Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Atherosclerosis ; 244: 29-37, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26584136

RESUMEN

OBJECTIVE: Dietary l-carnitine can be metabolized by intestinal microbiota to trimethylamine, which is absorbed by the gut and further oxidized to trimethylamine N-oxide (TMAO) in the liver. TMAO plasma levels have been associated with atherosclerosis development in ApoE(-/-) mice. To better understand the mechanisms behind this association, we conducted in vitro and in vivo studies looking at the effect of TMAO on different steps of atherosclerotic disease progression. METHODS: J774 mouse macrophage cells were used to evaluate the effect of TMAO on foam cell formation. Male ApoE(-/-) mice transfected with human cholesteryl ester transfer protein (hCETP) were fed l-carnitine and/or methimazole, a flavin monooxygenase 3 (FMO3) inhibitor that prevents the formation of TMAO. Following 12 week treatment, l-carnitine and TMAO plasma levels, aortic lesion development, and lipid profiles were determined. RESULTS: TMAO at concentrations up to 10-fold the Cmax reported in humans did not affect in vitro foam cell formation. In ApoE(-/-)mice expressing hCETP, high doses of l-carnitine resulted in a significant increase in plasma TMAO levels. Surprisingly, and independently from treatment group, TMAO levels inversely correlated with aortic lesion size in both aortic root and thoracic aorta. High TMAO levels were found to significantly correlate with smaller aortic lesion area. Plasma lipid and lipoprotein levels did not change with treatment nor with TMAO levels, suggesting that the observed effects on lesion area were independent from lipid changes. CONCLUSION: These findings suggest that TMAO slows aortic lesion formation in this mouse model and may have a protective effect against atherosclerosis development in humans.


Asunto(s)
Aterosclerosis/sangre , Carnitina/farmacocinética , Proteínas de Transferencia de Ésteres de Colesterol/biosíntesis , Metilaminas/sangre , Animales , Apolipoproteínas E/genética , Células Cultivadas , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Noqueados
2.
Steroids ; 74(13-14): 1015-24, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19665469

RESUMEN

Endometriosis, defined as the presence of endometrial glands and stroma at extra-uterine sites, is a gynecological condition that affects women of reproductive age. Consistent with its uterine origins, endometriotic lesions and resulting symptoms are hormonally responsive. To investigate Progesterone Receptor (PR)-based therapies, we measured physiological endpoints and gene expression in rat models of uterine cell estrogenic activity. Estrogen-induced ELT-3 rat leiomyoma cell proliferation was significantly inhibited by progesterone (P4), while the antiprogestin RU486 or the Selective PR Modulator (SPRM) asoprisnil, did not block proliferation. Stromal cell-derived factor-1 (SDF-1/Cxcl12) gene expression was induced by estrogen, and was repressed by the Selective Estrogen Receptor Modulators (SERMs), the antiestrogen ICI 182,780, and P4, but not by RU486 or the ERbeta-selective ligand ERB-041. In ELT-3 cells, asoprisnil demonstrated partial PR agonism on SDF-1 gene repression. Magnetic Resonance Imaging was used to monitor development of ectopic cysts in a rat surgical model of endometriosis. SERMs and P4 significantly decreased cyst volumes comparably by approximately 60%. However, ERB-041 and asoprisnil had no effect on cyst volume, and RU486 increased cyst volume by 20%. SDF-1 expression was modestly, but significantly, increased in the cyst compared to eutopic uterus, and P4 and raloxifene could repress the expression. We showed that SDF-1 was similarly regulated in human cells. These data suggest that transcriptional regulation of SDF-1 is a surrogate marker of estrogenic activities via ERalpha in rat uterine cells, and that SDF-1 repression by PR agonists can predict the ability to oppose the actions of estrogen in vivo.


Asunto(s)
Quimiocina CXCL12/antagonistas & inhibidores , Endometriosis/tratamiento farmacológico , Progesterona/uso terapéutico , Progestinas/uso terapéutico , Receptores de Progesterona/agonistas , Moduladores Selectivos de los Receptores de Estrógeno/uso terapéutico , Útero/efectos de los fármacos , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quimiocina CXCL12/agonistas , Quimiocina CXCL12/metabolismo , Quistes/tratamiento farmacológico , Quistes/metabolismo , Quistes/patología , Modelos Animales de Enfermedad , Endometriosis/metabolismo , Endometriosis/patología , Estradiol/análogos & derivados , Estradiol/farmacología , Estrenos/farmacología , Antagonistas de Estrógenos/farmacología , Receptor alfa de Estrógeno/metabolismo , Estrógenos/farmacología , Femenino , Fulvestrant , Expresión Génica , Antagonistas de Hormonas/farmacología , Humanos , Mifepristona/farmacología , Oxazoles/farmacología , Oximas/farmacología , Progesterona/farmacología , Progestinas/farmacología , Ratas , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Útero/metabolismo
3.
Bioorg Med Chem Lett ; 19(15): 4292-5, 2009 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-19487124

RESUMEN

This Letter discloses a series of 2-aminothiadiazole amides as selective EP(3) receptor antagonists. SAR optimization resulted in compounds with excellent functional activity in vitro. In addition, efforts to optimize DMPK properties in the rat are discussed. These efforts have resulted in the identification of potent, selective EP(3) receptor antagonists with excellent DMPK properties suitable for in vivo studies.


Asunto(s)
Amidas/química , Química Farmacéutica/métodos , Receptores de Prostaglandina E/antagonistas & inhibidores , Receptores de Prostaglandina E/química , Tiadiazoles/química , Administración Oral , Animales , Perros , Diseño de Fármacos , Humanos , Modelos Químicos , Estructura Molecular , Unión Proteica , Ratas , Ratas Sprague-Dawley , Subtipo EP3 de Receptores de Prostaglandina E , Relación Estructura-Actividad
4.
J Pharmacol Exp Ther ; 326(2): 432-42, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18499743

RESUMEN

The transient receptor potential (TRP) vanilloid 4 (TRPV4) member of the TRP superfamily has recently been implicated in numerous physiological processes. In this study, we describe a small molecule TRPV4 channel activator, (N-((1S)-1-{[4-((2S)-2-{[(2,4-dichlorophenyl)sulfonyl]amino}-3-hydroxypropanoyl)-1-piperazinyl]carbonyl}-3-methylbutyl)-1-benzothiophene-2-carboxamide (GSK1016790A), which we have used as a valuable tool in investigating the role of TRPV4 in the urinary bladder. GSK1016790A elicited Ca2+ influx in mouse and human TRPV4-expressing human embryonic kidney (HEK) cells (EC50 values of 18 and 2.1 nM, respectively), and it evoked a dose-dependent activation of TRPV4 whole-cell currents at concentrations above 1 nM. In contrast, the TRPV4 activator 4alpha-phorbol 12,13-didecanoate (4alpha-PDD) was 300-fold less potent than GSK1016790A in activating TRPV4 currents. TRPV4 mRNA was detected in urinary bladder smooth muscle (UBSM) and urothelium of TRPV4+/+ mouse bladders. Western blotting and immunohistochemistry demonstrated protein expression in both the UBSM and urothelium that was absent in TRPV4-/- bladders. TRPV4 activation with GSK1016790A contracted TRPV4+/+ mouse bladders in vitro, both in the presence and absence of the urothelium, an effect that was undetected in TRPV4-/- bladders. Consistent with the effects on TRPV4 HEK whole-cell currents, 4alpha-PDD demonstrated a weak ability to contract bladder strips compared with GSK1016790A. In vivo, urodynamics in TRPV4+/+ and TRPV4-/- mice revealed an enhanced bladder capacity in the TRPV4-/- mice. Infusion of GSK1016790A into the bladders of TRPV4+/+ mice induced bladder overactivity with no effect in TRPV4-/- mice. Overall TRPV4 plays an important role in urinary bladder function that includes an ability to contract the bladder as a result of the expression of TRPV4 in the UBSM.


Asunto(s)
Leucina/análogos & derivados , Contracción Muscular/efectos de los fármacos , Sulfonamidas/farmacología , Canales Catiónicos TRPV/agonistas , Vejiga Urinaria/efectos de los fármacos , Urodinámica/efectos de los fármacos , Urotelio/efectos de los fármacos , Animales , Peso Corporal/efectos de los fármacos , Femenino , Leucina/farmacología , Masculino , Ratones , Ratones Noqueados , Estructura Molecular , Músculo Liso/efectos de los fármacos , Músculo Liso/metabolismo , Forboles/farmacología , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/fisiología , Vejiga Urinaria/metabolismo , Urotelio/metabolismo
5.
J Pharmacol Exp Ther ; 326(2): 443-52, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18499744

RESUMEN

The transient receptor potential (TRP) vanilloid subtype 4 (V4) is a nonselective cation channel that exhibits polymodal activation and is expressed in the endothelium, where it contributes to intracellular Ca2+ homeostasis and regulation of cell volume. The purpose of the present study was to evaluate the systemic cardiovascular effects of GSK1016790A, a novel TRPV4 activator, and to examine its mechanism of action. In three species (mouse, rat, and dog), the i.v. administration of GSK1016790A induced a dose-dependent reduction in blood pressure, followed by profound circulatory collapse. In contrast, GSK1016790A had no acute cardiovascular effects in the TRPV4-/- null mouse. Hemodynamic analyses in the dog and rat demonstrate a profound reduction in cardiac output. However, GSK1016790A had no effect on rate or contractility in the isolated, buffer-perfused rat heart, and it produced potent endothelial-dependent relaxation of rodent-isolated vascular ring segments that were abolished by nitric-oxide synthase (NOS) inhibition (N-nitro-L-arginine methyl ester; L-NAME), ruthenium red, and endothelial NOS (eNOS) gene deletion. However, the in vivo circulatory collapse was not altered by NOS inhibition (L-NAME) or eNOS gene deletion but was associated with (concentration and time appropriate) profound vascular leakage and tissue hemorrhage in the lung, intestine, and kidney. TRPV4 immunoreactivity was localized in the endothelium and epithelium in the affected organs. GSK1016790A potently induced rapid electrophysiological and morphological changes (retraction/condensation) in cultured endothelial cells. In summary, inappropriate activation of TRPV4 produces acute circulatory collapse associated with endothelial activation/injury and failure of the pulmonary microvascular permeability barrier. It will be important to determine the role of TRPV4 in disorders associated with edema and microvascular congestion.


Asunto(s)
Aorta Torácica/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Hemodinámica/efectos de los fármacos , Leucina/análogos & derivados , Sulfonamidas/efectos adversos , Canales Catiónicos TRPV/agonistas , Función Ventricular Izquierda/efectos de los fármacos , Animales , Aorta Torácica/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Línea Celular , Perros , Relación Dosis-Respuesta a Droga , Endotelio Vascular/metabolismo , Femenino , Humanos , Inmunohistoquímica , Leucina/efectos adversos , Leucina/farmacocinética , Masculino , Ratones , Ratones Noqueados , Estructura Molecular , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Sulfonamidas/farmacocinética , Canales Catiónicos TRPV/genética , Vasoconstricción/efectos de los fármacos
6.
Mol Endocrinol ; 21(5): 1066-81, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17356170

RESUMEN

Selective progesterone receptor modulators (SPRMs) have been suggested as therapeutic agents for treatment of gynecological disorders. One such SPRM, asoprisnil, was recently in clinical trials for treatment of uterine fibroids and endometriosis. We present the crystal structures of progesterone receptor (PR) ligand binding domain complexed with asoprisnil and the corepressors nuclear receptor corepressor (NCoR) and SMRT. This is the first report of steroid nuclear receptor crystal structures with ligand and corepressors. These structures show PR in a different conformation than PR complexed with progesterone (P4). We profiled asoprisnil in PR-dependent assays to understand further the PR-mediated mechanism of action. We confirmed previous findings that asoprisnil demonstrated antagonism, but not agonism, in a PR-B transfection assay and the T47D breast cancer cell alkaline phosphatase activity assay. Asoprisnil, but not RU486, weakly recruited the coactivators SRC-1 and AIB1. However, asoprisnil strongly recruited the corepressor NCoR in a manner similar to RU486. Unlike RU486, NCoR binding to asoprisnil-bound PR could be displaced with equal affinity by NCoR or TIF2 peptides. We further showed that it weakly activated T47D cell gene expression of Sgk-1 and PPL and antagonized P4-induced expression of both genes. In rat leiomyoma ELT3 cells, asoprisnil demonstrated partial P4-like inhibition of cyclooxygenase (COX) enzymatic activity and COX-2 gene expression. In the rat uterotrophic assay, asoprisnil demonstrated no P4-like ability to oppose estrogen. Our data suggest that asoprisnil differentially recruits coactivators and corepressors compared to RU486 or P4, and this specific cofactor interaction profile is apparently insufficient to oppose estrogenic activity in rat uterus.


Asunto(s)
Estrenos/química , Estrenos/farmacología , Oximas/química , Oximas/farmacología , Receptores de Progesterona/efectos de los fármacos , Neoplasias de la Mama , Línea Celular Tumoral , Cristalografía por Rayos X , Estradiol/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Modelos Moleculares , Plásmidos , Reacción en Cadena de la Polimerasa , Conformación Proteica , Receptores de Progesterona/química , Receptores de Progesterona/genética , Receptores de Progesterona/fisiología , Transfección
7.
Fertil Steril ; 88(4 Suppl): 1058-64, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17362941

RESUMEN

OBJECTIVE: To non-invasively characterize ectopic uterine tissue (EUT) development in a modified autologous rat surgical model of endometriosis using magnetic resonance imaging (MRI). DESIGN: Investigational MRI study. SETTING: A pharmaceutical company. ANIMAL(S): Female Sprague Dawley rats. INTERVENTION(S): Uterine tissue was autotransplanted on the right peritoneal wall of rats. Rats were serially imaged after surgery and after endogenous hormone suppression, hormone supplementation, or ovariectomy. In addition, an MRI contrast agent was administered to examine EUT perfusion characteristics. MAIN OUTCOME MEASURE(S): Changes in transplanted EUT volume and perfusion were monitored using MRI. RESULT(S): The EUT growth could be readily monitored non-invasively by MRI. Although EUT growth was rapid during the initial 4 days after surgery, volume stabilized by the third week and maintained for at least 9 weeks after transplantation. The EUT volumes varied with the estrous cycle and were hormonally sensitive to ovariectomy, to Antide (GnRH antagonist), and to Antide followed by 17beta-E(2) supplementation. The use of an MRI contrast agent facilitated visualization of EUT wall perfusion. CONCLUSION(S): MRI allows for noninvasive, dynamic evaluation of transplanted EUT growth in the rat. This reproducible model will allow for performing quantifiable pharmacologic studies in pre-clinical drug discovery for therapies targeting endometriosis.


Asunto(s)
Coristoma/diagnóstico , Imagen por Resonancia Magnética/métodos , Cavidad Peritoneal , Útero , Animales , Coristoma/fisiopatología , Endometriosis/diagnóstico , Endometriosis/fisiopatología , Ciclo Estral/fisiología , Femenino , Ratas , Ratas Sprague-Dawley
8.
J Pharmacol Exp Ther ; 315(3): 1306-13, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16144980

RESUMEN

We describe N-[(2S)-2-(mercaptomethyl)-3-methylbutanoyl]-4-(1H-pyrazol-1-yl)-L-phenylalanine (GW796406), a vasopeptidase inhibitor (VPI) that possessed approximately 3-fold selectivity for neutral endopeptidase 24.11 (NEP) versus angiotensin-converting enzyme (ACE) in in vitro assays using rat and human enzymes. In the same assays, omapatrilat, the most extensively studied VPI, displayed approximately 3-fold selectivity for ACE. The in vivo ACE and NEP inhibition profile and the liability of the compounds to increase plasma extravasation were compared at two (low and high) therapeutically equivalent intravenous doses in the rat. At the low dose, both agents inhibited ACE activity by approximately 85%. Consistent with their in vitro ACE/NEP selectivity, omapatrilat produced 49% inhibition, whereas GW796406 produced >95% inhibition of NEP. Neither compound increased plasma extravasation. When the low dose was administered to rats pretreated with the NEP inhibitor ecadotril to normalize NEP background to <5% of control, only omapatrilat significantly increased plasma extravasation. At the high dose, omapatrilat and GW796406 produced profound, nonselective inhibition of ACE (>90%) and NEP (>95%), and they significantly increased plasma extravasation. The activity of the agents as inhibitors of dipeptidylpeptidase IV (DPP IV) and aminopeptidase P (APP) was also investigated. Neither compound inhibited DPP IV. Interestingly, omapatrilat, but not GW796406, was a relatively potent inhibitor of APP (IC50 = 260 nM). We investigated whether APP inhibition increased the plasma extravasation liability of GW796406. The low dose of GW796406 administered with apstatin, an APP inhibitor, did not increase plasma extravasation. This finding inferred that APP inhibition is not involved in plasma extravasation in the rat and that APP inhibition does not explain the increased plasma extravasation produced by omapatrilat in NEP-inhibited rats.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Neprilisina/farmacología , Plasma/efectos de los fármacos , Piridinas/farmacología , Tiazepinas/farmacología , Aminopeptidasas/análisis , Aminopeptidasas/antagonistas & inhibidores , Inhibidores de la Enzima Convertidora de Angiotensina/análisis , Animales , Dipeptidil Peptidasa 4/análisis , Relación Dosis-Respuesta a Droga , Humanos , Concentración 50 Inhibidora , Riñón/efectos de los fármacos , Riñón/enzimología , Pulmón/efectos de los fármacos , Pulmón/enzimología , Masculino , Neprilisina/análisis , Péptidos/farmacología , Fenilalanina/análogos & derivados , Fenilalanina/química , Fenilalanina/farmacología , Plasma/fisiología , Pirazoles/química , Pirazoles/farmacología , Conejos , Ratas , Ratas Sprague-Dawley , Ratas Wistar
9.
J Pharmacol Exp Ther ; 309(3): 1141-7, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-14769834

RESUMEN

The effect of angiotensin-converting enzyme (ACE) and neutral endopeptidase (NEP) inhibition on microvascular plasma leakage (extravasation) was evaluated in a rat model. Progressive inhibition of ACE using captopril caused increased extravasation when lung ACE was inhibited by >55%. In contrast, the selective inhibition of renal NEP by >90% using ecadotril did not increase extravasation. In NEP-inhibited rats, extravasation produced by the ACE inhibitors captopril and lisinopril was markedly enhanced. The dual ACE and NEP inhibitor omapatrilat, at oral doses of 0.03, 0.1, and 0.3 mg/kg, selectively inhibited lung ACE by 19, 61, and 76%, respectively, and did not cause significant extravasation. Doses of 1 and 10 mg/kg omapatrilat, which produced >90% inhibition of ACE and also inhibited renal NEP by 54 and 78%, respectively, significantly increased extravasation. In this model, bradykinin and substance P produced extravasation that could be abolished by the bradykinin 2 (B2) receptor antagonist Hoe 140 (icatibant) or the neurokinin1 (NK1) antagonist CP99994 [(+)-(2S,3S)-3-(2-methoxybenzylamino)-2-phenylpiperidine], respectively. Bradykinin induced extravasation was also partially ( approximately 40%) inhibited by CP99994, indicating that a portion of the response involves B2 receptor-mediated release of substance P. In conclusion, this study is the first to relate the degree of ACE and/or NEP inhibition to extravasation liability in the rat model. Our data clearly demonstrate that ACE inhibitor-induced plasma extravasation is enhanced by concomitant inhibition of NEP. In addition, this study provides further evidence for the role for B2 and NK1 receptors in mediating plasma extravasation in the rat.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Bradiquinina/análogos & derivados , Captopril/farmacología , Neprilisina/antagonistas & inhibidores , Peptidil-Dipeptidasa A/metabolismo , Plasma/efectos de los fármacos , Tiorfan/análogos & derivados , Animales , Bradiquinina/farmacología , Relación Dosis-Respuesta a Droga , Pulmón/efectos de los fármacos , Pulmón/enzimología , Masculino , Plasma/fisiología , Ratas , Ratas Sprague-Dawley , Receptor de Bradiquinina B1/efectos de los fármacos , Receptor de Bradiquinina B1/fisiología , Receptor de Bradiquinina B2/efectos de los fármacos , Receptor de Bradiquinina B2/fisiología , Receptores de Neuroquinina-1/efectos de los fármacos , Receptores de Neuroquinina-1/fisiología , Sustancia P/farmacología , Tiorfan/farmacología
10.
J Pharmacol Exp Ther ; 301(3): 987-92, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12023529

RESUMEN

Two endogenous receptors for the potent smooth muscle-stimulating peptide neuromedin U (NmU) have recently been identified and cloned. Pharmacological, binding, and expression studies were conducted in an attempt to determine the receptor(s) involved in the smooth muscle-stimulating effects of NmU. The NmU peptides caused a concentration-dependent contraction of canine isolated urinary bladder. NmU did not have this same effect in the urinary bladder from rat, guinea pig, rabbit, mouse, or ferret. Although NmU had no effect on canine uterus it did cause contraction of canine stomach, ileum, and colon. As well as causing contraction of canine bladder in vitro, NmU administered systemically resulted in a significant increase in urinary bladder pressure in vivo. High-affinity binding sites for NmU were identified in canine bladder. The four NmU peptides porcine NmU-8, rat NmU-23, human NmU-25, and porcine NmU-25 displaced (125)I-NmU-25 binding with similar K(i) values (0.08-0.24 nM). A different binding profile was revealed in human embryonic kidney-293 cells transiently expressed with the canine NmU-2 receptor where porcine NmU-8 (K(i) = 147.06 nM) was much less potent than the other NmU peptides. Using TaqMan, expression of NmU-1 was detected in human urinary bladder, small intestine, colon, and uterus. Expression of NmU-2 was much lower or absent in these human tissues and undetectable in canine bladder and stomach. The results of this study reveal significant species differences in the activity of NmU. The contractile activity in human and canine smooth muscle seems to be mediated by the recently cloned NmU-1 receptor.


Asunto(s)
Proteínas de la Membrana , Músculo Liso/efectos de los fármacos , Músculo Liso/metabolismo , Neuropéptidos/metabolismo , Receptores de Neurotransmisores , Animales , Sitios de Unión/efectos de los fármacos , Sitios de Unión/fisiología , Línea Celular , Perros , Evaluación Preclínica de Medicamentos/métodos , Femenino , Hurones , Cobayas , Humanos , Mucosa Intestinal/metabolismo , Intestinos/efectos de los fármacos , Masculino , Ratones , Contracción Muscular/efectos de los fármacos , Contracción Muscular/fisiología , Neuropéptidos/farmacología , Conejos , Ratas , Receptores de Superficie Celular/biosíntesis , Vejiga Urinaria/efectos de los fármacos , Vejiga Urinaria/metabolismo , Útero/efectos de los fármacos , Útero/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...