Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros

Banco de datos
Tipo de estudio
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Biochim Biophys Acta ; 1831(1): 117-25, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23127512

RESUMEN

Due to its antiapoptotic action, derivatives of the lipid mediator lysophosphatidic acid (LPA) provide potential therapeutic utility in diseases associated with programmed cell death. Apoptosis is one of the major pathophysiological processes elicited by radiation injury to the organism. Consequently, therapeutic explorations applying compounds that mimic the antiapoptotic action of LPA have begun. Here we present a brief account of our decade-long drug discovery effort aimed at developing LPA mimics with a special focus on specific agonists of the LPA(2) receptor subtype, which was found to be highly effective in protecting cells from apoptosis. We describe new evidence that 2-((3-(1,3-dioxo-1H-benzo[de]isoquinolin-2(3H)-yl)propyl)thio)benzoic acid (GRI977143), a prototypic nonlipid agonist specific to the LPA(2) receptor subtype, rescues apoptotically condemned cells in vitro and in vivo from injury caused by high-dose γ-irradiation. GRI977143 shows the features of a radiomitigator because it is effective in rescuing the lives of mice from deadly levels of radiation when administered 24h after radiation exposure. Our findings suggest that by specifically activating LPA(2) receptors GRI977143 activates the ERK1/2 prosurvival pathway, effectively reduces Bax translocation to the mitochondrion, attenuates the activation of initiator and effector caspases, reduces DNA fragmentation, and inhibits PARP-1 cleavage associated with γ-irradiation-induced apoptosis. GRI977143 also inhibits bystander apoptosis elicited by soluble proapoptotic mediators produced by irradiated cells. Thus, GRI977143 can serve as a prototype scaffold for lead optimization paving the way to more potent analogs amenable for therapeutic exploration. This article is part of a Special Issue entitled Advances in Lysophospholipid Research.


Asunto(s)
Traumatismos por Radiación/metabolismo , Traumatismos por Radiación/prevención & control , Receptores del Ácido Lisofosfatídico/metabolismo , Síndrome de Radiación Aguda/tratamiento farmacológico , Síndrome de Radiación Aguda/patología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Efecto Espectador/efectos de los fármacos , Efecto Espectador/efectos de la radiación , Inhibidores de Caspasas/farmacología , Caspasas/metabolismo , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/efectos de la radiación , Citoprotección/efectos de los fármacos , Citoprotección/efectos de la radiación , Fragmentación del ADN/efectos de los fármacos , Fragmentación del ADN/efectos de la radiación , Embrión de Mamíferos/citología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/efectos de la radiación , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Fibroblastos/patología , Fibroblastos/efectos de la radiación , Rayos gamma , Lisofosfolípidos/química , Lisofosfolípidos/metabolismo , Lisofosfolípidos/farmacología , Ratones , Ratones Noqueados , Compuestos Organofosforados/farmacología , Hidrolasas Diéster Fosfóricas/genética , Hidrolasas Diéster Fosfóricas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Traumatismos por Radiación/patología , Análisis de Supervivencia
2.
Xenobiotica ; 41(11): 1006-12, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21864202

RESUMEN

KZ-41, a quinic acid derivative, significantly reduces mortality in a murine model of hematopoietic acute radiation syndrome. The purpose of this study was to evaluate the systemic pharmacokinetics, elimination, and oral bioavailability of KZ-41 in rats. Male Sprague-Dawley rats (n = 6 per group) received a single dose (10 mg/kg) of KZ-41 administered either intravenously via the jugular vein or orally via gavage. In vitro stability was determined using both rat liver microsomes and the bacteria Gluconobacter oxydans. KZ-41 concentrations were determined using LC-MS/MS (liquid chromatography tandom mass spectrometry). Half-life of KZ-41 was ≈3 hr after either intravenous or oral administration. Mean volume of distribution was 3.3 L/kg. Extent of absorption (F) after oral administration was estimated to be ~100%, which was consistent with the finding that KZ-41 was stable to liver microsomal and bacterial degradation. Following intravenous administration, KZ-41 demonstrated a medium clearance and volume of distribution with a terminal half-life of ≈3 hr. KZ-41 was rapidly and completely absorbed (F ≅ 1), which was consistent with the findings that KZ-41 is resistant to presystemic elimination mechanisms (i.e. enteric bacterial degradation and hepatic metabolism). Thus, KZ-41 represents an excellent candidate for further development as an orally available agent for the mitigation of radiation injury.


Asunto(s)
Ácido Quínico/análogos & derivados , Ácido Quínico/farmacocinética , Protectores contra Radiación/farmacocinética , Administración Oral , Animales , Cromatografía Liquida , Estabilidad de Medicamentos , Inyecciones Intravenosas , Masculino , Espectrometría de Masas , Ácido Quínico/sangre , Ácido Quínico/química , Ácido Quínico/farmacología , Protectores contra Radiación/química , Protectores contra Radiación/farmacología , Ratas , Ratas Sprague-Dawley , Estándares de Referencia , Reproducibilidad de los Resultados , Factores de Tiempo
3.
Bioorg Med Chem Lett ; 19(18): 5458-60, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19674895

RESUMEN

Quinic acid (QA) esters found in hot water extracts of Uncaria tomentosa (a.k.a. cat's claw) exert anti-inflammatory activity through mechanisms involving inhibition of the pro-inflammatory transcription factor nuclear factor kappa B (NF-kappaB). Herein, we describe the synthesis and biological testing of novel QA derivatives. Inhibition of NF-kappaB was assessed using A549 (Type II alveolar epithelial-like) cells that stably express a secreted alkaline phosphatase (SEAP) reporter driven by an NF-kappaB response element. A549-NF-kappaB cells were stimulated with TNF-alpha (10 ng/mL) in the presence or absence of QA derivative for 18 hours followed by measurement of SEAP activity. Amide substitution at the carboxylic acid position yielded potent inhibitors of NF-kappaB. A variety of modifications to the amide substitution were tolerated with the N-propyl amide derivative being the most potent. Further examination of the SAR demonstrated that acetylation of the hydroxyl groups reduced NF-kappaB inhibitory activity. QA amide derivatives lacked anti-oxidant activity and were found to be neither anti-proliferative nor cytotoxic at concentrations up to 100 microM. In conclusion, we have discovered a novel series of non-toxic QA amides that potently inhibit NF-kappaB, despite their lack of anti-oxidant activity. Mechanistic studies and pre-clinical efficacy studies in various inflammatory animal models are on-going.


Asunto(s)
Antiinflamatorios/química , Antiinflamatorios/farmacología , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Ácido Quínico/química , Ácido Quínico/farmacología , Fosfatasa Alcalina/metabolismo , Antiinflamatorios/síntesis química , Línea Celular , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Humanos , Alveolos Pulmonares/citología , Ácido Quínico/síntesis química , Uncaria/química
4.
Radiat Res ; 183(4): 465-75, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25807318

RESUMEN

We have previously demonstrated that the small molecule octadecenyl thiophosphate (OTP), a synthetic mimic of the growth factor-like mediator lysophosphatidic acid (LPA), showed radioprotective activity in a mouse model of total-body irradiation (TBI) when given orally or intraperitoneally 30 min before exposure to 9 Gy γ radiation. In the current study, we evaluated the effects of OTP, delivered subcutaneously, for radioprotection or radiomitigation from -24 h before to up to +72 h postirradiation using a mouse TBI model with therapeutic doses at around 1 mg/kg. OTP was injected at 10 mg/kg without observable toxic side effects in mice, providing a comfortable safety margin. Treatment of C57BL/6 mice with a single dose of OTP over the time period from -12 h before to +26 h after a lethal dose of TBI reduced mortality by 50%. When administered at +48 h to +72 h postirradiation (LD50/30 to LD100/30), OTP reduced mortality by ≥34%. OTP administered at +24 h postirradiation significantly elevated peripheral white blood cell and platelet counts, increased crypt survival in the jejunum, enhanced intestinal glucose absorption and reduced endotoxin seepage into the blood. In the 6.4-8.6 Gy TBI range using LD50/10 as the end point, OTP yielded a dose modification factor of 1.2. The current data indicate that OTP is a potent radioprotector and radiomitigator ameliorating the mortality and tissue injury of acute hematopoietic as well as acute gastrointestinal radiation syndrome.


Asunto(s)
Síndrome de Radiación Aguda/prevención & control , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/efectos de la radiación , Hematopoyesis/efectos de los fármacos , Hematopoyesis/efectos de la radiación , Lisofosfolípidos/metabolismo , Compuestos Organofosforados/farmacología , ATPasas Asociadas con Actividades Celulares Diversas , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Antígenos CD34/metabolismo , Transporte Biológico/efectos de los fármacos , Transporte Biológico/efectos de la radiación , Materiales Biomiméticos/efectos adversos , Materiales Biomiméticos/farmacocinética , Materiales Biomiméticos/farmacología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Células Clonales/citología , Células Clonales/efectos de los fármacos , Células Clonales/efectos de la radiación , Relación Dosis-Respuesta a Droga , Femenino , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/microbiología , Glucosa/metabolismo , Células HEK293 , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de la radiación , Humanos , Proteínas con Dominio LIM/metabolismo , Recuento de Leucocitos , Ratones , Ratones Endogámicos C57BL , Compuestos Organofosforados/efectos adversos , Compuestos Organofosforados/farmacocinética , Fosfoproteínas/metabolismo , Recuento de Plaquetas , Complejo de la Endopetidasa Proteasomal , Protectores contra Radiación/efectos adversos , Protectores contra Radiación/farmacocinética , Protectores contra Radiación/farmacología , Intercambiadores de Sodio-Hidrógeno/metabolismo , Factores de Transcripción/metabolismo , Irradiación Corporal Total/efectos adversos
5.
Invest Ophthalmol Vis Sci ; 53(4): 2439-45, 2012 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-22427570

RESUMEN

Purpose. Super-selective intra-ophthalmic artery chemotherapy (SSIOAC) is an eye-targeted drug-delivery strategy to treat retinoblastoma, the most prevalent primary ocular malignancy in children. Unfortunately, recent clinical reports associate adverse vascular toxicities with SSIOAC using melphalan, the most commonly used chemotherapeutic. Methods. To explore reasons for the unexpected vascular toxicities, we examined the effects of melphalan, as well as carboplatin (another chemotherapeutic used with retinoblastoma), in vitro using primary human retinal endothelial cells, and in vivo using a non-human primate model, which allowed us to monitor the retina in real time during SSIOAC. Results. Both melphalan and carboplatin triggered human retinal endothelial cell migration, proliferation, apoptosis, and increased expression of adhesion proteins intracellullar adhesion molecule-1 [ICAM-1] and soluble chemotactic factors (IL-8). Melphalan increased monocytic adhesion to human retinal endothelial cells. Consistent with these in vitro findings, histopathology showed vessel wall endothelial cell changes, leukostasis, and vessel occlusion. Conclusions. These results reflect a direct interaction of chemotherapeutic drugs with both the vascular endothelium and monocytes. The vascular toxicity may be related to the pH, the pulsatile delivery, or the chemotherapeutic drugs used. Our long-term goal is to determine if changes in the drug of choice and/or delivery procedures will decrease vascular toxicity and lead to better eye-targeted treatment strategies.


Asunto(s)
Antineoplásicos Alquilantes/toxicidad , Células Endoteliales/efectos de los fármacos , Melfalán/toxicidad , Arteria Oftálmica/efectos de los fármacos , Animales , Antineoplásicos/toxicidad , Carboplatino/toxicidad , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucina-8/metabolismo , Macaca mulatta , Neutrófilos/metabolismo , ARN Mensajero/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA