMP) and cyclic adenosine monophosphate (cAMP) has been newly developed. This novel MS/MS method was applied for the evaluation of the inhibitory effect of a novel 2-oxo-1,2-dihydropyridine-3-carbonitrile derivative, also named DF492, on PDE3 enzyme activity in comparison to its parent drug milrinone. Molecule DF492, with an IC50 of 409.5 nM, showed an inhibition of PDE3 greater than milrinone (IC50 = 703.1 nM). To explain the inhibitory potential of DF492, molecular docking studies toward the human PDE3A were carried out with the aim of predicting the binding mode of DF492. The presence of different bulkier decorating fragments in DF492 was pursued to shift affinity of this novel molecule toward PDE3A compared to milrinone in accordance with both the theoretical and experimental results. The described mass spectrometric approach could have a wider potential use in kinetic and biomedical studies and could be applied for the determination of other phosphodiesterase inhibitor molecules.


Asunto(s)
Adenosina Monofosfato/química , AMP Cíclico/química , Espectrometría de Masas , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Inhibidores de Fosfodiesterasa 3/química , Adenosina Monofosfato/farmacología , Sitios de Unión , AMP Cíclico/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Enlace de Hidrógeno , Milrinona/farmacología , Estructura Molecular , Inhibidores de Fosfodiesterasa 3/farmacología , Unión Proteica , Relación Estructura-Actividad , Espectrometría de Masas en Tándem
19.
Am J Physiol Lung Cell Mol Physiol ; 318(5): L908-L920, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32159371

RESUMEN

Over 2,000 mutations have been reported in the cystic fibrosis transmembrane conductance regulator (cftr) gene, many of which cause disease but are rare and have no effective treatment. Thus, there is an unmet need for new, mutation-agnostic therapies for cystic fibrosis (CF). Phosphodiesterase (PDE) inhibitors are one such class of therapeutics that have been shown to elevate intracellular cAMP levels and stimulate CFTR-dependent anion secretion in human airway epithelia; however, the number of people with CF that could be helped by PDE inhibitors remains to be determined. Here we used Fisher rat thyroid (FRT) cells stably transduced with rare human CFTR mutants and studied their responsiveness to the dual phosphodiesterase 3/4 inhibitor RPL554 (Verona Pharma). Through its inhibitory effect on PDE4D, we find that RPL554 can elevate intracellular cAMP leading to a potentiation of forskolin-stimulated current mediated by R334W, T338I, G551D, and S549R mutants of CFTR when used alone or in combination with CFTR modulators. We also were able to reproduce these effects of RPL554 on G551D-CFTR when it was expressed in primary human bronchial epithelial cells, indicating that RPL554 would have stimulatory effects on rare CFTR mutants in human airways and validating FRT cells as a model for PDE inhibitor studies. Furthermore, we provide biochemical evidence that VX-809 causes surprisingly robust correction of several class III and IV CFTR mutants. Together, our findings further support the therapeutic potential of RPL554 for patients with CF with class III/IV mutations and emphasize the potential of PDEs as potential drug targets that could benefit patients with CF.


Asunto(s)
AMP Cíclico/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Isoquinolinas/farmacología , Inhibidores de Fosfodiesterasa 3/farmacología , Inhibidores de Fosfodiesterasa 4/farmacología , Pirimidinonas/farmacología , Células Epiteliales Tiroideas/efectos de los fármacos , Aminopiridinas/farmacología , Animales , Benzodioxoles/farmacología , Bronquios/citología , Bronquios/efectos de los fármacos , Bronquios/metabolismo , Línea Celular , Colforsina/farmacología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/agonistas , Regulador de Conductancia de Transmembrana de Fibrosis Quística/clasificación , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Humanos , Mutación , Cultivo Primario de Células , Ratas , Ratas Endogámicas F344 , Células Epiteliales Tiroideas/citología , Células Epiteliales Tiroideas/metabolismo , Transgenes
20.
Life Sci ; 241: 117100, 2020 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-31783052

RESUMEN

AIMS: The present study aims to define maturation, yield, health, and ease of collection of murine immature oocytes recovered using the conventional method or from mice treated with cilostazol. MAIN METHODS: The conventional method included the superovulation of mice and the recovery of germinal vesicle (GV) or metaphase (MI) oocytes from preovulatory follicles. The cilostazol method included the oral treatment of superovulated mice with 7.5 mg cilostazol once or twice to result in the ovulation of MI or GV oocytes, respectively. KEY FINDINGS: The cilostazol method resulted in >95% of GV or MI oocytes with a diameter range of 60-90 µm or 50.1-70 µm in comparison to <60.0% of GV or MI oocytes resulting from the conventional method, respectively (P < 0.0001). The cilostazol method resulted in GV oocytes having higher levels of co-occurrence of peripheral cortical granules (CG) and chromatin configuration of surrounded nucleolus and MI oocytes having higher levels of co-occurrence of normally organized spindles/chromosomes and peripheral CG with free domains than did the conventional method (P < 0.001). The cilostazol method was more time and labor efficient and resulted in higher oocyte yields of normal morphology than did the conventional method (P < 0.01). SIGNIFICANCE: The presented method provides not only oocytes with uniform size and synchronized developmental maturation but also a technique of oocyte collection that is efficient and resourceful. It is possible that not all immature oocytes resulting from the conventional method are from preovulatory follicles nor have been developed adequately and consequently ovulated as opposed to the presented method.


Asunto(s)
Cilostazol/farmacología , Recuperación del Oocito/métodos , Oocitos/citología , Oocitos/fisiología , Inhibidores de Fosfodiesterasa 3/farmacología , Animales , Nucléolo Celular , Núcleo Celular , Cromatina/ultraestructura , Femenino , Técnicas de Maduración In Vitro de los Oocitos/métodos , Metafase , Ratones , Ovulación/efectos de los fármacos , Superovulación/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...