Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
J Pharmacol Exp Ther ; 329(3): 928-37, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19255283

RESUMEN

ABT-869 [N-(4-(3-amino-1H-indazol-4-yl)phenyl)-N'-(2-fluoro-5-methylphenyl)urea] is a novel multitargeted inhibitor of the vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF) receptor tyrosine kinase family members. ABT-869 demonstrates tumor growth inhibition in multiple preclinical animal models and in early clinical trials. VEGF receptor inhibition is also associated with reversible hypertension that may limit its benefit clinically. To evaluate optimal therapeutic approaches to prevent hypertension with VEGF receptor inhibition, we characterized the dose-dependent effects of seven antihypertensive agents from three mechanistic classes [angiotensin-converting enzyme inhibitors (ACEis), angiotensin receptor blockers (ARBs), calcium channel blockers (CCBs)] on hypertension induced by ABT-869 in conscious telemetry rats. We report that ABT-869-induced hypertension can be prevented and reversed with subtherapeutic or therapeutic doses of antihypertensive drugs with a general rank order of ACEi > ARB > CCB. In SCID mice, the ACE inhibitor, enalapril (C(20)H(28)N(2)O(5) x C(4)H(4)O(4)) at 30 mg/kg, prevented hypertension, with no attenuation of the antitumor efficacy of ABT-869. These studies demonstrate that the adverse cardiovascular effects of the VEGF/PDGF receptor tyrosine kinase inhibitor, ABT-869, are readily controlled by conventional antihypertensive therapy without affecting antitumor efficacy.


Asunto(s)
Antihipertensivos/farmacología , Presión Sanguínea/efectos de los fármacos , Indazoles/farmacología , Neoplasias/tratamiento farmacológico , Compuestos de Fenilurea/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Acrilatos/farmacología , Amlodipino/farmacología , Antagonistas de Receptores de Angiotensina , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Bencimidazoles/farmacología , Benzoatos/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Relación Dosis-Respuesta a Droga , Enalapril/farmacología , Humanos , Imidazoles/farmacología , Indazoles/efectos adversos , Indazoles/uso terapéutico , Lisinopril/farmacología , Masculino , Ratones , Ratones SCID , Neoplasias/patología , Nifedipino/farmacología , Compuestos de Fenilurea/efectos adversos , Compuestos de Fenilurea/uso terapéutico , Inhibidores de Proteínas Quinasas/sangre , Inhibidores de Proteínas Quinasas/uso terapéutico , Ramipril/farmacología , Ratas , Ratas Sprague-Dawley , Telmisartán , Tiofenos/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
2.
J Cardiovasc Pharmacol ; 53(2): 173-8, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19188829

RESUMEN

ABT-869 is a novel multitargeted inhibitor of vascular endothelial growth factor and platelet-derived growth factor receptor tyrosine kinases (RTKs) with potent antiangiogenic properties that slow tumor progression. Vascular endothelial growth factor receptor blockade has been shown to produce hypertension. Atrasentan is a potent and selective endothelin (ETA) receptor antagonist that lowers blood pressure and affects tumor growth. To assess the utility of ETA receptor blockade in controlling hypertension with RTK inhibition, we evaluated the ability of atrasentan to block hypertension with ABT-869 in conscious, telemetry-instrumented rats. Changes in mean arterial pressure (MAP) and heart rate (HR) were evaluated using mean values and the area under the curve (AUC). Atrasentan (0.5, 1.5, and 5.0 mg kg(-1) d(-1) for 5 days) elicited dose-dependent decreases in MAP-AUC (-16.7 +/- 1.3, -20.94 +/- 3.68, and -30.12 +/- 3.57 mm Hg x day, respectively) compared with vehicle. ABT-869 (1, 3, 10, 30 mg kg(-1) d(-1) for 5 days) increased MAP compared with vehicle (MAP-AUC values of -5.52 +/- 3.75, 12.7 +/- 8.4, 37.5 +/- 4.4, and 63.8 +/- 3.3 mm Hg x day, respectively). Pretreatment with atrasentan (5 mg/kg for 5 days) prevented and abolished the hypertensive effects of ABT-869. Thus, ETA receptor blockade effectively alleviated hypertension with RTK inhibition and may serve a dual therapeutic role by preventing hypertension and slowing tumor progression.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antagonistas de los Receptores de la Endotelina A , Hipertensión/prevención & control , Indazoles/farmacología , Compuestos de Fenilurea/farmacología , Pirrolidinas/farmacología , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Inhibidores de la Angiogénesis/efectos adversos , Animales , Área Bajo la Curva , Atrasentán , Presión Sanguínea/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Frecuencia Cardíaca/efectos de los fármacos , Hipertensión/inducido químicamente , Hipertensión/fisiopatología , Indazoles/efectos adversos , Masculino , Compuestos de Fenilurea/efectos adversos , Pirrolidinas/uso terapéutico , Ratas , Ratas Sprague-Dawley , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Telemetría
3.
J Med Chem ; 50(22): 5439-48, 2007 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-17918921

RESUMEN

A new structural series of histamine H3 receptor antagonist was developed. The new compounds are based on a quinoline core, appended with a required basic aminoethyl moiety, and with potency- and property-modulating heterocyclic substituents. The analogs have nanomolar and subnanomolar potency for the rat and human H3R in various in vitro assays, including radioligand competition binding as well as functional tests of H3 receptor-mediated calcium mobilization and GTPgammaS binding. The compounds possessed favorable drug-like properties, such as good PK, CNS penetration, and moderate protein binding across species. Several compounds were found to be efficacious in animal behavioral models of cognition and attention. Further studies on the pharmaceutic properties of this series of quinolines discovered a potential problem with photochemical instability, an issue which contributed to the discontinuation of this series from further development.


Asunto(s)
Pirazoles/síntesis química , Pirimidinas/síntesis química , Quinolinas/síntesis química , Receptores Histamínicos H3/metabolismo , Animales , Atención/efectos de los fármacos , Reacción de Prevención/efectos de los fármacos , Proteínas Sanguíneas/metabolismo , Barrera Hematoencefálica/metabolismo , Calcio/metabolismo , Línea Celular , Cognición/efectos de los fármacos , Perros , Agonismo Inverso de Drogas , Estabilidad de Medicamentos , Haplorrinos , Humanos , Unión Proteica , Pirazoles/farmacocinética , Pirazoles/farmacología , Pirimidinas/farmacocinética , Pirimidinas/farmacología , Quinolinas/farmacocinética , Quinolinas/farmacología , Ensayo de Unión Radioligante , Ratas , Ratas Endogámicas SHR , Reconocimiento en Psicología/efectos de los fármacos , Conducta Social , Estereoisomerismo , Relación Estructura-Actividad , Distribución Tisular
4.
Nephron Physiol ; 106(4): p76-81, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17622742

RESUMEN

BACKGROUND/AIMS: Vitamin D receptor activators (VDRAs) may suppress renin expression and VDR-mediated renin inhibitors may offer a novel mechanism to control the RAS. METHODS: We delineated the effects of paricalcitol and calcitriol on PTH, renin, and iCa(2+) in C57/BL6 mice administered vehicle, paricalcitol, or calcitriol (0.01, 0.03, 0.10, 0.33, 1.0 microg/kg s.c.) 3 days/week for 9 days. RESULTS: Paricalcitol produced PTH suppression from 0.03 to 1.0 microg/kg (values between 9.7 +/- 3.3 and 20.7 +/- 4.7 pg/ml; vehicle = 88.0 +/- 16.9) and elicited dose-dependent reductions in renin/GAPDH expression at 0.33 and 1.0 microg/kg (0.037 +/- 0.002, 0.027 +/- 0.003; vehicle = 0.054 +/- 0.003) but produced no increases iCa(2+) at any dose tested. Calcitrol produced PTH suppression at all doses tested (between 6.4 +/- 1.2 and 29.5 +/- 17.2 pg/ml) and renin suppression at 0.10, 0.33, and 1.0 microg/kg (0.029 +/- 0.002, 0.031 +/- 0.003, and 0.038 +/- 0.02). However, at 0.33 and 1.0 mg/kg, calcitriol produced increases iCa(2+) (1.31 +/- 0.03 and 1.48 +/- 0.02 mmol/l; vehicle = 1.23 +/- 0.02 mmol/l). CONCLUSIONS: Paricalcitol produces significant, dose-dependent suppression of renin expression in the absence of hypercalcemia at doses 10-fold above those necessary for PTH suppression. Calcitriol also produced suppression of renin at doses at least 10-fold above those required for PTH suppression, but increases in iCa(2+) were observed at doses only 3-fold above those necessary to elicit renin suppression.


Asunto(s)
Calcitriol/administración & dosificación , Calcio/metabolismo , Ergocalciferoles/administración & dosificación , Riñón/metabolismo , Hormona Paratiroidea/metabolismo , Renina/metabolismo , Activación Transcripcional/fisiología , Animales , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Riñón/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Renina/genética , Activación Transcripcional/efectos de los fármacos
5.
Int J Parasitol ; 47(12): 753-763, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28899690

RESUMEN

Improvements have been made to the safety and efficacy of bumped kinase inhibitors, and they are advancing toward human and animal use for treatment of cryptosporidiosis. As the understanding of bumped kinase inhibitor pharmacodynamics for cryptosporidiosis therapy has increased, it has become clear that better compounds for efficacy do not necessarily require substantial systemic exposure. We now have a bumped kinase inhibitor with reduced systemic exposure, acceptable safety parameters, and efficacy in both the mouse and newborn calf models of cryptosporidiosis. Potential cardiotoxicity is the limiting safety parameter to monitor for this bumped kinase inhibitor. This compound is a promising pre-clinical lead for cryptosporidiosis therapy in animals and humans.


Asunto(s)
Criptosporidiosis/tratamiento farmacológico , Cryptosporidium parvum/efectos de los fármacos , Inhibidores de Proteínas Quinasas/uso terapéutico , Administración Oral , Animales , Animales Recién Nacidos , Bovinos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Corazón/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Interferón gamma/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Pruebas de Mutagenicidad , Embarazo , Unión Proteica , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/sangre , Inhibidores de Proteínas Quinasas/toxicidad , Seguridad
6.
Artículo en Inglés | MEDLINE | ID: mdl-27432021

RESUMEN

Screening novel, poorly soluble small-molecule candidates for cardiovascular liabilities represents a key challenge in early drug discovery. This report describes a novel vehicle composed of 20% N,N-Dimethylacetamide (DMA)/40% Propylene glycol (PG)/40% Polyethylene Glycol (PEG-400) (DPP) for administration of new chemical entities (NCEs) by slow intravenous (i.v.) infusion in a preclinical anesthetized rat model. The vehicle was designed considering both available excipient safety information and solubilization potential for poorly soluble NCEs. DPP solubilized 11 drugs, 8 of which were insoluble in 5% dextrose in water (D5W), and 5 insoluble in PEG-400 to a target concentration of 30mg/mL. DPP elicits no adverse cardiovascular responses in the anesthetized rat model despite containing 40% PEG-400, a commonly used organic solvent which elicits hypertension and bradycardia that often confounds interpretation of drug effects. Three compounds demonstrating adequate solubility in both DPP and D5W were screened in the anesthetized rat model. When normalized to plasma exposure, atenolol, sotalol and enalaprilat exhibited comparable mean arterial pressure, heart rate, and cardiac contractility responses regardless of formulation. While the antihypertensive effect of nifedipine was evident with both DPP and PEG-400 formulations, pressor effects from PEG-400 confounded interpretation of the magnitude of nifedipine's response. Plasma concentrations of atenolol and enalaprilat were greater in D5W formulation whereas sotalol exposures were greater when using DPP as a vehicle. These results demonstrate the utility of DPP as an intravenous vehicle for formulating poorly soluble compounds in early preclinical screening for cardiovascular safety studies.


Asunto(s)
Portadores de Fármacos/química , Excipientes/química , Hemodinámica/efectos de los fármacos , Modelos Cardiovasculares , Preparaciones Farmacéuticas/administración & dosificación , Bibliotecas de Moléculas Pequeñas/administración & dosificación , Acetamidas/administración & dosificación , Acetamidas/química , Acetamidas/toxicidad , Animales , Portadores de Fármacos/administración & dosificación , Portadores de Fármacos/toxicidad , Descubrimiento de Drogas/métodos , Evaluación Preclínica de Medicamentos , Excipientes/administración & dosificación , Excipientes/toxicidad , Infusiones Intravenosas , Dosificación Letal Mediana , Masculino , Preparaciones Farmacéuticas/sangre , Preparaciones Farmacéuticas/química , Polietilenglicoles/administración & dosificación , Polietilenglicoles/química , Polietilenglicoles/toxicidad , Propilenglicol/administración & dosificación , Propilenglicol/química , Propilenglicol/toxicidad , Ratas Sprague-Dawley , Bibliotecas de Moléculas Pequeñas/efectos adversos , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacocinética , Solubilidad
7.
J Med Chem ; 59(10): 4926-47, 2016 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-27077528

RESUMEN

Transient receptor potential vanilloid 3 (TRPV3) is a Ca(2+)- and Na(+)-permeable channel with a unique expression pattern. TRPV3 is found in both neuronal and non-neuronal tissues, including dorsal root ganglia, spinal cord, and keratinocytes. Recent studies suggest that TRPV3 may play a role in inflammation, pain sensation, and skin disorders. TRPV3 studies have been challenging, in part due to a lack of research tools such as selective antagonists. Herein, we provide the first detailed report on the development of potent and selective TRPV3 antagonists featuring a pyridinyl methanol moiety. Systematic optimization of pharmacological, physicochemical, and ADME properties of original lead 5a resulted in identification of a novel and selective TRPV3 antagonist 74a, which demonstrated a favorable preclinical profile in two different models of neuropathic pain as well as in a reserpine model of central pain.


Asunto(s)
Ciclobutanos/síntesis química , Ciclobutanos/farmacología , Piridinas/síntesis química , Piridinas/farmacología , Canales Catiónicos TRPV/antagonistas & inhibidores , Calcio/metabolismo , Ciclobutanos/química , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Conformación Molecular , Piridinas/química , Relación Estructura-Actividad , Canales Catiónicos TRPV/metabolismo
8.
Biochem Pharmacol ; 83(3): 406-18, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22153861

RESUMEN

Blockade of voltage-gated Ca²âº channels on sensory nerves attenuates neurotransmitter release and membrane hyperexcitability associated with chronic pain states. Identification of small molecule Ca²âº channel blockers that produce significant antinociception in the absence of deleterious hemodynamic effects has been challenging. In this report, two novel structurally related compounds, A-686085 and A-1048400, were identified that potently block N-type (IC50=0.8 µM and 1.4 µM, respectively) and T-type (IC50=4.6 µM and 1.2 µM, respectively) Ca²âº channels in FLIPR based Ca²âº flux assays. A-686085 also potently blocked L-type Ca²âº channels (EC50=0.6 µM), however, A-1048400 was much less active in blocking this channel (EC50=28 µM). Both compounds dose-dependently reversed tactile allodynia in a model of capsaicin-induced secondary hypersensitivity with similar potencies (EC50=300-365 ng/ml). However, A-686085 produced dose-related decreases in mean arterial pressure at antinociceptive plasma concentrations in the rat, while A-1048400 did not significantly alter hemodynamic function at supra-efficacious plasma concentrations. Electrophysiological studies demonstrated that A-1048400 blocks native N- and T-type Ca²âº currents in rat dorsal root ganglion neurons (IC50=3.0 µM and 1.6 µM, respectively) in a voltage-dependent fashion. In other experimental pain models, A-1048400 dose-dependently attenuated nociceptive, neuropathic and inflammatory pain at doses that did not alter psychomotor or hemodynamic function. The identification of A-1048400 provides further evidence that voltage-dependent inhibition of neuronal Ca²âº channels coupled with pharmacological selectivity vs. L-type Ca²âº channels can provide robust antinociception in the absence of deleterious effects on hemodynamic or psychomotor function.


Asunto(s)
Analgésicos/administración & dosificación , Bloqueadores de los Canales de Calcio/administración & dosificación , Hemodinámica/fisiología , Neuronas/fisiología , Dimensión del Dolor , Piperidonas/administración & dosificación , Piperidonas/química , Administración Oral , Animales , Relación Dosis-Respuesta a Droga , Células HEK293 , Hemodinámica/efectos de los fármacos , Humanos , Masculino , Neuronas/efectos de los fármacos , Dimensión del Dolor/efectos de los fármacos , Dimensión del Dolor/métodos , Ratas , Ratas Sprague-Dawley
9.
Pain ; 142(1-2): 27-35, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19135797

RESUMEN

Transient receptor potential vanilloid type 1 (TRPV1) is a ligand-gated ion channel that functions as an integrator of multiple pain stimuli including heat, acid, capsaicin and a variety of putative endogenous lipid ligands. TRPV1 antagonists have been shown to decrease inflammatory pain in animal models and to produce limited hyperthermia at analgesic doses. Here, we report that ABT-102, which is a potent and selective TRPV1 antagonist, is effective in blocking nociception in rodent models of inflammatory, post-operative, osteoarthritic, and bone cancer pain. ABT-102 decreased both spontaneous pain behaviors and those evoked by thermal and mechanical stimuli in these models. Moreover, we have found that repeated administration of ABT-102 for 5-12 days increased its analgesic activity in models of post-operative, osteoarthritic, and bone cancer pain without an associated accumulation of ABT-102 concentration in plasma or brain. Similar effects were also observed with a structurally distinct TRPV1 antagonist, A-993610. Although a single dose of ABT-102 produced a self-limiting increase in core body temperature that remained in the normal range, the hyperthermic effects of ABT-102 effectively tolerated following twice-daily dosing for 2 days. Therefore, the present data demonstrate that, following repeated administration, the analgesic activity of TRPV1 receptor antagonists is enhanced, while the associated hyperthermic effects are attenuated. The analgesic efficacy of ABT-102 supports its advancement into clinical studies.


Asunto(s)
Analgésicos/administración & dosificación , Fiebre/tratamiento farmacológico , Indazoles/administración & dosificación , Umbral del Dolor/efectos de los fármacos , Dolor/tratamiento farmacológico , Canales Catiónicos TRPV/metabolismo , Urea/análogos & derivados , Animales , Temperatura Corporal/efectos de los fármacos , Neoplasias Óseas/complicaciones , Calcio/metabolismo , Modelos Animales de Enfermedad , Interacciones Farmacológicas , Fiebre/inducido químicamente , Inflamación/complicaciones , Masculino , Ratones , Ratones Endogámicos C3H , Actividad Motora/efectos de los fármacos , Osteoartritis/complicaciones , Dolor/etiología , Dimensión del Dolor , Ratas , Ratas Sprague-Dawley , Canales Catiónicos TRPV/antagonistas & inhibidores , Urea/administración & dosificación
10.
Am J Physiol Heart Circ Physiol ; 294(1): H238-48, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17982006

RESUMEN

Levosimendan enhances cardiac contractility via Ca(2+) sensitization and induces vasodilation through the activation of ATP-dependent K(+) and large-conductance Ca(2+)-dependent K(+) channels. However, the hemodynamic effects of levosimendan, as well as its metabolites, OR-1896 and OR-1855, relative to plasma concentrations achieved, are not well defined. Thus levosimendan, OR-1896, OR-1855, or vehicle was infused at 0.01, 0.03, 0.1, and 0.3 mumol.kg(-1).30 min(-1), targeting therapeutic to supratherapeutic concentrations of total levosimendan (62.6 ng/ml). Results were compared with those of the beta(1)-agonist dobutamine and the phosphodiesterase 3 inhibitor milrinone. Peak concentrations of levosimendan, OR-1896, and OR-1855 were 455 +/- 21, 126 +/- 6, and 136 +/- 6 ng/ml, respectively. Levosimendan and OR-1896 produced dose-dependent reductions in mean arterial pressure (-31 +/- 2 and -42 +/- 3 mmHg, respectively) and systemic resistance without affecting pulse pressure, effects paralleled by increases in heart rate; OR-1855 produced no effect at any dose tested. Dobutamine, but not milrinone, increased mean arterial pressure and pulse pressure (17 +/- 2 and 23 +/- 2 mmHg, respectively). Regarding potency to elicit reductions in time to peak pressure and time to systolic pressure recovery: OR-1896 > levosimendan > milrinone > dobutamine. Levosimendan and OR-1896 elicited dose-dependent increases in change in pressure over time (118 +/- 10 and 133 +/- 13%, respectively), concomitant with reductions in left ventricular end-diastolic pressure and ejection time. However, neither levosimendan nor OR-1896 produced increases in myocardial oxygen consumption at inotropic and vasodilatory concentrations, whereas dobutamine increased myocardial oxygen consumption (79% above baseline). Effects of the levosimendan and OR-1896 were limited to the systemic circulation; neither compound produced changes in pulmonary pressure, whereas dobutamine produced profound increases (74 +/- 13%). Thus levosimendan and OR-1896 are hemodynamically active in the anesthetized dog at concentrations observed clinically and elicit cardiovascular effects consistent with activation of both K(+) channels and Ca(2+) sensitization, whereas OR-1855 is inactive on endpoints measured in this study.


Asunto(s)
Acetamidas/farmacología , Cardiotónicos/farmacología , Sistema Cardiovascular/efectos de los fármacos , Dobutamina/farmacología , Hidrazonas/farmacología , Milrinona/farmacología , Consumo de Oxígeno/efectos de los fármacos , Piridazinas/farmacología , Resistencia Vascular/efectos de los fármacos , Vasodilatadores/farmacología , Animales , Presión Sanguínea/efectos de los fármacos , Calcio/metabolismo , Gasto Cardíaco/efectos de los fármacos , Cardiotónicos/sangre , Sistema Cardiovascular/metabolismo , Perros , Relación Dosis-Respuesta a Droga , Frecuencia Cardíaca/efectos de los fármacos , Hidrazonas/sangre , Masculino , Contracción Miocárdica/efectos de los fármacos , Potasio/metabolismo , Canales de Potasio/efectos de los fármacos , Canales de Potasio/metabolismo , Circulación Pulmonar/efectos de los fármacos , Piridazinas/sangre , Simendán , Factores de Tiempo , Vasodilatadores/sangre , Función Ventricular Izquierda/efectos de los fármacos
11.
J Pharmacol Exp Ther ; 323(1): 217-26, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17636005

RESUMEN

Calcium-sensing receptor (CaR) activation decreases serum parathyroid hormone (PTH) and Ca2+ and, despite long-term reductions in mean arterial blood pressure (MAP), may produce acute hypertension in rats, an effect we hypothesized was mediated by constriction of multiple vascular beds. Rats were subjected to 5/6 nephrectomy (NX) or no surgery (Normal); at 7 to 8 weeks, uremia animals were anesthetized and instrumented to record MAP and regional blood flow (carotid, mesenteric, and hindlimb). Cinacalcet [N-(1-naphthalen-1-ylethyl)-3-[3-(trifluoromethyl)phenyl]-propan-1-amine; 1, 3, and 10 mg/kg; 30 min/dose] was infused over 90 min. In NX rats, cinacalcet dose-dependently decreased ionized calcium (iCa2+), elicited a 90% reduction in PTH, and produced dose-dependent self-limiting increases in MAP (from 119 +/- 6 to 129 +/- 5, 142 +/- 4, and 145 +/- 3 mm Hg at the end of each infusion). At 1 mg/kg, carotid vascular resistance (CVR) and mesenteric vascular resistance (MVR) increased to 16 +/- 6 and 18 +/- 6% above baseline, respectively. Hindlimb vascular resistance (HVR) also trended upward (13 +/- 8%). At 3 mg/kg, increases in CVR (38 +/- 10%), MVR (40 +/- 8%), and HVR (39 +/- 14%) were exacerbated; at 10 mg/kg, values remained at or near these levels. The effects of cinacalcet in Normal rats were similar to NX and were attenuated by ganglionic blockade with hexamethonium at low doses but remained significantly elevated at higher doses. Thus, CaR activation acutely increases MAP in uremic and nonuremic rats, responses that occur in parallel to vasoconstriction in multiple vascular beds through both a central and peripheral mechanism of action. Moreover, subsequent mechanistic studies suggest that increases in MAP produced by cinacalcet may be mediated by reduced tonic NO synthase-dependent NO production subsequent to reductions in blood iCa2+.


Asunto(s)
Vías Aferentes/fisiología , Presión Sanguínea/efectos de los fármacos , Vías Eferentes/fisiología , Tono Muscular/efectos de los fármacos , Músculo Liso Vascular , Naftalenos/farmacología , Receptores Sensibles al Calcio/metabolismo , Uremia , Animales , Calcio/sangre , Cinacalcet , Frecuencia Cardíaca/efectos de los fármacos , Masculino , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/inervación , Hormona Paratiroidea/sangre , Ratas , Ratas Sprague-Dawley , Flujo Sanguíneo Regional/efectos de los fármacos , Uremia/tratamiento farmacológico , Uremia/metabolismo , Uremia/fisiopatología , Resistencia Vascular/efectos de los fármacos
12.
Am J Physiol Heart Circ Physiol ; 290(6): H2554-9, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16399858

RESUMEN

The net contribution of endothelin type A (ET(A)) and type B (ET(B)) receptors in blood pressure regulation in humans and experimental animals, including the conscious mouse, remains undefined. Thus we assessed the role of ET(A) and ET(B) receptors in the control of basal blood pressure and also the role of ET(A) receptors in maintaining the hypertensive effects of systemic ET(B) blockade in telemetry-instrumented mice. Mean arterial pressure (MAP) and heart rate were recorded continuously from the carotid artery and daily (24 h) values determined. At baseline, MAP ranged from 99 +/- 1 to 101 +/- 1 mmHg and heart rate ranged between 547 +/- 15 and 567 +/- 19 beats/min (n = 6). Daily oral administration of the ET(B) selective antagonist A-192621 [10 mg/kg twice daily] increased MAP to 108 +/- 1 and 112 +/- 2 mmHg on days 1 and 5, respectively. Subsequent coadministration of the ET(A) selective antagonist atrasentan (5 mg/kg twice daily) in conjunction with A-192621 (10 mg/kg twice daily) decreased MAP to baseline values on day 6 (99 +/- 2 mmHg) and to below baseline on day 8 (89 +/- 3 mmHg). In a separate group of mice (n = 6) in which the treatment was reversed, systemic blockade of ET(B) receptors produced no hypertension in animals pretreated with atrasentan, underscoring the importance of ET(A) receptors to maintain the hypertension produced by ET(B) blockade. In a third group of mice (n = 10), ET(A) blockade alone (atrasentan; 5 mg/kg twice daily) produced an immediate and sustained decrease in MAP to values below baseline (baseline values = 101 +/- 2 to 103 +/- 2 mmHg; atrasentan decreased pressure to 95 +/- 2 mmHg). Thus these data suggest that ET(A) and ET(B) receptors play a physiologically relevant role in the regulation of basal blood pressure in normal, conscious mice. Furthermore, systemic ET(B) receptor blockade produces sustained hypertension in conscious telemetry-instrumented mice that is absent in mice pretreated with an ET(A) antagonist, suggesting that ET(A) receptors maintain the hypertension produced by ET(B) blockade.


Asunto(s)
Presión Sanguínea/efectos de los fármacos , Antagonistas de los Receptores de la Endotelina B , Hipertensión/fisiopatología , Receptor de Endotelina A/efectos de los fármacos , Animales , Atrasentán , Relación Dosis-Respuesta a Droga , Frecuencia Cardíaca/efectos de los fármacos , Hipertensión/inducido químicamente , Ratones , Ratones Endogámicos C57BL , Pirrolidinas/farmacología , Telemetría
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA