Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Int J Mol Sci ; 24(22)2023 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-38003456

RESUMEN

Radiation-induced lung fibrosis (RILF) is a common complication of radiotherapy in lung cancer. However, to date no effective treatment has been developed for this condition. NXC736 is a novel small-molecule compound that inhibits NLRP3, but its effect on RILF is unknown. NLRP3 activation is an important trigger for the development of RILF. Thus, we aimed to evaluate the therapeutic effect of NXC736 on lung fibrosis inhibition using a RILF animal model and to elucidate its molecular signaling pathway. The left lungs of mice were irradiated with a single dose of 75 Gy. We observed that NXC736 treatment inhibited collagen deposition and inflammatory cell infiltration in irradiated mouse lung tissues. The damaged lung volume, evaluated by magnetic resonance imaging, was lower in NXC736-treated mice than in irradiated mice. NXC736-treated mice exhibited significant changes in lung function parameters. NXC736 inhibited inflammasome activation by interfering with the NLRP3-ASC-cleaved caspase-1 interaction, thereby reducing the expression of IL-1ß and blocking the fibrotic pathway. In addition, NXC736 treatment reduced the expression of epithelial-mesenchymal transition markers such as α-SMA, vimentin, and twist by blocking the Smad 2,3,4 signaling pathway. These data suggested that NXC736 is a potent therapeutic agent against RILF.


Asunto(s)
Fibrosis Pulmonar , Traumatismos por Radiación , Ratones , Animales , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/etiología , Fibrosis Pulmonar/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Pulmón/patología , Fibrosis , Inflamasomas/metabolismo , Traumatismos por Radiación/metabolismo , Transducción de Señal , Síndrome de Fibrosis por Radiación
2.
Cell Mol Gastroenterol Hepatol ; 13(3): 925-947, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34890841

RESUMEN

BACKGROUND & AIMS: Sphingosine 1-phosphate receptors (S1PRs) are a group of G-protein-coupled receptors that confer a broad range of functional effects in chronic inflammatory and metabolic diseases. S1PRs also may mediate the development of nonalcoholic steatohepatitis (NASH), but the specific subtypes involved and the mechanism of action are unclear. METHODS: We investigated which type of S1PR isoforms is activated in various murine models of NASH. The mechanism of action of S1PR4 was examined in hepatic macrophages isolated from high-fat, high-cholesterol diet (HFHCD)-fed mice. We developed a selective S1PR4 functional antagonist by screening the fingolimod (2-amino-2-[2-(4- n -octylphenyl)ethyl]-1,3- propanediol hydrochloride)-like sphingolipid-focused library. RESULTS: The livers of various mouse models of NASH as well as hepatic macrophages showed high expression of S1pr4. Moreover, in a cohort of NASH patients, expression of S1PR4 was 6-fold higher than those of healthy controls. S1pr4+/- mice were protected from HFHCD-induced NASH and hepatic fibrosis without changes in steatosis. S1pr4 depletion in hepatic macrophages inhibited lipopolysaccharide-mediated Ca++ release and deactivated the Nod-like receptor pyrin domain-containning protein 3 (NLRP3) inflammasome. S1P increased the expression of S1pr4 in hepatic macrophages and activated NLRP3 inflammasome through inositol trisphosphate/inositol trisphosphate-receptor-dependent [Ca++] signaling. To further clarify the biological function of S1PR4, we developed SLB736, a novel selective functional antagonist of SIPR4. Similar to S1pr4+/- mice, administration of SLB736 to HFHCD-fed mice prevented the development of NASH and hepatic fibrosis, but not steatosis, by deactivating the NLRP3 inflammasome. CONCLUSIONS: S1PR4 may be a new therapeutic target for NASH that mediates the activation of NLRP3 inflammasome in hepatic macrophages.


Asunto(s)
Inflamasomas , Enfermedad del Hígado Graso no Alcohólico , Animales , Humanos , Inflamasomas/metabolismo , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Receptores de Esfingosina-1-Fosfato
3.
Am J Physiol Gastrointest Liver Physiol ; 299(5): G1147-53, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20724528

RESUMEN

Aging changes in the stomach lead to a decreased capacity for tissue repair in response to gastric acid. The aim of this study was to determine the mechanism associated with the increased susceptibility to injury of aging mucosa including reactive oxygen species (5), apoptosis, angiogenesis, and sensory neuron activity. Fischer 344 rats at four different ages (6, 31, 74 wk, and 2 yr of age) were studied. The connective tissue indicators [salt-soluble collagen and sulfated glycosaminoglycan (sGAG)], lipid hydroperoxide (LPO), myeloperoxidase (MPO), and hexosamine were assessed. We also evaluated the expression of early growth response-1 (Egr-1), phosphatase and tension homologue deleted on chromosome 10 (PTEN), caspase-9 (index of apoptosis), VEGF (index of angiogenesis), calcitonin gene-related peptide (CGRP, index of sensory neurons), and neuronal nitric oxide synthase (nNOS). The histological connective tissue area in the lower part of rat gastric mucosa increased with aging, with increase of salt-soluble collagen and sGAG. LPO and MPO in old rats were significantly greater than in the young rats, whereas hexosamine was significantly reduced. The old gastric mucosa had increased expression of Egr-1, PTEN, and caspase-9, whereas the VEGF, CGRP, and nNOS expression were significantly reduced. These results indicate that the lower part of rat gastric mucosa was found to be replaced by connective tissue with accumulation of oxidative products with aging. In addition, impairment of apoptosis, angiogenesis, and sensory neuron activity via the activation of Egr-1 and PTEN might increase the susceptibility of gastric mucosa to injury during aging.


Asunto(s)
Envejecimiento/fisiología , Apoptosis/fisiología , Mucosa Gástrica/fisiología , Neovascularización Fisiológica/fisiología , Especies Reactivas de Oxígeno/metabolismo , Células Receptoras Sensoriales/fisiología , Animales , Western Blotting , Tejido Conectivo/metabolismo , Masculino , Ratas , Ratas Endogámicas F344 , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estadísticas no Paramétricas
4.
J Pharm Pharm Sci ; 13(1): 93-106, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20456834

RESUMEN

PURPOSE: This study was performed to find which types of hepatic CYP isoforms are responsible for the metabolism of mirodenafil (a new erectogenic) and one of its metabolite, SK3541, using various hepatic CYP inducers and inhibitors in rats. METHODS: Mirodenafil at a dose of 20 mg/kg was administered intravenously to control rats and rats pretreated with various CYP inducers and inhibitors. The disappearance of SK3541 was also measured in vitro hepatic microsomes of rats with and without CYP inducer and inhibitors. RESULTS: Compared to controls, in rats pretreated with 3-methylcholanthrene, orphenadrine, and dexamethasone (main inducers of CYP1A1/2, 2B1/2, and 3A1/2, respectively), the non-renal clearances (CLNRs) of mirodenafil were significantly faster (by 39.4%, 59.3%, and 63.9%, respectively). However, compared to controls, in rats pretreated with quinine and troleandomycin (main inhibitors of CYP2D subfamily and 3A1/2, respectively), the CLNRs of mirodenafil were significantly slower (by 36.1% and 33.2%, respectively). In rat hepatic microsomes spiked with furafylline, quinine, and troleandomycin (main inhibitors of CYP1A2, 2D subfamily, and 3A1/2, respectively), the intrinsic clearances (CLints) for the disappearance of SK3541 were significantly slower (by 18.4%, 35.3%, and 51.5%, respectively) than controls. Also in rat hepatic microsomes pretreated with orphenadrine (a main inducer of CYP2B1/2), the CLint for the disappearance of SK3541 was significantly faster (by 55.5%) than controls. CONCLUSIONS: The above data suggest that hepatic CYP1A1/2, 2B1/2, 2D subfamily, and 3A1/2 are involved in the metabolism of both mirodenafil and SK3541 in rats.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Pirimidinonas/metabolismo , Sulfonamidas/metabolismo , Animales , Inducción Enzimática , Inhibidores Enzimáticos/farmacología , Masculino , Microsomas Hepáticos/metabolismo , Inhibidores de Fosfodiesterasa 5 , Ratas , Ratas Sprague-Dawley
5.
Biosci Biotechnol Biochem ; 74(10): 2022-8, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20944425

RESUMEN

The parenteral route has many merits over the oral route, including greater predictability, reproducibility of absorption, and rapid drug action, but injectable phytomedicines are uncommon due to protein precipitating tannin and hemolytic saponin components. In this study, in an effort to develop a safe injectable analgesic phytomedicine, we prepared a tannin and saponin-free Lonicera japonica extract, SKLJI, through fractionation and column purification, and evaluated its anti-inflammatory and analgesic activities in in vivo experimental models of inflammation and pain. The removal of tannin and saponin resulted in loganin and sweroside-enriched SKLJI and it showed reduced hemolysis and protein precipitation. In efficacy tests, SKLJI inhibited croton oil- and arachidonic acid-induced ear edema, acetic acid-induced writhing, and carrageenan-induced rat hind paw hyperalgesia. Inhibition of cylcooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), and 5-lipoxyfenase (5-LO) activities by SKLJI appeared to be the mechanism underlying anti-inflammatory and analgesic efficacy. Loganin and sweroside also showed anti-inflammatory and analgesic activities, suggesting that they might be active principles in the efficacy of SKLJI. These results suggest that SKLJI is a viable candidate for a new anti-inflammatory and analgesic phytomedicine that can be administered by the parenteral route.


Asunto(s)
Analgésicos/aislamiento & purificación , Analgésicos/farmacología , Antiinflamatorios/aislamiento & purificación , Antiinflamatorios/farmacología , Lonicera/química , Extractos Vegetales/aislamiento & purificación , Extractos Vegetales/farmacología , Analgésicos/administración & dosificación , Analgésicos/uso terapéutico , Animales , Antiinflamatorios/administración & dosificación , Antiinflamatorios/uso terapéutico , Araquidonato 5-Lipooxigenasa/metabolismo , Ciclooxigenasa 2/metabolismo , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/aislamiento & purificación , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Hemólisis/efectos de los fármacos , Inflamación/tratamiento farmacológico , Inflamación/enzimología , Inflamación/inmunología , Inyecciones Intravenosas , Glucósidos Iridoides/farmacología , Glucósidos Iridoides/uso terapéutico , Iridoides/farmacología , Iridoides/uso terapéutico , Masculino , Ratones , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Extractos Vegetales/administración & dosificación , Extractos Vegetales/uso terapéutico , Ratas , Ratas Sprague-Dawley
6.
J Korean Med Sci ; 25(6): 875-81, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20514308

RESUMEN

Cochinchina momordica seed is the dried ripe seed of Momordica cochinchinensis, a perennial vine. The antiulcer effect of an extract from cochinchina momordica seeds (SK-MS10) was evaluated in a rat model of acetic acid-induced gastric ulcers. Gastric ulcers were produced by subserosal injection of acetic acid. SK-MS10 (200 mg/kg) or vehicle was administered orally once per day for 14 days after the acetic acid injection. The stomach was removed and the ulcer size measured at day 7 and 14 of the treatment. Expression of vascular endothelial growth factor (VEGF) was assessed by real-time RT-PCR and Western blot analysis. In addition, the microvasculature density (MVD) adjacent to the ulcer margin was examined by immunohistochemistry. The treatment with SK-MS10 for 7 and 14 days significantly accelerated ulcer healing and increased the expression of mRNA (at day 7) as well as VEGF protein (at day 14) compared to the vehicle-treated rats. The MVD for factor VIII was also higher in the SK-MS10 treatment group compared to the vehicle-treated rats; however, these differences were not statistically significant. These results suggest that SK-MS10 treatment accelerates the healing of gastric ulcers via upregulation of VEGF and angiogenesis in an acetic acid rat model.


Asunto(s)
Antiulcerosos/uso terapéutico , Momordica/química , Neovascularización Fisiológica/efectos de los fármacos , Extractos Vegetales/uso terapéutico , Úlcera Gástrica/tratamiento farmacológico , Ácido Acético/toxicidad , Administración Oral , Animales , Factor VIII/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Semillas/química , Úlcera Gástrica/inducido químicamente , Úlcera Gástrica/patología , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
7.
Dig Dis Sci ; 54(12): 2549-60, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19130224

RESUMEN

Cochinchina momordica seed extract (SKMS10), which is composed of the major compounds momordica saponins, has been evaluated for its gastroprotective effects in rat models of acute gastric mucosal damage. Ethanol and water immersion restraint stress (WRS) induced gastric damage, including hemorrhages and edema, was significantly attenuated by pretreatment with SK-MS10. In addition, SK-MS10 reduced increases of mucosal myeloperoxidase (MPO), IL-1ß, and TNFα levels and the expression of cPLA(2), and 5-LOX induced by ethanol or WRS. SK-MS10 also increased hexosamine, adherent mucus, and the expression of MUC5AC. Furthermore, SK-MS10 enhanced the mucosal expression of the CGRP gene and its serum levels.N(G)-methyl L-arginine (L-NMMA) or capsaicin desensitization reversed the SK-MS10-induced gastroprotection effect. These results suggest that SK-MS10 is a gastroprotective agent against acute gastric mucosal damage by suppressing proinflammatory cytokines, downregulating cPLA(2), 5-LOX, and increasing the synthesis of mucus. Furthermore, CGRP-NO pathway was found to play an important role in these gastroprotective effects of SK-MS10.


Asunto(s)
Araquidonato 5-Lipooxigenasa/metabolismo , Péptido Relacionado con Gen de Calcitonina/metabolismo , Mucosa Gástrica/efectos de los fármacos , Fármacos Gastrointestinales/farmacología , Fosfolipasas A2 Grupo IV/metabolismo , Momordica , Extractos Vegetales/farmacología , Transducción de Señal/efectos de los fármacos , Úlcera Gástrica/prevención & control , Animales , Péptido Relacionado con Gen de Calcitonina/sangre , Péptido Relacionado con Gen de Calcitonina/genética , Ciclooxigenasa 2/metabolismo , Citoprotección , Dinoprostona/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo , Etanol , Mucosa Gástrica/enzimología , Mucosa Gástrica/patología , Hexosaminas/metabolismo , Mediadores de Inflamación/metabolismo , Interleucina-1beta/metabolismo , Leucotrieno B4/metabolismo , Masculino , Mucina 5AC/metabolismo , Mucina 6/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo I , Peroxidasa/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Restricción Física , Semillas , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/metabolismo , Úlcera Gástrica/enzimología , Úlcera Gástrica/etiología , Úlcera Gástrica/patología , Factores de Tiempo , Factor de Necrosis Tumoral alfa/metabolismo
8.
J Pharm Biomed Anal ; 49(2): 513-8, 2009 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-19095395

RESUMEN

The purpose of the present study was to determine sildenafil and a novel PDE-5 inhibitor, mirodenafil in the plasma and corpus cavernosum tissue of rats to compare their pharmacokinetic properties. The concentrations of mirodenafil and sildenafil in the rat plasma and corpus cavernosum tissue samples were analyzed using LC-MS/MS after a single oral administration at a dose of 40mg/kg to rats. Although the T(max), Tlambda(1/2) and MRT were not different between mirodenafil and sildenafil, the C(max) and AUC of mirodenafil were significantly higher than those of sildenafil in the plasma and corpus cavernosum tissue. Consequently mirodenafil remained longer than sildenafil in the plasma and tissue. This may provide pharmacokinetic evidence for assessment of the in vivo efficacy of mirodenafil and sildenafil.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/sangre , Pene/irrigación sanguínea , Inhibidores de Fosfodiesterasa/sangre , Piperazinas/sangre , Pirimidinonas/sangre , Sulfonamidas/sangre , Sulfonas/sangre , Administración Oral , Animales , Área Bajo la Curva , Cromatografía Liquida/métodos , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/farmacocinética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/farmacología , Ayuno , Semivida , Concentración de Iones de Hidrógeno , Masculino , Espectrometría de Masas/métodos , Tasa de Depuración Metabólica , Estructura Molecular , Peso Molecular , Pene/efectos de los fármacos , Inhibidores de Fosfodiesterasa/farmacocinética , Inhibidores de Fosfodiesterasa/farmacología , Piperazinas/administración & dosificación , Piperazinas/química , Piperazinas/farmacocinética , Piperazinas/farmacología , Polietilenglicoles/química , Purinas/administración & dosificación , Purinas/sangre , Purinas/química , Purinas/farmacocinética , Purinas/farmacología , Pirimidinonas/administración & dosificación , Pirimidinonas/química , Pirimidinonas/farmacocinética , Pirimidinonas/farmacología , Control de Calidad , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Estándares de Referencia , Reproducibilidad de los Resultados , Citrato de Sildenafil , Soluciones/química , Organismos Libres de Patógenos Específicos , Sulfonamidas/administración & dosificación , Sulfonamidas/química , Sulfonamidas/farmacocinética , Sulfonamidas/farmacología , Sulfonas/administración & dosificación , Sulfonas/química , Sulfonas/farmacocinética , Sulfonas/farmacología
9.
J Pharm Biomed Anal ; 48(5): 1425-9, 2008 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-18947958

RESUMEN

A simple, rapid, and sensitive liquid chromatography-tandem mass spectrometry (LC-MS/MS) method was applied to pharmacokinetic study of a neuroactive oleanolic-glycoside saponin, hederacolchiside E from SK-PC-B70M, a standardized extract of Pulsatilla koreana in rat. Rat plasma samples were pretreated by protein precipitation with acetonitrile, eluted from C(18) column, and analyzed using electrospray ionization (ESI)-MS/MS in negative ion mode. Digoxin was used as an internal standard. The standard curves were linear (r>0.997) over the concentration ranges of 2-500 ng/mL. The intra- and inter-day precisions were measured to be below 9% and accuracy between 90 and 111% for all quality control samples at 2, 20, 100, and 500 ng/mL (n=5). The lower limits of quantification (LLOQ) for hederacolchiside E was 2 ng/mL and the limit of detection (LOD) 0.5 ng/mL using 20 microL of plasma sample. Subsequently, hederacolchiside E was determined in rat plasma samples after oral administration of SK-PC-B70M. The mean maximum plasma concentrations of hederacolchiside E were 0.07, 0.13, and 0.36 microg/mL and the mean areas under the plasma concentration versus time curve 0.56, 1.27, and 6.46 microg h/mL at doses of 100, 200, and 400 mg/kg, respectively, which indicated non-linear pharmacokinetic pattern. In conclusion, this method was successfully applied to the pharmacokinetic study of hederacolchiside E after an oral administration of SK-PC-B70M to rats.


Asunto(s)
Medicamentos Herbarios Chinos/farmacocinética , Pulsatilla/química , Saponinas/sangre , Espectrometría de Masas en Tándem/métodos , Administración Oral , Animales , Área Bajo la Curva , Calibración , Cromatografía Liquida/métodos , Semivida , Masculino , Estructura Molecular , Extractos Vegetales/administración & dosificación , Extractos Vegetales/química , Extractos Vegetales/aislamiento & purificación , Extractos Vegetales/normas , Control de Calidad , Ratas , Ratas Sprague-Dawley , Estándares de Referencia , Reproducibilidad de los Resultados , Saponinas/administración & dosificación , Saponinas/química , Saponinas/aislamiento & purificación , Saponinas/normas , Sensibilidad y Especificidad , Factores de Tiempo
10.
PLoS One ; 10(9): e0138823, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26401847

RESUMEN

Antiangiogenic agents have been widely investigated in combination with standard chemotherapy or targeted cancer agents for better management of advanced cancers. Therapeutic agents that concurrently inhibit epidermal growth factor receptor and other angiokinases could be useful alternatives to combination therapies for epidermal growth factor receptor-dependent cancers. Here, we report the synthesis of an indole derivative of pazopanib using a bioisosteric replacement strategy, which was designated MKP101. MKP101 inhibited not only the epidermal growth factor receptor with an IC50 value of 43 nM but also inhibited angiokinases as potently as pazopanib. In addition, MKP101 effectively inhibited vascular endothelial growth factor-induced endothelial proliferation, tube formation, migration of human umbilical vein endothelial cells and proliferation of HCC827, an epidermal growth factor receptor-addicted cancer cell line. A docking model of MKP101 and the kinase domain of the epidermal growth factor receptor was generated to predict its binding mode, and validated by synthesizing and evaluating MKP101 derivatives. Additionally, a study of structure-activity relationships of indolylamino or indolyloxy pyrimidine analogues derived from MKP101 demonstrated that selectivity for epidermal growth factor receptor and other angiokinases, especially vascular endothelial growth factor receptor 2 depends on the position of substituents on pyrimidine and the type of link between pyrimidine and the indole moiety. We believe that this study could provide a basis for developing angiokinase inhibitors having high affinity for the epidermal growth factor receptor, from the pyrimidine scaffold.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Receptores ErbB/antagonistas & inhibidores , Indoles/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/metabolismo , Pirimidinas/farmacología , Inhibidores de la Angiogénesis/síntesis química , Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/patología , Proliferación Celular/efectos de los fármacos , Receptores ErbB/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Indazoles , Indoles/síntesis química , Indoles/química , Indoles/uso terapéutico , Concentración 50 Inhibidora , Neoplasias Pulmonares/patología , Simulación del Acoplamiento Molecular , Neovascularización Patológica/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirimidinas/síntesis química , Pirimidinas/química , Pirimidinas/uso terapéutico , Relación Estructura-Actividad , Sulfonamidas/química , Sulfonamidas/farmacología , Factor A de Crecimiento Endotelial Vascular/farmacología
11.
J Pharm Pharmacol ; 67(11): 1519-27, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26255780

RESUMEN

OBJECTIVES: This study was conducted to examine the tissue distribution of human recombinant epidermal growth factor (EGF) after multiple intravenous and subcutaneous injections in mice. METHODS: Male BALB/c mice were divided into (1) EGF 1 mg/kg intravenous dose, (2) EGF 5 mg/kg intravenous dose, (3) drug-free intravenous control, (4) EGF 1 mg/kg subcutaneous dose, (5) EGF 5 mg/kg subcutaneous dose and (6) drug-free subcutaneous control groups. EGF and drug-free dosing solutions were injected by intravenous and subcutaneous injections once a day for 3 days. EGF concentrations in serum and tissues of kidney, liver, lung, small intestine and tongue were determined by ELISA. KEY FINDINGS: As the intravenous and subcutaneous doses were increased from 1 to 5 mg/kg, serum Cmax and area under the concentration-time curve (AUC) values were increased dose-proportionally. In lung, tongue and small intestine, increases in AUC were dose-proportional after intravenous injections, but greater than dose-proportional after subcutaneous injections. The fold-increases in Cmax and AUC values were lowest in liver and highest in kidney. CONCLUSION: Based on Cmax and AUC data, the systemic exposure achieved by subcutaneous injections was comparable with that achieved by intravenous injections.


Asunto(s)
Factor de Crecimiento Epidérmico/farmacocinética , Proteínas Recombinantes/farmacocinética , Animales , Área Bajo la Curva , Relación Dosis-Respuesta a Droga , Factor de Crecimiento Epidérmico/administración & dosificación , Humanos , Inyecciones Intravenosas , Inyecciones Subcutáneas , Masculino , Ratones , Ratones Endogámicos BALB C , Proteínas Recombinantes/administración & dosificación , Distribución Tisular
12.
Exp Mol Med ; 44(5): 311-8, 2012 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-22282402

RESUMEN

In this study, the synergistic effect of 6-[4-(1-cyclohexyl- 1H-tetrazol-5-yl) butoxy]-3,4-dihydro-2(1H )-quinolinone (cilostazol) and Ginkgo biloba extract (GbE) was examined in apolipoprotein E (ApoE) null mice. Co-treatment with GbE and cilostazol synergistically decreased reactive oxygen species (ROS) production in ApoE null mice fed a high-fat diet. Co-treatment resulted in a significantly decreased atherosclerotic lesion area compared to untreated ApoE mice. The inflammatory cytokines and adhesion molecules such as monocyte chemoattractant-1 (MCP-1), soluble vascular cell adhesion molecule-1 (sVCAM-1), and VCAM-1 which can initiate atherosclerosis were significantly reduced by the co-treatment of cilostazol with GbE. Further, the infiltration of macrophages into the intima was decreased by co-treatment. These results suggest that co-treatment of GbE with cilostazol has a more potent anti-atherosclerotic effect than treatment with cilostazol alone in hyperlipidemic ApoE null mice and could be a valuable therapeutic strategy for the treatment of atherosclerosis.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Ginkgo biloba/química , Extractos Vegetales/administración & dosificación , Especies Reactivas de Oxígeno/metabolismo , Tetrazoles/administración & dosificación , Animales , Apolipoproteínas E/genética , Apolipoproteínas E/fisiología , Cilostazol , Citocinas/metabolismo , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Humanos , Macrófagos/citología , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Desnudos , Extractos Vegetales/química
13.
Clin Ther ; 32(1): 193-205, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20171424

RESUMEN

BACKGROUND: S-amlodipine gentisate, consisting entirely of the (S)-enantiomer, was developed to increase the potency and improve the safety profile of amlodipine. Regulatory requirements for marketing of S-amlodipine gentisate in Korea require comparison of this agent versus amlodipine racemate. OBJECTIVE: This study was conducted to compare the pharmacodynamic (PD) and pharmacokinetic (PK) characteristics of the S-amlodipine formulation (S-amlodipine gentisate) and amlodipine racemate (amlodipine besylate). METHODS: This study consisted of 2 separate substudies; PD and PK parameters were evaluated separately. Both studies were conducted using a doubleblind, randomized, 2-period, 2-treatment, 2-sequence, double-dummy, single-dose crossover design with S-amlodipine 5 mg and amlodipine racemate 10 mg, separated by a 2-week washout period. Blood pressure (BP) and heart rate were measured in the sitting position before dosing and at 1, 2, 4, 5, 6, 7, 8, 10, 12, 14, 24, 48, and 72 hours after oral administration of S-amlodipine or amlodipine racemate. Impedance cardiography parameters (stroke volume, cardiac index, and systemic vascular resistance) were measured before and at 1, 2, 4, 5, 6, 7, 8, 10, and 12 hours after dosing. For PK assessments, serial blood samples were collected before dosing and at 1, 2, 4, 6, 8, 10, 12, 14, 24, 48, 72, 96, 120, 144, and 168 hours after dosing, and drug concentrations were determined by HPLC-MS/MS. Adverse events (AEs) were collected using self-report or general health-related questions. RESULTS: The PD study included 24 healthy men (mean [SD] age, 23.1 [3.1] years; weight, 69.2 [6.1] kg), and the PK study included 24 different healthy men (mean age, 25.1 [2.1] years; weight, 65.9 [5.9] kg). There were no statistically significant differences between the treatment groups in terms of systolic BP, diastolic BP, or heart rate by repeated-measures ANOVA. Likewise, in the analysis of impedance cardiography, the treatment groups did not display any significant differences in stroke volume, cardiac index, or systemic vascular resistance by repeatedmeasures ANOVA. The mean (SD) AUC(0-last) was 129.7 (62.8) ng . h/mL after dosing with S-amlodipine and 129.0 (59.6) ng . h/mL after dosing with amlodipine racemate. The geometric mean ratio (S-amlodipine: amlodipine racemate) of the S-amlodipine AUC(0-last) was 1.01 (90% CI, 0.90-1.13). In the PD study, 4 AEs in 3 volunteers (3/24; 12.5%) and 8 AEs in 5 volunteers (5/24; 20.8%) were reported after dosing with S-amlodipine and amlodipine racemate, respectively. In the PK study, 18 AEs in 11 volunteers (11/24; 45.8%) and 20 AEs in 9 volunteers (9/24; 37.5%) were reported after dosing with S-amlodipine and amlodipine racemate, respectively. Five volunteers reported AEs after dosing with both S-amlodipine and amlodipine racemate. For the PD and PK studies combined, 30 AEs were judged to be possibly related to S-amlodipine (16 cases) or amlodipine racemate (14 cases). Twenty AEs were judged not to be related to S-amlodipine (6 cases) or amlodipine racemate (14 cases). The most common AEs considered at least possibly related to the study drug in both studies were headache (18 cases) and nausea (3 cases). CONCLUSIONS: In these single-dose studies, no significant differences were found in PD (hemodynamic) or PK parameters between S-amlodipine 5 mg and amlodipine racemate 10 mg. S-amlodipine had a safety profile comparable to that of amlodipine racemate in these healthy male volunteers.


Asunto(s)
Amlodipino/farmacocinética , Presión Sanguínea/efectos de los fármacos , Bloqueadores de los Canales de Calcio/farmacocinética , Frecuencia Cardíaca/efectos de los fármacos , Hemodinámica/efectos de los fármacos , Administración Oral , Adulto , Amlodipino/uso terapéutico , Área Bajo la Curva , Pueblo Asiatico , Bloqueadores de los Canales de Calcio/uso terapéutico , Cardiografía de Impedancia , Cromatografía Líquida de Alta Presión/métodos , Estudios Cruzados , Método Doble Ciego , Electrocardiografía , Humanos , Corea (Geográfico) , Masculino , Espectrometría de Masas en Tándem/métodos , Adulto Joven
14.
Thromb Res ; 124(3): 328-34, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19349067

RESUMEN

Thrombosis and thromboembolic occlusions of major and minor blood vessels are a major complication in various peripheral vascular diseases. Antiplatelet agents (APA), key tools in the treatment of atherothrombosis, therefore became a mainstay medication for a wide range of vascular diseases. Cilostazol and Ginkgo biloba extract (GB), commonly used remedies for peripheral arterial disease, inhibit platelet aggregation with distinct therapeutic mechanisms. In this study, we have investigated if GB can potentiate the antiplatelet effects of cilostazol to explore the utility of combination therapy of cilostazol and GB against peripheral occlusive vascular diseases. GB or cilostazol was evaluated alone or in combination for the antiplatelet activity using in vitro and in vivo models. In addition, potential bleeding side effect of the combinative therapy was assessed by measuring bleeding time, prothrombin time (PT) and activated partial thromboplastin time (aPTT) in vivo after oral administration. In in vitro assays using freshly isolated human platelets, the combination of cilostazol and GB showed superior inhibition of both the shear and the collagen-induced platelet aggregation to those of each drug alone. In accordance with these enhanced in vitro antiplatelet activities, the combinative therapy showed enhanced anti-thrombotic effects in in vivo pulmonary embolism model and arterial thrombosis model. In particular, the increase of survival rate in pulmonary embolism model by combination treatment of cilostazol (25 mg/kg) and GB (20 mg/kg) was higher more than two-fold of those of the respective drugs. Notably, the combination of cilostazol and GB did not show a significant effect on the bleeding time, PT and aPTT increase, suggesting that GB may potentiate the antiplatelet effect of cilostazol without the prolongation of bleeding time or coagulation time. With these studies, we suggest that combinative therapy of GB and cilostazol might offer enhanced anti-thrombotic efficacies without increasing side-effects.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Fibrinolíticos/administración & dosificación , Ginkgo biloba/química , Extractos Vegetales/administración & dosificación , Inhibidores de Agregación Plaquetaria/administración & dosificación , Tetrazoles/administración & dosificación , Trombosis/tratamiento farmacológico , Animales , Tiempo de Sangría , Células Cultivadas , Cilostazol , Sinergismo Farmacológico , Humanos , Masculino , Ratas , Terapéutica , Trombosis/diagnóstico , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA